You are on page 1of 228

Volume 143

Number 5

November 24, 2010

www.cell.com

Muscle Aging Cues Systemic Aging


Modifications
Reviews: Posttranslational Modifications
SAN TA CR UZ B I OT ECH N O LO GY, I N C . SANTA CRUZ
®

Leading (#1 worldwide) supplier BIOTECHNOLOGY

of specialty biochemicals for


The Power to Question

proteomics research.

Products include over 71,300 unique biochemicals


sorted alphabetically, by CAS number, by formula
and on the basis of functional activity.

See our web site for product details


and ordering information.

www.scbt.com

HEADQUARTERS
TOLL FREE: 800.457.3801 TEL: 831.457.3800
FAX: 831.457.3801 E-MAIL: scbt@scbt.com

EUROPEAN SUPPORT
TOLL FREE: +00800.4573.8000 PHONE: +49.6221.4503.0
FAX: +49.6221.4503.45 E-MAIL: europe@scbt.com

ASIAN SUPPORT
JAPAN TOLL FREE: (010) 800.40402026 S. KOREA TOLL FREE: 00798.1.1.002.0297
N. CHINA TOLL FREE: 10.800.711.0752 S. CHINA TOLL FREE: 10.800.110.0694
FAX: 831.457.3801 E-MAIL: asia@scbt.com

ChemCruz ™
.
E W Intelligent design

N +
inCuSaFe™ copper enriched
stainless steel interior

+ Single-beam, dual capture


infrared CO2 sensor

+ SafeCell UV
protection in situ

+ Hydrogen peroxide
vapor sterilization in situ

1
Good laboratory technique

Sterisonic™ GxP
Performance and Productivity
Delivers Best Efficiency Value:

2 Hours 14 Hours

SANYO Brand X
Sterisonic™

H2O2 sterilization vs. high heat sterilization


= Uptime (Hours) = Downtime (Hours)

The rapid in situ H2O2 sequence returns the


fully sterilized Sterisonic™ GxP to normal use quicker

Spot on results.
than any competitive incubator in the world.

The industry’s first in situ H2O2 sterilization with the fastest turnaround.
For maximum productivity in clinical, general purpose or the most highly compliant GMP applications,
the new SANYO Sterisonic™ GxP CO2 incubator offers an impressive return on investment. With multiple
contamination control safeguards, exclusive on-board H2O2 sterilization, FDA-21CFR data capture and
graphical LCD display, the Sterisonic™ GxP rewards good laboratory technique with performance
you can trust. Learn more, visit www.sterisonic.com or call 800-858-8442.

pictured above: Sterisonic™ GxP MCO-19AICUVH with rapid H2O2 vapor sterilization system.

FREEER
OFF
[ D E TA IL
S
!
O N L IN E ]
FREE!
FREE!
H2O2 sterilization system acessory, plus BD labware consumables kit.
Limited time upgrade offer with purchase of the Sterisonic™ GxP MCO 19AICUVH. ($900 Value)
BD stem cell starter kit with Sterisonic™ GxP quote.
No purchase necessary! Act now. Supplies are limited. ($150 Value)
www.sterisonic.com
©2009 Sanyo Biomedical OWS 1015 05/09
60
years of leadership in
human genetics research,
education and service.
1948–2008
www.ashg.org
Do A 180

Turn to a world of true


CO2 incubator sterilization.
Binder CO2 incubators offer:
A 180°C hot-air sterilization cycle that
4
eliminates contamination.
A hot-air jacket that delivers consistent,
4

uniform temperatures.
Drift-free real-time CO2 measurement that
4

ensures stable pH values.
Condensation-free humidification that
4

maintains dry interior walls.
A one-piece weld-free interior with rounded
4

corners and integrated shelf supports.

Get directions at www.true-sterilization.com


or 866-885-9794.

BINDER Inc. | Toll Free 866 885 9794


sales@binder-world.us | www.binder-world.us
xCELLigence System: E-Plate VIEW 96

Real-Time Cell Analysis in a Whole New Light


Combine real-time impedance-based monitoring of cell behavior with visual inspection in a single
96-well plate. The new E-Plate VIEW 96 for xCELLigence RTCA SP and MP Instruments is an upgraded
device that features both measuring electrodes and a clear inspection window on the well bottoms,
enabling you to get the most out of your cell analysis experiments.
 Monitor cell activity continuously in real-time
 Observe measured changes using microscopes and the Cellavista System, or other automated
cell-imaging systems
 Precisely identify the best time points for further experiments (see Figure 1)
1.7
1.5
1.3
Normalized Cell Index

1.1
0.9
0.7
0.5
0.3 Control Paclitaxel
0.1
-0.1
0 11 22 33 44 55 66 77
Time [h]
Figure 1: Combine impedance-based, real-time monitoring with microscopic optical detection. Cell proliferation and cell
death were continuously monitored using the xCELLigence System. The optimal time point for visual inspection was determined and
images were taken 24 hours after compound treatment using a Z16 Apo Microscope with light base (Leica Micro systems).

For more information please visit: www.xcelligence.roche.com

For life science research only.


Not for use in diagnostic procedures.
XCELLIGENCE is a trademark of Roche. Roche Diagnostics GmbH
E-PLATE and ACEA BIOSCIENCES are registered trademarks
of ACEA Biosciences, Inc. in the US.
Roche Applied Science
© 2010 Roche Diagnostics GmbH. All rights reserved. 82372 Penzberg, Germany
Editor Editorial Board Douglas Green Dana Pe’er
Emilie Marcus Abul Abbas Leonard Guarente Kathrin Plath
C. David Allis Taekjip Ha Carol Prives
Senior Deputy Editor
Geneviève Almouzni Daniel Haber Klaus Rajewsky
Elena Porro
Uri Alon Ulrike Heberlein Venki Ramakrishnan
Deputy Editors Angelika Amon Nobutaka Hirokawa Rama Ranganathan
Robert Kruger Johan Auwerx Mark Hochstrasser Anne Ridley
Connie M. Lee Richard Axel Arthur Horwich James Roberts
Cori Bargmann Tony Hunter Alexander Rudensky
Scientific Editors Konrad Basler James Hurley Helen Saibil
Karen Carniol
Bonnie Bassler Richard Hynes Joshua Sanes
Michaeleen Doucleff David Baulcombe Thomas Jessell Randy Schekman
Fabiola Rivas Jeffrey Benovic Narry Kim Ueli Schibler
Niki Scaplehorn Carolyn Bertozzi Mary-Claire King Joseph Schlessinger
Lara Szewczak
Wendy Bickmore David Kingsley Hans Schöler
Senior Managing Editor Elizabeth Blackburn Frank Kirchhoff Trina Schroer
Meredith Adinolfi Joan Brugge Richard Kolodner Geraldine Seydoux
Lewis Cantley John Kuriyan Kevan Shokat
Deputy Managing Editor Joanne Chory Robert Lamb Pamela Sklar
Andy Smith David Clapham Mark Lemmon Nahum Sonenberg
Lead Illustrator Andrew Clark Beth Levine James Spudich
Andrew A. Tang Hans Clevers Wendell Lim Paul Sternberg
Stephen Cohen Jennifer Lippincott-Schwartz Bruce Stillman
Illustrators Pascale Cossart Dan Littman Azim Surani
Yvonne Blanco Jeff Dangl Richard Losick Keiji Tanaka
Kate Mahan Ted Dawson Scott Lowe Craig Thompson
Pier Paolo di Fiore Tom Maniatis Robert Tjian
Production Staff
Marileen Dogterom Matthias Mann Jürg Tschopp
Reyna Clancy
Julian Downward Kelsey Martin Ulrich von Andrian
Editorial Assistant Bruce Edgar Joan Massagué Gerhard Wagner
Mary Beth O’Leary Steve Elledge Iain Mattaj Detlef Weigel
Anne Ephrussi Satyajit Mayor Alan Weiner
Ronald Evans Ruslan Medzhitov Jonathan Weissman
Witold Filipowicz Craig Mello Matthew Welch
Marco Foiani Tom Misteli Tian Xu
Elaine Fuchs Tim Mitchison Shinya Yamanaka
Yukiko Goda Alex Mogilner Marino Zerial
Stephen Goff Paul Nurse Xiaowei Zhuang
Joe Goldstein Roy Parker Huda Zoghbi

Cell Office
Cell, Cell Press, 600 Technology Square, 5th Floor, Cambridge, Massachusetts 02139
Phone: (+1) 617 661 7057, Fax: (+1) 617 661 7061, E-mail: celleditor@cell.com
Online Publication: http://www.cell.com

Cell (ISSN 0092-8674) is published biweekly by Cell Press, 600 Technology Square, 5th Floor, Cambridge, Massachusetts 02139. The institutional subscription rate for
2010 is $1,360 (US and Canada) or $1,532 (elsewhere). The individual subscription rate is $202 (US and Canada) or $305 (elsewhere). The individual copy price is $50.
Periodicals postage paid at Boston, Massachusetts and additional mailing offices. Postmaster: send address changes to Elsevier Customer Service Americas,
Cell Press Journals, 11830 Westline Industrial Drive, St. Louis, MO 63146, USA.
The paper used in this publication meets the requirments of ANSI/NISO Z39.48-1992 (Permanence of Paper). Printed by Dartmouth Printing Company, Hanover, NH.
XenoWorks Microinjection Workstation
TM

Smooth, responsive, precise. Always.

Micromanipulator
Highly ergonomic inverted joystick
One-touch coarse and fine control
Superior mechanical stability

Digital Microinjector
Dual channel pneumatic microinjector
Holds, transfers and injects - all from a
single remote keypad

Analog Microinjector
Use with oil, water, or air
Interchangeable syringes

ONE DIGITAL DRIVE, NOVATO, CA. 9494 9 PHONE: 415.883.0128 | FAX: 415.883.057 2
EMAIL: INFO@SUTTER.COM | WWW.SUTTER.COM

Naturally beautiful.
FINE SURGICAL

INSTRUMENTS

FOR RESEARCH™

SHIPPING GLOBALLY
SINCE 1974

Request a catalog
at finescience.com
or call 1-800-521-2109.
Cell Press Display Advertising
Northeast/Mid-Atlantic: Victoria Macomber, ph: 508 928
President & CEO 1255; fax: 508 928 1256; e-mail: v.macomber@elsevier.com
Lynne Herndon Midwest/Southeast/Eastern Canada: Inez Herrero-Redman,
ph: 585 678 4395; fax: 585 678 4722; e-mail: i.herrero@elsevier.
Editor in Chief, Vice President of Content Development
com
Emilie Marcus
Northwest/Southwest/Western Canada: Lori Young, ph: 646
Vice President of Marketing and Publishing 370 6312; fax: 212 462 1915; e-mail: l.young@elsevier.com
Els Bosma California: Elizabeth Loennborn, ph: 714 655 1877; fax: 214 452
Vice President of Web Development and Operations 9627; e-mail: e.loennborn@elsevier.com
Keith Wollman UK/Europe: James Kenney, ph: +44 20 7424 4216; fax: +44 18
6585 3136; e-mail: j.kenney@elsevier.com
Director of Marketing
Jonathan Atkinson Asia: Wendy Xie, ph: +86 10 8520 8827; e-mail: w.xie@
elsevier.com
Production Manager
Meredith Adinolfi Classified Advertising
United States and Canada:
Press Officer Gordon Sheffield, Key Account Manager, ph: 617 386 2189; fax:
Cathleen Genova 617 397 2805; e-mail: g.sheffield@elsevier.com
UK, Europe, and Asia:
Sabrina Dodge, Key Account Manager, ph: +44 20 7424 4997;
fax: +44 18 6585 3136; e-mail: s.dodge@elsevier.com

ª2010 Elsevier Inc. All rights reserved. advances in the medical sciences, in particular, independent verification
This journal and the individual contributions contained in it are protected of diagnoses and drug dosages should be made. Although all advertis-
under copyright by Elsevier Inc., and the following terms and conditions ing material is expected to conform to ethical (medical) standards, inclu-
apply to their use: sion in this publication does not constitute a guarantee or endorsement
Photocopying: of the quality or value of such product or of the claims made of it by its
Single photocopies of single articles may be made for personal use as al- manufacturer.
lowed by national copyright laws. Permission of the Publisher and payment Reprints:
of a fee are required for all other photocopying, including multiple or system- Article reprints are available through Cell’s reprint service; for informa-
atic copying, copying for advertising or promotional purposes, resale, and tion, contact Nicholas Pavlow (e-mail: n.pavlow@elsevier.com; ph: (+1)
all forms of document delivery. Special rates are available for educational 212 633 3960).
institutions that wish to make photocopies for nonprofit educational class- Subscription Orders and Inquiries:
room use. For information on how to seek permission, visit www.elsevier. Mail, fax, or e-mail address changes to Elsevier Customer Service Amer-
com/permissions or call (+44) 1865 843830 (UK) / (+1) 215 239 3804 (US). icas, allowing 4–6 weeks for processing. Lost or damaged issues will be
Permissions: replaced, subject to availability, if Cell Press is notified within the claim
For information on how to seek permission, visit www.elsevier.com/ period (US and airmail delivery: 3 months from issue date; surface deliv-
permissions or call (+44) 1865 843830 (UK) / (+1) 215 239 3804 (US). ery: 4 months from issue date). Periodical delivery in the US can take up
Derivative Works: to 3 weeks. Airmail delivery can take 2–4 weeks.
Subscribers may reproduce tables of contents or prepare lists of articles The price of a single copy of Cell is $50 (excluding special issues).
including summaries for internal circulation within their institutions. All orders must be prepaid and in writing. Please include the volume
Permission of the Publisher is required for resale or distribution outside and issue number, payment (check or credit card, MasterCard, Visa,
the institution. Permission of the Publisher is required for all other deriv- or American Express only), and a delivery address. Allow 4–6 weeks
ative works, including compilations and translations (please consult for delivery.
www.elsevier.com/permissions). Mailing address: Elsevier Customer Service Americas, Cell Press
Journals, 11830 Westline Industrial Drive, St. Louis, MO 63146,
Electronic Storage or Usage:
USA. Toll-free phone within USA/Canada: 866 314 2355; phone for
Permission of the Publisher is required to store or use electronically any
outside US/Canada: (+1) 314 453 7038; fax: (+1) 314 523 5170; e-mail:
material contained in this journal, including any article or part of an article
subs@cell.com; internet: www.cellpress.com or <www.cell.com>.
(please consult www.elsevier.com/permissions). Except as outlined
above, no part of this publication may be reproduced, stored in a retrieval Funding Body Agreements and Policies:
system, or transmitted in any form or by any means, electronic, mechan- Elsevier has established agreements and developed policies to allow au-
ical, photocopying, recording, or otherwise, without prior written permis- thors whose articles appear in journals published by Elsevier to comply
sion of the Publisher. with potential manuscript archiving requirements as specified as condi-
Notice: tions of their grant awards. To learn more about existing agreements and
No responsibility is assumed by the Publisher for any injury and/or dam- policies, visit http://www.cell.com/cellpress/FundingBodyAgreements.
age to persons or property as a matter of products liability, negligence, Guide for Authors:
or otherwise, or from any use or operation of any methods, products, For a full and complete guide for authors, please go to www.cell.com/
instructions, or ideas contained in the material herein. Because of rapid authors.
UNDERSTANDING CHANGE

New tools to advance epigenetics research


For over 35 years, New England Biolabs has been committed to understanding the mechanisms of restriction and
methylation of DNA. This expertise in enzymology has recently led to the development of a suite of validated
products for epigenetics research. These unique solutions to study DNA methylation are designed to address some
of the challenges of the current methods. EpiMark™ validated reagents simplify epigenetics research and expand the
potential for biomarker discovery.

Simplify DNA methylation analysis with MspJI EpiMark™ validated products include:
Plant • Newly discovered methylation-dependent restriction enzymes
Hela (Maize) Yeast
– + – + – + MspJI • A novel kit for 5-hmC and 5-mC analysis and quantitation
• Methyltransferases
• Histones
• Genomic DNAs

To learn how these products can help you to better


32 bp understand epigenetic changes, visit neb.com/epigenetics.

MspJI recognizes methylated and hydroxymethylated DNA and cleaves


out 32 bp fragments for downstream sequencing analysis. Overnight
digestion of 1 µg of genomic DNA from various sources with or without
MspJI is shown. Note: Yeast DNA does not contain methylated DNA,
therefore no 32-mer is detected.

DNA AMPLIFICATION PROTEIN EXPRESSION GENE EXPRESSION


CLONING & MAPPING RNA ANALYSIS
& PCR & ANALYSIS & CELLULAR ANALYSIS
www.neb.com
Leading Edge
Cell Volume 143 Number 5, November 24, 2010

IN THIS ISSUE

SELECT
657 Cell Cycle

PREVIEWS

665 ER Sheets Get Roughed Up C. Barlowe


667 SIRT3 in Calorie Restriction: C. Sebastian and R. Mostoslavsky
Can You Hear Me Now?
669 ATP Consumption Promotes W.J. Israelsen and M.G. Vander Heiden
Cancer Metabolism

ESSAYS
672 Glycomics Hits the Big Time G.W. Hart and R.J. Copeland

677 What Determines the Specificity F. Ikeda, N. Crosetto, and I. Dikic


and Outcomes of Ubiquitin Signaling?

MINIREVIEW

682 Ubiquitin: Same Molecule, M.J. Clague and S. Urbe


Different Degradation Pathways

PERSPECTIVE

686 Will the Ubiquitin System Furnish as P. Cohen and M. Tcherpakov


Many Drug Targets as Protein Kinases?

REVIEWS

694 Pathogen-Mediated Posttranslational D. Ribet and P. Cossart


Modifications: A Re-emerging Field

703 Modifications of Small RNAs Y.-K. Kim, I. Heo, and V.N. Kim
and Their Associated Proteins

SNAPSHOT

848 The SUMO System S. Creton and S. Jentsch


Antibodies and Related Reagents for Signal Transduction Research

The highest quality antibodies for the study of

© 2010 Cell Signaling Technology, Inc.


DNA Damage from Cell Signaling Technology
untreated +UV
Unparalleled product quality, validation,
phospho-histone H2A.X

clonal antibodies are produced under license (granting certain rights including those under U.S. Patents No. 5,675,063 and 7,429,487).
Cell Signaling Technology® is a trademark of Cell Signaling Technology, Inc. DRAQ5® is a registered trademark of Biostatus Limited. Selected rabbit mono-
and technical support
HeLa

:: Innovative products from Cell Signaling Technology


offer unsurpassed sensitivity, specificity,
reproducibility, and performance.
:: Extensive in-house validation means optimization
is not left up to you.
phospho-p53
HT-29

:: Technical support provided by the same scientists


who produce and validate the products translates
into a thorough, fast, and accurate response.
Confocal IF analysis of HeLa (upper) and HT-29 cells (lower), untreated (left) or UV- TOP IMAGE: To the right, the nuclear pore complex is located in the nuclear double bilayer. To the
treated (right), using Phospho-Histone H2A.X (Ser139) (20E3) Rabbit mAb left, nuclear proteins are interspersed between DNA and nucleosomes in various levels of compact-
#9718 (green, upper) or Phospho-p53 (Ser15) (16G8) Mouse mAb #9286 ness. Red = histone H2A.X; blue = histones H2B, H3, H4; green = p53 (with nearby ATM below);
(green, lower). Actin filaments were labeled with DY-554 phalloidin (red). purple = Rb (with a nearby E2F dimer on the right); orange = damage/repair MRN complex, loaded
onto a DNA double strand break. Please visit www.cellsignal.com for the complete story.

for quality products you can trust...


www.cellsignal.com
Orders (toll-free) 1-877-616-2355 | Technical support (toll-free) 1-877-678-8324 support@cellsignal.com | Inquiries info@cellsignal.com | Environmental Commitment eco.cellsignal.com
Articles
Cell Volume 143 Number 5, November 24, 2010

711 The ER UDPase ENTPD5 Promotes Protein M. Fang, Z. Shen, S. Huang, L. Zhao, S. Chen,
N-Glycosylation, the Warburg Effect, T.W. Mak, and X. Wang
and Proliferation in the PTEN Pathway

725 Stepwise Histone Replacement by SWR1 E. Luk, A. Ranjan, P.C. FitzGerald, G. Mizuguchi,
Requires Dual Activation with Histone Y. Huang, D. Wei, and C. Wu
H2A.Z and Canonical Nucleosome

737 Sororin Mediates Sister Chromatid T. Nishiyama, R. Ladurner, J. Schmitz, E. Kreidl,


Cohesion by Antagonizing Wapl A. Schleiffer, V. Bhaskara, M. Bando, K. Shirahige,
A.A. Hyman, K. Mechtler, and J.-M. Peters

750 Nonenzymatic Rapid Control A. Raveh, A. Cooper, L. Guy-David, and E. Reuveny


of GIRK Channel Function
by a G Protein-Coupled Receptor Kinase

761 Sequence-Dependent Sorting M.A. Puthenveedu, B. Lauffer, P. Temkin, R. Vistein,


of Recycling Proteins by Actin-Stabilized P. Carlton, K. Thorn, J. Taunton, O.D. Weiner,
Endosomal Microdomains R.G. Parton, and M. von Zastrow

774 Mechanisms Determining the Morphology Y. Shibata, T. Shemesh, W.A. Prinz, A.F. Palazzo,
of the Peripheral ER M.M. Kozlov, and T.A. Rapoport

789 Abortive HIV Infection Mediates G. Doitsh, M. Cavrois, K.G. Lassen, O. Zepeda,
CD4 T Cell Depletion and Inflammation Z. Yang, M.L. Santiago, A.M. Hebbeler,
in Human Lymphoid Tissue and W.C. Greene

802 Sirt3 Mediates Reduction of Oxidative S. Someya, W. Yu, W.C. Hallows, J. Xu,
Damage and Prevention of Age-Related J.M. Vann, C. Leeuwenburgh, M. Tanokura,
Hearing Loss under Caloric Restriction J.M. Denu, and T.A. Prolla

813 FOXO/4E-BP Signaling in Drosophila F. Demontis and N. Perrimon


Muscles Regulates Organism-wide
Proteostasis during Aging

(continued)
BE THE FIRSTto read the latest issue of any Cell Press journal.

Register for Cell Press Email Alerts and get the complete table of contents as soon as
the issue publishes online — FREE!

Cell Press Email Alerts deliver the news, research, and commentaries featured in each
journal’s latest issue, including the full title of every article, direct links to the articles,
and the complete author list. Plus, to save you time, each research article has a brief
summary highlighting its significant findings.

You don’t have to be a subscriber to sign up for Cell Press Email Alerts. While subscribers have
instant access to the full text of all articles listed in the Email Alerts, non-subscribers can read
the abstracts of all articles as well as the full text of the issue’s Featured Article.

www.cellpress.com
826 Reelin and Stk25 Have Opposing T. Matsuki, R.T. Matthews, J.A. Cooper,
Roles in Neuronal Polarization M.P. van der Brug, M.R. Cookson, J.A. Hardy,
and Dendritic Golgi Deployment E.C. Olson, and B.W. Howell

RESOURCE
837 A Human Genome Structural Variation J.M. Kidd, T. Graves, T.L. Newman, R. Fulton,
Sequencing Resource Reveals Insights H.S. Hayden, M. Malig, J. Kallicki, R. Kaul,
into Mutational Mechanisms R.K. Wilson, and E.E. Eichler

POSITIONS AVAILABLE

CONFERENCES AND COURSES

On the cover: The progressive decrease of muscle strength in the elderly is one of the first
signs of aging in many organisms. Here, Demontis and Perrimon (pp. 813–825) demonstrate
that FOXO/4E-BP activity in Drosophila muscles is essential for maintaining protein homeo-
stasis and muscle function and is beneficial for systemic aging by extending life span. The
image is a view of old Drosophila flight muscles with highlighted nuclei (white), myofibrils
(blue), protein aggregates (red), and mitochondria (green).
375 nm 405 nm
100

Laser-based 90
80

Fluorescence 70

Transmission (%)
60
Typical Spectra:

Imaging:
50 Exciter
Dichroic
40 Emitter
Laser Bands
30

What you need


20
10
0

to know
350 375 400 425 450 475 500 525 550
Wavelength (nm)
Long-pass filter set for laser microscopy - LF405/LP-A

Lasers are replacing conventional broadband light Dichroic beamsplitters for laser applications must be
sources for fluorescence imaging applications due anti-reflection (AR) coated to maximize transmission and
to desirable laser properties like high brightness, eliminate coherent interference artifacts. They should
stability, long lifetime, and narrow spectral bandwidth. also have high optical damage ratings like the exciters
These features enable higher sensitivity, better image and low autofluorescence glass like the emitters. And it
fidelity, and superior axial resolution in a variety of is critical for laser dichroics to exhibit sufficient flatness
imaging applications using laser-scanning and to eliminate axial focal shift and transverse aberrations
spinning-disk confocal microscopes and total-internal- associated with reflected laser light.
reflection fluorescence (TIRF) microscopes. The
narrow beam divergence, high spatial and temporal Download our free white paper on this subject at:
coherence, and well-defined polarization properties www.semrock.com
of lasers have enabled new fluorescence imaging
techniques, such as super-resolution. Advances in thin-film filter technology pioneered by
Semrock and embodied in all BrightLine® fluorescence
The use of lasers as fluorescence light sources filters permit the highest-performance fluorescence
imposes new constraints on imaging systems and imaging, while resolving the longevity and handling
their components. For example, all optical filter issues that plague older, soft-coating technology.
wavelengths should be precisely keyed to the
important laser lines and the spectra should offer
steep transitions from the laser wavelength to
fluorescence transmission. And exceptionally We invite you to experience the difference
high transmission is critical to maximize system at no risk. All Semrock filters have a 30 day,
throughput, thus reducing acquisition time. no questions asked return policy. We’ll even
help you make the evaluation. Give us a call
Excitation filters act as “clean-up” filters, minimizing at 866-736-7625 for details.
noise background resulting from the light away from
the laser line, including spontaneous emission
observed in solid-state lasers and the plasma lines of
gas lasers. They should be hard-coated to withstand
the high intensity of the laser beam. Emission filters
should have deep blocking (optical density > 6) at all
possible laser wavelengths to eliminate the intense
stray laser light at the detector and very high transmission
(> 97%) especially for low-light-level applications like
single-molecule imaging. Emitters should also have
low autofluorescence glass and excellent wavefront
performance for minimal beam deviation and aberrations. A Unit of Corporation
Visit www.semrock.com for a complete list
of exclusively hard-coated filters.
Leading Edge

In This Issue

An EnERgy Boost for Cancer


PAGE 711
Rapidly growing cancer cells increase their rate of aerobic glycolysis in a meta-
bolic shift known as the Warburg effect. Their proliferation also demands high
protein folding capacity in the endoplasmic reticulum (ER). Fang et al. identify
an ER-localized enzyme, ENTPD5, that is responsible for both of these features
of tumor cells. Inhibition of ENTPD5, which is commonly upregulated in human
cancers, blocked tumor growth in mice. Thus, ENTPD5 inhibition could poten-
tially become an anticancer therapy.

A Nudge and a Kick for Histone Replacement


PAGE 725
Most promoters in eukaryotes are marked with nucleosomes carrying a special
histone H2A.Z, which is important for gene regulation. SWR1 incorporates
H2A.Z into nucleosomes in a histone replacement reaction. Luk et al. now report a mechanism that ensures that only nucle-
osomes containing the canonical histone H2A are targeted for replacement. SWR1’s ATPase activity is sequentially stimu-
lated by H2A-containing nucleosomes and free H2A.Z-H2B dimers, leading to eviction of nucleosomal H2A-H2B and depo-
sition of H2A.Z-H2B. These stepwise events ensure the specificity of the nucleosome replacement reaction.

Locking Chromosome Cohesion during Replication


PAGE 737
In eukaryotic cells, sister chromatids remain physically connected from the time of their
synthesis during DNA replication until their separation during mitosis. Sister chromatid
cohesion depends on the stable association of cohesin with DNA. Nishiyama et al.
now show that Sororin binds cohesin during replication and stabilizes the cohesin-
DNA complex by displacing the cohesin ‘‘unloading’’ protein Wapl. Distant orthologs
of Sororin exist in many species, implying that this may be a widespread mechanism
for the maintenance of sister chromatid cohesion.

G Protein Lockdown for Channels


PAGE 750
G protein-coupled potassium channels need to be turned off quickly, on a timescale faster than that afforded by either ligand
clearance or receptor endocytosis. Raveh et al. now show that the GPCR kinase, GRK2, achieves rapid desensitization of the
GIRK potassium channel by sequestering the G protein subunits required for GIRK activity. This kinase-independent function
of GRK2 thus allows rapid control of ligand-stimulated channel function.

Actin Cherry Picks Recycling Receptors


PAGE 761
Signaling receptors recycle efficiently during endocytosis in a manner that differs from bulk membrane recycling. Puthen-
veedu et al. use live cell imaging to show that distinct endosomal subdomains mediate active recycling of signaling receptors.
The actin cytoskeleton binds in a sequence-dependent manner to the receptors, further concentrating and stabilizing these
domains for recycling.

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 653


R&D Systems Tools for Cell Biology Research™

Cancer

Development

Endocrinology

Glycobiology

Immunology R&D Systems Products for


Neuroscience

Proteases
Neuroscience Research
R&D Systems offers a wide range of high quality products for neuroscience research. In addition to high performance
Signal Transduction antibodies, we offer the most referenced collection of premium quality proteins and ELISA kits in the industry.
Our catalog also includes primary rat and mouse cortical stem cells, and kits for the expansion, differentiation, and
Stem Cells identification of neural stem cells.

Plexin-B2 Notch-2 O4

Performance.
Results.
RGM-B
Progress. HSPH1

GFAP BSRP-A Vanilloid R1

For more information visit our website at www.RnDSystems.com/go/Neuroscience

For research use only. Not for use in diagnostic procedures.

R&D Systems, Inc. www.RnDSystems.com

R&D Systems Europe, Ltd. www.RnDSystems.co.uk

R&D Systems China Co., Ltd. www.RnDSystemsChina.com.cn


Shapewear for the ER
PAGE 774
The endoplasmic reticulum (ER) consists of the nuclear envelope and an exten-
sive peripheral network of tubules and membrane sheets. Shibata et al. demon-
strate that ER sheets are formed through stabilization of their highly curved
edges by the reticulon/DP1/Yop1 p proteins. The membrane protein Climp63
further shapes the sheets, acting as a spacer to regulate their area and luminal
width.

HIV Pushes the T Cell Self-Destruct Button


PAGE 789
The depletion of CD4 T cells during HIV infection is a hallmark of AIDS. Doitsh
et al. show that abortive infection of CD4 T cells elicits cell death. Incomplete
reverse transcripts of the virus accumulate in these cells and activate suicidal
innate antiviral and inflammatory responses. Thus, T cell death is not triggered
by new virus production but, rather, by a suicide mechanism, which likely evolved to protect the host but in fact contributes to
immunodeficiency.

Hungry but Still Hearing


PAGE 802
Caloric restriction (CR) extends the life span of many species and slows the progression of age-related hearing loss (AHL).
Here, Someya et al. report that mitochondrial Sirt3 mediates the prevention of AHL and reduces oxidative damage in
calorie-restricted mice. In response to CR, Sirt3 deacetylates and activates isocitrate dehydrogenase 2, leading to an
enhanced glutathione antioxidant defense system in mitochondria. These results suggest that Sirt3-dependent mitochondrial
adaptations may be a central mechanism to delay aging in mammals.

Outfoxing Aging
PAGE 813
Loss of muscle strength is one of the most obvious changes that we experience
as we age, but how this connects with systemic aging is unclear. Demontis and
Perrimon report that accumulation of protein aggregates in aging Drosophila
muscle is reduced by FOXO/4E-BP signaling, delaying muscle senescence.
This pathway in muscle prevents overall aging and protein aggregation in other
tissues. These results provide a framework to understand the coordination of
tissue and organismal aging.

Golgi Decides, Axon or Dendrite


PAGE 826
Neuronal cells polarize to develop an axon at one pole and dendrites at the
other. Matsuki et al. identify two signaling pathways that influence Golgi
morphogenesis to regulate this polarization. The Stk25 kinase acts through
the Golgi protein GM130 to promote a condensed Golgi morphology and axon development. The Reelin-Dab1 signaling
pathway, previously known to regulate other aspects of nervous system development, antagonizes the Stk25 pathway to
promote Golgi extension and dendrite development. Thus, Golgi distribution is a central factor in neuronal development.

Structural Fingerprints of the Human Genome


PAGE 837
Genomic structural variation—insertions, duplications, and deletions—are important contributors to human disease and
genetic diversity. The precise molecular characteristics of these variants have been difficult to ascertain by standard high-
throughput genome sequencing. Kidd et al. now report a resource of fosmid clones obtained from the genomes of 17 indi-
viduals. The authors characterize the breakpoints of more than a thousand structural variants, allowing inference of the molec-
ular pathways that generated them and offering an in-depth view of the characteristics of human genomic variation.

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 655


bioreprogramming

Biopotential.
Unlock extraordinary potential with stem cell technologies from Sigma . ®

Stem cell biology offers astonishing


research potential; Sigma® Life Science has
the innovations you need to discover the
promise it holds. Access a world of RNAi with
the MISSION® RNAi Library, efficiently edit
genes of interest using advanced CompoZr®
ZFN technology, and characterize your stem
cells with our Prestige Antibodies®, powered
by Atlas Antibodies. The applications are
endless—and so are the possibilities.

wherebiobegins.com/bioreprogramming

Sigma, MISSION, CompoZr and Prestige Antibodies are registered trademarks of


Sigma-Aldrich and its affiliate Sigma-Aldrich Biotechnology, L.P.
Leading Edge

Select: Cell Cycle

The phases of the cell cycle must be exquisitely timed and tightly regulated in order to ensure proper
chromosome replication and segregation and cell division. New findings described in this issue’s Select
address key regulatory events in the cell cycle and reveal potential clinical outcomes of errors in these
processes.

An Epigenetic License to Replicate


Chromosome replication needs to occur once and only once during the cell cycle
to produce daughter cells with accurate genetic content. Licensing of replication
origins is one form of DNA synthesis regulation, in which origins are loaded with
pre-replication complex (RC) proteins during the end of M phase and throughout
G1. Without this licensing event, replication origins cannot be activated. New
findings from Tardat et al. identify the methyltransferase PR-Set7—and the
histone modification that it catalyzes, methylation of histone H4 lysine 20
(H4K20me1)—as a key regulator of the onset of licensing in mammalian cells.
The authors show that PR-Set7 and H4K20me1 levels are cell cycle regu-
lated—both are high during M and G1 phases, dropping in S when synthesis
begins—and that proteasomal degradation of PR-Set7 is needed to prevent
DNA re-replication. The authors also show that silencing PR-Set7 leads to
Re-replicating G2 cells (cyclin B1, red; decreased chromatin loading of pre-RC proteins and reduced origin firing during
EdU, green). Image courtesy of E. Julien. S phase, whereas targeting PR-Set7 to nonorigin sites on the chromatin is suffi-
cient to induce H4K20me1 and the assembly of pre-RC proteins. Future studies
are needed to investigate how H4K20me1 facilitates chromatin loading of pre-RC proteins.
M. Tardat et al. (2010). Nat. Cell Biol. Published online October 17, 2010. 10.1038/ncb2113.

Getting a Toehold on Microtubules


The ability of the kinetochore to maintain an attachment between chromo-
somes and microtubules is necessary for proper chromosomal segregation
during anaphase. The Ndc80 complex is known to be a key regulatory site
for microtubule attachment, but, given the highly dynamic process of micro-
tubule assembly and disassembly occurring during segregation, it has been
a challenge to identify how the Ndc80 complex physically holds on to such
a rapidly changing structure. Alushin et al. address this using cryo-electron
microscopy to better reveal the metazoan Ndc80 complex bound to micro-
tubules. The authors find that the Ndc80 complex binds both a- and
b-tubulin monomers and identify a ‘‘toe’’—a short section of the NDC80
protein that recognizes a site between two tubulin monomers, a hinge point
for tubulin bending. The toe appears to prefer binding straight tubulin, sug-
gesting that it could act as a sensor for tubulin conformation. At the same
time, the N terminus of NDC80 allows high-affinity microtubule binding and
appears to mediate self-assembly of Ndc80 complexes in a manner that is
modulated via phosphorylation by Aurora B kinase. The authors propose
a model in which phosphorylated Ndc80 complexes bind a microtubule
and spindle forces then pull the bound complex out of the Aurora B kinase
phosphorylation zone. The resulting dephosphorylation of NDC80 results in Two Ndc80 molecules (blue and yellow;
high-affinity clusters forming in linear arrays along the microtubule. This N terminus, magenta) binding tubulin
cluster arrangement is consistent with a biased diffusion model of kineto- (green; C terminus, red). Image courtesy of
E. Nogales.
chore attachment and movement. On a shrinking microtubule, the Ndc80-
microtubule interaction would be reduced due to conformational changes
in tubulin at the disassembling or depolymerizing end, and the cluster would diffuse along the microtubules toward
the pole, thereby moving the chromosome in that direction.
G.M. Alushin et al. (2010). Nature 467, 805–810.

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 657


Announcing an innovative new textbook from Academic Cell
Primer to The Immune Response, Academic Cell Update Edition
By Tak W. Mak and Mary Saunders

Primer to The Immune Response, Academic Cell Update


Edition, is an invaluable resource for students who
need a concise but complete and understandable
introduction to immunology.

Academic Cell textbooks contain premium journal


content from Cell Press and are part of a new
cutting-edge textbook/journal collaboration
designed to help today’s instructors teach students to
“think like a scientist.”

academiccell.com

Primer to The Immune Response


Academic Cell Update Edition
Academic Cell is a dynamic textbook publishing partnership between
Tak W. Mak Academic Press and Cell Press, two market-leading publishers bringing
The Campbell Family Institute for Breast scientific advances from the world of life science research into the classroom.
Cancer Research, Ontario, Canada
Mary Saunders Order online now from: elsevierdirect.com/9780123847430
The Campbell Family Institute for Breast Request and examination copy from textbooks.elsevier.com
Cancer Research, Ontario, Canada

Paperback/456 pages
ISBN: 9780123847430 Twitter.com/academiccell Facebook.com/academiccell
$79.95/£54.99/€64.95
Mounting Tension in Lead-Up to Fateful Decision
Asymmetric cell division, which generates daughter cells with different developmental fates, is often achieved through
asymmetric positioning of the mitotic spindle. However, some dividing cells start out with a centered spindle that
becomes displaced during anaphase. This progressive asymmetry has been postulated to arise from greater elonga-
tion of microtubules on one side of the spindle. New findings from Ou et al. suggest that nonmuscle myosin II might also
play a role. The authors show that in the QR.a neuroblast of Caenorhabditis elegans, myosin II becomes asymmetrically
distributed during anaphase, concentrating at the anterior side of the cleavage furrow. Consequently, the anterior
membrane becomes less dynamic and shrinks inward, whereas the posterior membrane expands like a balloon, sug-
gesting that cortical tension and contractile forces driven by myosin II could be a factor in developing asymmetry. The
authors also used CALI (chromophore-assisted laser inactivation) to specifically inactivate myosin II at the anterior
membrane and find that this increases the size of the anterior daughter cell and can alter its fate from apoptosis to
differentiation into a neuron-like cell. Future work is needed to better understand the respective contributions of micro-
tubule elongation, myosin polarization, and perhaps other unknown mechanisms to the regulation of asymmetric divi-
sion and cell fate.
G. Ou et al. (2010). Science. Published online September 30, 2010. 10.1126/science.1196112.

Spindle Position, a Neuronal Mover and Maker


Human microcephaly is a neurodevelopmental disorder characterized by a small brain,
fewer surface ridges, and reduced cortical neuron numbers. Two recent papers used
linkage analysis and genome capture in affected families to identify WDR62 as
a common cause of genetic microcephaly and characterized the WDR62 protein as
a spindle pole protein expressed in mitotic neural precursors. After sequencing
affected individuals to identify specific disease-causing mutations, Nicholas et al. ex-
pressed mutant WDR62 in HeLa cells and showed that the normal accumulation of the
protein at the spindle poles during mitosis is disrupted. Given the phenotype of
reduced neuron numbers and small brain seen in microcephaly, one possibility the
authors suggest is that WDR62 could be involved in proper positioning of the mitotic
spindle and cleavage furrow, such that mutant WDR62 results in insufficient symmetric
Photograph of human microce- divisions—needed to produce neural precursors—early in cortical development. In
phalic brain. Image courtesy of agreement, Yu et al. show that the brain of an affected individual has profound cortical
C. Walsh. defects, with thin sparse cortical layers and aberrant repositioning of neurons to
subcortical regions, suggesting deficits in neurogenesis and migration. Further
description of the specific role of WDR62 at the spindle will clarify how it is involved in cerebral development and
aid in our understanding of the etiology of microcephaly.
A.K. Nicholas et al. (2010). Nat. Genet. Published online October 3, 2010. 10.1038/ng.682.
T.W. Yu et al. (2010). Nat. Genet. Published online October 3, 2010. 10.1038/ng.683.

Rebecca Alvania

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 659


Expand your stem cell library and save
today on the latest books on stem cells
and regenerative medicine

Stem Cells Stem Cell Anthology


Scientific Facts and Fiction From Stem Cell Biology, Tissue Engineering,
Christine Mummery, Ian Wilmut, Anja Van de Stolpe Regenerative Medicine, Cloning and Stem
and Bernard Roelen
Cell Methods
November 2010 | 400 pages | Paperback | Bruce M. Carlson
$79.95 | €57.95 | £48.99 | October 2009 | 450 pp. | Hardback | $150.00 |
ISBN: 9780123815354 €100.00 | £95.00 |AU$222.00 |
ISBN: 9780123756824

Principles of Regenerative
Medicine, 2nd Edition Essential Stem Cell Methods
Anthony Atala and Robert Lanza A Volume in the Reliable Lab Solutions Series
November 2010 | 1400 pages | Hardback | Robert Lanza and Irina Klimanskaya
$199.95 | €143.00 | £125.00 | April 2009 | 628 pp. | Paperback | $75.00 | €50.95
ISBN: 9780123814227 | £45.99 |AU$111.00 | ISBN: 9780123750617

Heart Development and Tissue Engineering


Regeneration, 2-Volume Set Clemens van Blitterswijk, Peter Thomsen, Jeffrey Hubbell,
Ranieri Cancedda, Anders Lindahl Sahlgrenska,
Nadia Rosenthal and Richard P. Harvey
Jerome Sohier and David F. Williams
June 2010 | 1072 pp. | Hardback
| $199.95 | €143.00 | £125.00 | March 2008 | 760 pp. | Hardback | $115.00 |
AU$296.00 | ISBN: 9780123813329 €76.95 | £69.99 |AU$170.00 |
ISBN: 9780123708694

Essentials of Stem Cell


Biology, 2nd Edition Human Stem Cell Manual
Robert Lanza, Roger Pedersen, John Gearhart, E. A Laboratory Guide
Donnall Thomas, Brigid Hogan, James Thomson, Jeanne F. Loring, Robin L. Wesselschmidt and Philip H. Schwartz
Douglas Melton and Sir Ian Wilmut June 2007 | 488 pp. | Spiral bound | $88.95 |
June 2009 | 600 pp. | Hardback | $199.95 €59.95 | £53.99 |AU$132.00 |
| €134.00 | £125.00 | AU$302.00 | ISBN: ISBN: 9780123704658
9780123747297

Handbook of Stem Cells


Foundations of 2-Volume Set with CD-ROM Vol. 1–2
Regenerative Medicine Vol. 1 – Embryonic Stem Cells
Clinical and Therapeutic Applications Vol. 2 – Adult & Fetal Stem Cells
Anthony Atala, Robert Lanza, James Thomson and Robert Lanza, Roger Pedersen, Helen Blau, E. Donnall Thomas,
Robert Nerem John Gearhart, James Thomson, Brigid Hogan, Catherine
Verfaillie, Douglas Melton, Irving Weissman, Malcolm Moore
September 2009 | 750 pp. | Hardback |
and Michael West
$99.95 | €66.95 | £60.99|AU$148.00 |
ISBN: 9780123750853 September 2004 | 1,760 pp. | Hardback | $566.00
| €380.00 | £345.00 | AU$817.00 |
ISBN: 9780124366435

Cell Stem Cell subscribers save 25% on their book order


Secure ordering online at elsevierdirect.com
Enter promo code 28024 at check out
Prices and publication dates subject to change without notice.
Your Power for Health

Challenging Experiments?
Advanced TC™ cell culture vessels from Greiner Bio-One
Innovative polymer modification improves cellular
adhesion
Positive effects on cell functionality and performance
Enhanced propagation of fastidious cells
Improved cell expansion under limited growth
conditions
Better assay consistency
Long-term stability and storage at room temperature

Germany (Main office): Greiner Bio-One GmbH, info@de.gbo.com l Austria: Greiner Bio-One GmbH, office@at.gbo.com
Belgium: Greiner Bio-One BVBA/SPRL, info@be.gbo.com l Brazil: Greiner Bio-One Brasil, office@br.gbo.com l China: Greiner Bio-One GmbH, info@cn.gbo.com
France: Greiner Bio-One SAS, infos@fr.gbo.com l Japan: Greiner Bio-One Co. Ltd., info@jp.gbo.com l Netherlands: Greiner Bio-One B.V., info@nl.gbo.com
UK: Greiner Bio-One Ltd., info@uk.gbo.com l USA: Greiner Bio-One North America Inc., info@us.gbo.com www.gbo.com/bioscience
¬,OOK¬!GAIN

$ISCOVER¬-ORE
s¬!CCESS¬TO¬THE¬¬#ELL¬0RESS¬PRIMARY¬RESEARCH¬JOURNALS¬AND¬
¬4RENDS¬REVIEWS¬TITLES ¬ALL¬ON¬THE¬SAME¬PLATFORM
s¬)MPROVED ¬MORE¬ROBUST¬ARTICLE¬AND¬AUTHOR¬SEARCH
s¬6IDEO ¬ANIMATIONS¬AND¬SOUND¬lLES
s¬%ASY¬TO¬NAVIGATE¬HOME¬PAGE ¬ARTICLES¬PAGES¬AND¬ARCHIVE

WWWCELLCOM
017.A1.0115.A © 2010 Eppendorf AG
 Joystick provides
intuitive control

microinjection

search level

B
A
 Patented axial
injection level
C
injection movement
of the capillary
carrier

 Semi-Automatic
microinjection into
adherent cells

 Pre-pull capillaries for


reproducible injection
of adherent cells
InjectMan
NI 2

Microinjection simplified!
Microinjection is one of the core methods to introduce Eppendorf InjectMan NI 2 microinjector has it all:
foreign DNA and other non-permeable molecules into  Motorized X-Y-Z movements provide precise movement
cells. Nuclear injection of plasmid DNA enables rapid  Pre-setting and storage of up to 2 locations in X-Y-Z,
expression of proteins in specific cells within saves time in returning to pre-set work locations
a population.  Automated Home function for rapid capillary exchange
 Joystick-controlled provides overall ergonomic manipulator
The menu-controlled, programmable micromanipulator  Fine adjustment of work speed made easy with
InjectMan NI 2 is ideally suited for microinjection of adherent positioning wheel
cells. Connection with the FemtoJet and the axial mounting  Can be adapted to all common microscopes
allows injections at 45˚ angle reducing cell damage during
injection and increases cell viability. This guarantees a very For more information visit www.eppendorf.com
rapid, safe and reproducible microinjection process.

www.eppendorf.com • Email: info@eppendorf.com


In the U.S.: Eppendorf North America, Inc. 800-645-3050 • In Canada: Eppendorf Canada Ltd. 800-263-8715
experience
[ constant contamination control ]
Closed Loop HEPA Filtration
NuAire's closed loop HEPA filtration system creates a mini ISO Class 5 cleanroom inside
Microbiological CO2 Incubators
the growth chamber. Air and gas pass through 99.99% HEPA filters to create Class 100 air.
All filters are kept out of the growth chamber ensuring a true contaminate free work zone.
The chamber is maintained at positive pressure to prevent potential contaminates from
entering the growth chamber.

Sensitivity and Accuracy of Gas Control


A microprocessor-based, non-dispersive, digital solid-state infrared [IR] CO2 sensor
provides a stable drift-free output requiring less calibrations.

Precision of Electronic Microprocessor Controls


Run by a programmable micro-controller, AutoFlow™ samples input sensors, sets control
outputs, and makes necessary corrections to the chamber environment.

Dual Sterilization Cycles - Self Cleaning System


The DHD AutoFlow™ NU-5510 features dual sterilization cycles to eradicated contamination
in the growth chamber. The chamber heats up to 95°C Humidified or 145°C Dry for a
constant uniform high temperature on chamber walls, brackets, shelves, and water pan.

®
ASCB booth 622
Follow, Find, Enjoy:
2100 Fernbrook Lane | Plymouth, MN 55447 | U.S.A. | 763.553.1270 | WWW.NUAIRE.COM
Leading Edge
Previews

ER Sheets Get Roughed Up


Charles Barlowe1,*
1Department of Biochemistry, Dartmouth Medical School, Hanover, NH 03755, USA

*Correspondence: charles.barlowe@dartmouth.edu
DOI 10.1016/j.cell.2010.11.011

The molecular machinery that shapes the endoplasmic reticulum’s (ER’s) membrane into ordered
networks of ‘‘smooth’’ tubules and ‘‘rough’’ sheets is poorly defined. Shibata et al. (2010) now report
that sheet-inducing proteins, such as Climp-63, are enriched in the ‘‘rough’’ ER by their association
with membrane-bound ribosomes, whereas curvature-inducing proteins localize at highly bent
edges of membrane sheets.

The elaborate morphologies of the endo- a wedge into ER membranes (Figure 1). groups of ribosomes bound to the ER
plasmic reticulum have fascinated cell Indeed, reconstitution of purified reticulon membranes (i.e., polysomes), proteins
biologists for years. Compartments of the and DP1 proteins into synthetic lipo- of the translocon complex redistribute
endoplasmic reticulum (ER) membrane somes (i.e., artificial vesicles with a lipid between ER sheets and tubules. This
form the nuclear envelope and then bilayer) was sufficient to generate mem- finding suggests that actively translating
extend throughout the cell periphery in brane tubules with a high degree of curva- polysomes concentrate translocon com-
an interconnected network of mem- ture (Hu et al., 2008). Thus, intrinsic plexes into sheet subdomains of the ER.
brane tubules and flattened discs called properties of the reticulon and DP1 pro- To identify the structural components
cisternae. How do these ordered arrays teins are sufficient to induce membrane of these ER sheet domains, Shibata and
of membranes form, and how are their tubulation. colleagues then perform a proteomic
structures connected to their cellular However, ER tubules also form analysis of rough ER membranes from
function? In this issue of Cell, Shibata branched, reticular morphologies. Gener- pancreatic secretory cells. Indeed, the
and coworkers define a class of sheet- ation of these net-like structures requires most abundant protein constituents in
inducing membrane proteins that are en- additional factors, specifically atlastin ER sheets are components of the translo-
riched in the ribosome-studded ‘‘rough’’ GTPases, which drive fusion of ER con complex and Climp-63. Moreover,
ER. These proteins cooperate with tubules into branched networks (Hu microarray experiments reveal that
membrane curvature-stabilizing factors et al., 2009; Orso et al., 2009). Of interest, Climp-63 messenger RNA (mRNA) levels
to govern the relative level of sheets atlastin isoforms were detected in associ- are among the most highly induced
and tubules of the ER, providing a molec- ation with the reticulon proteins, sug- messages during proliferation of ER sheet
ular basis for the longstanding morpho- gesting that the formation of tubules and structures during the differentiation of
logical descriptions of ‘‘rough’’ and branching are coordinated processes immature B cells into IgG secreting
‘‘smooth’’ ER. (Hu et al., 2009). plasma cells. Climp-63 is an ER trans-
ER morphologies vary greatly across In contrast to our understanding of membrane protein that contains an
different species and cell types. For ER tubules, the molecular mechanisms extended coiled-coil domain in the interior
example, highly active secretory cells, underlying the formation of ER sheets of the ER (i.e., the ER lumen). Previous
such as pancreatic exocrine cells and have been elusive. Now, Shibata et al. studies suggested that this coiled-coil
plasma B cells, are packed full of flattened (2010) uncover an unexpected connec- domain contributes to ER morphology
cisternae of rough ER. Live cell imaging tion between the sheet-inducing factor by forming a scaffold in the ER lumen
also reveals that ER membranes are Climp-63 and the reticulon and DP1 (Klopfenstein et al., 2001).
highly dynamic networks, undergoing proteins. Their discovery begins with To test the functional role of Climp-63
constant remodeling often in response to a key observation regarding the translo- in ER sheet formation, Shibata and
physiological conditions. con complex, a large multisubunit chan- colleagues then overexpress Climp-63 in
Previous studies focusing on the nel that transports, or ‘‘translocates,’’ cultured cells, which causes a dramatic
smooth ER found that tubule formation nascent polypeptides across ER mem- proliferation of ER sheets. Moreover, the
depends on a class of integral membrane brane into the interior of the ER. distance between the sheets is 50 nm,
proteins belonging to the reticulon and Shibata and colleagues observe that the standard distance between ER sheets
DP1 families (Voeltz et al., 2006). Reticu- components of the translocon complex in mammalian cells (Figure 1). In contrast,
lon and DP1 proteins are highly enriched are not only highly enriched in ER sheets, decreasing the expression of Climp-63
in tubular ER elements, and they contain but they also form a specialized subdo- does not deplete cells of ER sheets, but
transmembrane segments with a double main within ER membranes. Moreover, instead, it causes a marked reduction in
hairpin structure that induces positive when the authors treat cells with the anti- the distance between cisternal sheets.
membrane curvature by inserting like biotic puromycin, which disassembles Further, these sheets are spread diffusely

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 665


throughout the cytoplasm, Finally, sheets and tubules
a similar phenotype as the are not the only morphologies
authors observe when they of ER membranes. For
treat cells with puromycin. example, specialized struc-
Finally, Climp-63 and the tural domains of the ER are
reticulon protein Rtn4 have involved in metabolism of
opposing effects on ER mor- hydrophobic compounds,
phology. Increased expres- formation of ER-mitochon-
sion of Rtn4 reduces the drial junctions, transport of
number of ER sheets, Ca2+, formation of lipid drop-
whereas co-overexpression lets, and protein export from
with Climp-63 restores sheet ER subdomains called transi-
structures in these cells. tional ER sites. The molecular
Importantly, reticulon pro- machinery that generates
teins strikingly localize to the these ER structures awaits
highly curved edges of ER elucidation. Although the
sheets, and this occurs components that sculpt ER
when reticulon genes are sheets and tubules might
expressed at endogenous Figure 1. Molecular Model for the Generation of ER Membrane also contribute to the mor-
levels or when both Climp-63 Sheets and Tubules phology of these other struc-
Cross-section of an endoplasmic reticulum (ER) cisterna showing the curva-
and reticulon genes were ture-inducing proteins reticulons and DP1 (purple) enriched in highly bent tures, it seems likely that
overexpressed together. membrane tubules and edges of the sheet. In contrast, the sheet-inducing novel mechanisms will also
The authors then propose protein Climp-63 (blue) is excluded from tubules and, instead, partitions into be discovered.
sheet domains with translocon complexes. Climp-63 could assemble into
the most basic mechanism parallel coiled-coil arrangements to flatten membranes and to serve as luminal
for sheet formation that is ER spacers that keep individual sheets a specific distance apart (50 nm in
REFERENCES
also consistent with their find- mammalian cells).
ings. In this model, reticulons Hu, J., Shibata, Y., Voss, C., She-
and DP1 proteins partition mesh, T., Li, Z., Coughlin, M.,
into the edges of sheets, where they 1998), suggesting an additional level of Kozlov, M.M., Rapoport, T.A., and Prinz, W.A.
induce a high degree of curvature at the ER organization that is connected to the (2008). Science 319, 1247–1250.
edges of closely apposed membrane bila- cell’s overall structure. Hu, J., Shibata, Y., Zhu, P.-P., Voss, C., Rismanchi,
yers (Figure 1). However, assembling the Although reticulon and DP1 proteins N., Prinz, W.A., Rapoport, T.A., and Blackstone, C.
ordered array of rough ER membranes in partition into sheet edges in vivo and ex- (2009). Cell 138, 549–561.
active secretory cells also depends on pressing Climp-63 drives ER sheet prolif- Klopfenstein, D.R., Kappeler, F., and Hauri, H.P.
the coiled-coil domain of Climp-63, which eration, it is still unknown whether these (1998). EMBO J. 17, 6168–6177.
serves as a spacer between the sheets factors are sufficient for sheet formation Klopfenstein, D.R., Klumperman, J., Lustig, A.,
in the ER lumen (Figure 1). Lastly, the or whether other factors contribute to Kammerer, R.A., Oorschot, V., and Hauri, H.P.
authors propose that Climp-63, together this process. A minimally reconstituted (2001). J. Cell Biol. 153, 1287–1300.
with translocon complexes, partition into liposome system successfully demon- Nikonov, A.V., Hauri, H.P., Lauring, B., and Krei-
sheet domains with membrane-bound strated that reticulon and DP1 proteins bich, G. (2007). J. Cell Sci. 120, 2248–2258.
polysomes to generate the rough ER. drive tubule formation in vitro (Hu et al.,
Orso, G., Pendin, D., Liu, S., Tosetto, J., Moss,
This model proposed by Shibata and 2008). This system should provide a T.J., Faust, J.E., Micaroni, M., Egorova, A., Marti-
colleagues is also supported by previous powerful tool for determining whether nuzzi, A., McNew, J.A., and Daga, A. (2009). Nature
studies showing that the coiled-coil adding purified Climp-63 is sufficient 460, 978–983.
domain of Climp-63 assembles into for sheet formation. Furthermore, varying Shibata, Y., Shemesh, T., Prinz, W.A., Palazzo,
a-helical rods that are required to restrict the ratio of curvature- and sheet- inducing A.F., Kozlov, M.M., and Rapoport, T.A. (2010).
the lateral mobility of Climp-63 (Klopfen- proteins in liposomes of defined lipid Cell 143, this issue, 774–788.
stein et al., 2001; Nikonov et al., 2007) compositions could provide insights into Voeltz, G.K., Prinz, W.A., Shibata, Y., Rist, J.M.,
(Figure 1). Moreover, Climp-63 is known the role that specific lipids play in gener- and Rapoport, T.A. (2006). Cell 124,
to bind microtubules (Klopfenstein et al., ating observed ER morphology. 573–586.

666 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


Leading Edge
Previews

SIRT3 in Calorie Restriction:


Can You Hear Me Now?
Carlos Sebastian1 and Raul Mostoslavsky1,*
1The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA

*Correspondence: rmostoslavsky@mgh.harvard.edu
DOI 10.1016/j.cell.2010.11.009

Caloric restriction decreases oxidative damage and extends life span in many organisms. Someya
et al. (2010) show that the sirtuin SIRT3 mediates the protective effects of caloric restriction on age-
related hearing loss by promoting the mitochondrial antioxidant system through the regulation of
isocitrate dehydrogenase 2 (Idh2).

Despite two decades of effort, caloric eration in mouse models. Remarkably, previous studies (Schwer et al., 2009).
restriction remains the only treatment early work demonstrated that caloric However, whereas wild-type mice display
demonstrated to extend life span and to restriction slows age-related hearing loss lower levels of serum insulin and triglycer-
delay the progression of several diseases in animal models (Sweet et al., 1988). ides when fed a calorie-restricted diet,
normally associated with aging, such as Moreover, in their previous work, Prolla SIRT3-deficient mice do not show this
cancer, diabetes, and neurological disor- and colleagues demonstrated that caloric response. Based on these results, the
ders. Early experiments in yeast showed restriction induces expression of the authors argue that SIRT3 plays a role in
that the life span extension mediated by SIRT3 gene in the cochlea (Someya et al., metabolic adaptations to caloric restric-
caloric restriction involves Sir2, the found- 2007). They now elegantly follow up on tion. It remains unclear, however, whether
ing member of the sirtuin family of histone these results, proving a role for this sirtuin SIRT3 can also mediate the effects of
deacetylases. However, later experi- in the delay in hearing loss due to caloric calorie restriction in other tissues or
ments have questioned this association restriction and elucidating the molecular whether it does so specifically in the
(Longo and Kennedy, 2006), and the role mechanisms underlying this effect. context of hearing loss.
of mammalian sirtuins in life span exten- Someya et al. use wild-type and SIRT3- The authors then investigate the molec-
sion by caloric restriction is still under deficient mice fed a diet in which caloric ular mechanisms involved in this process.
study. In this context, although SIRT1 intake is reduced to 75% and compare Given that caloric restriction reduces age-
appears to be the major mammalian sir- them to control mice fed with a regular associated oxidative damage to macro-
tuin involved in the metabolic effects of diet. The authors first look at the hearing molecules (Sohal and Weindruch, 1996),
caloric restriction (Haigis and Guarente, response of the animals and find that, as Someya et al. analyze levels of oxidative
2006), the precise role of sirtuins in the expected, aging leads to hearing loss in damage to DNA in several tissues. They
longevity response remains unclear. In both wild-type and SIRT3-deficient mice. find that a calorie-restricted diet reduces
this issue of Cell, Someya et al. (2010) However, whereas caloric restriction this type of damage in wild-type mice,
bring some light to the field by describing delays the progression of hearing loss in but not in SIRT3-deficient animals. Impor-
a new function for the mitochondrial wild-type mice, this effect is completely tantly, this is the first evidence that
SIRT3 protein in the prevention of hearing abolished in SIRT3-deficient animals. a mammalian sirtuin regulates levels of
loss mediated by caloric restriction during These results are consistent with the oxidative stress in response to caloric
aging. These tantalizing results suggest effects of caloric restriction on spiral restriction.
that SIRT3 might play an important role ganglion neurons and hair cells in these But how does SIRT3 regulate oxidative
in slowing the aging process in mammals. mice. In wild-type animals, a calorie damage during caloric restriction? Given
Age-related hearing loss is a hallmark of restricted diet reduces the age-related that SIRT3 localizes to the mitochondria,
mammalian aging and the most common loss of neurons and hair cells, whereas the authors hypothesize that SIRT3 could
sensory disorder in the elderly (Liu and this effect is abrogated in SIRT3-deficient regulate the antioxidant systems present
Yan, 2007). It is characterized by a gradual mice. Together, these results clearly pin- in this organelle. Using a combination of
loss of spiral ganglion neurons and point SIRT3 as a critical molecular determi- cellular and biochemical experiments,
sensory hair cells in the cochlea of the nant regulating the response to caloric they discover that SIRT3 regulates the
inner ear (Liu and Yan, 2007). Given that restriction in age-related hearing loss. mitochondrial glutathione antioxidant
the affected cells are postmitotic and do The authors next study the metabolic defense system. Glutathione is the main
not regenerate, their loss leads to an effects induced by caloric restriction in small molecule antioxidant in cells and is
age-associated decline in hearing func- SIRT3-deficient mice. With a normal diet, generated by glutathione reductase in
tion. Several groups have studied hearing SIRT3-deficient animals appear pheno- a reaction dependent on NADPH. The
loss as an example of age-related degen- typically normal, in accordance with authors show that SIRT3 modulates the

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 667


conversion of oxidized gluta- us closer to a healthier life
thione to reduced glutathione span. In the words of Francois
in response to caloric restric- Jacob, ‘‘In a world of unlimited
tion. They find that, under imagination, we are continu-
these conditions, SIRT3 ally inventing a possible world
binds and deacetylates the or a piece of a world, and then
mitochondrial isocitrate de- comparing it with the real
hydrogenase 2 enzyme world.’’ In the context of sir-
(Idh2), the enzyme that gener- tuins, it seems we are starting
ates NADPH, increasing the to put some of these pieces
enzyme’s activity. In agree- together.
ment with these results, Idh2
deacetylation and activity, as
ACKNOWLEDGMENTS
well as NADPH levels, in-
crease during caloric restric- We would like to thank all of the
tion in all wild-type tissues members of the Mostoslavsky lab
tested, whereas SIRT3 defi- for helpful comments. C.S. is the
ciency impairs this response. recipient of a Beatriu de Pinos Post-
Finally, overexpressing SIRT3 doctoral Fellowship (Generalitat de
Catalunya). R.M. is a Sidney Kimmel
and Idh2 promotes cell
Scholar, a Massachusetts Life
viability upon oxidative dam- Science Center New Investigator
age. Together, these data Scholar, and the recipient of an
lead the authors to propose AFAR Award. Work in the Mosto-
a model in which caloric slavsky lab is funded, in part, by
restriction promotes SIRT3 National Institutes of Health.
expression, leading to the de-
acetylation and activation of
REFERENCES
Idh2, thus providing resis- Figure 1. Caloric Restriction, SIRT3, and Age-Related Hearing Loss
tance to oxidative stress and During aging (left), oxidative damage (ROS, reactive oxygen species) leads to Finkel, T., Deng, C.X., and Mosto-
inhibiting the age-related the loss of spiral ganglion neurons and sensory hair cells in the ear, leading to slavsky, R. (2009). Nature 460,
age-related hearing loss. Caloric restriction (right) prevents the age-associ-
loss of spiral ganglion neu- ated loss of spiral ganglion neurons and sensory hair cells. Someya et al.
587–591.
rons and hair cells (Figure 1). (2010) show that caloric restriction leads to an increase in SIRT3 levels in Haigis, M.C., and Guarente, L.P.
Although Someya et al. the mitochondria. By promoting the deacetylation of isocitrate dehydrogenase (2006). Genes Dev. 20, 2913–2921.
provide enough data to 2 (Idh2), SIRT3 promotes the accumulation of NADPH, hence activating gluta-
thione reductase (GSR), which generates reduced glutathione (GSH), a cellular Hirschey, M.D., Shimazu, T., Goetz-
prove that the effects of antioxidant. man, E., Jing, E., Schwer, B.,
caloric restriction on age- Lombard, D.B., Grueter, C.A., Har-
related hearing loss are ris, C., Biddinger, S., Ilkayeva, O.R.,
dependent on SIRT3, key et al. (2010). Nature 464, 121–125.
questions remain. First, does SIRT3 effects of caloric restriction using SIRT3 Liu, X.Z., and Yan, D. (2007). J. Pathol. 211,
mediate the effects of caloric restriction activators? If so, such reagents would 188–197.
in other tissues? And if so, what are its have significant therapeutic potential. Longo, V.D., and Kennedy, B.K. (2006). Cell 126,
substrates? Multiple mitochondrial Finally, because other sirtuins also have 257–268.
proteins are deacetylated upon caloric prominent roles in metabolic regulation Schwer, B., Eckersdorff, M., Li, Y., Silva, J.C., Fer-
restriction in a SIRT3-dependent manner (Finkel et al., 2009), can we extend min, D., Kurtev, M.V., Giallourakis, C., Comb, M.J.,
(Schwer et al., 2009). It is therefore some of these findings to other sirtuins? Alt, F.W., and Lombard, D.B. (2009). Aging Cell 8,
604–606.
important to determine whether Idh2 is SIRT1, for example, has been linked to
the main SIRT3 target in preventing the response of mammals to caloric Sohal, R.S., and Weindruch, R. (1996). Science
273, 59–63.
oxidative stress or whether other SIRT3 restriction (Haigis and Guarente, 2006),
Someya, S., Yamasoba, T., Weindruch, R., Prolla,
substrates contribute as well. Second, and it is therefore possible that the
T.A., and Tanokura, M. (2007). Neurobiol. Aging
what is the relationship between the activity of this and other sirtuins may be 28, 1613–1622.
effect of SIRT3 on Idh2 and the recently regulated in a coordinated fashion
Someya, S., Yu, W., Hallows, W.C., Xu, J., Vann,
described role for SIRT3 in fatty acid following nutrient starvation. J.M., Leeuwenburg, C., Tanokura, M., Denu,
oxidation during nutrient stress (Hirschey Regardless of the utopian dream of life J.M., and Prolla, T.A. (2010). Cell 143, this issue,
et al., 2010)? Are these functions coordi- span extension, answering some of these 802–812.
nated? If they are not, how is specificity questions may provide a step forward for Sweet, R.J., Price, J.M., and Henry, K.R. (1988).
achieved? Third, can we mimic the treating age-related pathologies, bringing Audiology 27, 305–312.

668 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


Leading Edge
Previews

ATP Consumption Promotes


Cancer Metabolism
William J. Israelsen1 and Matthew G. Vander Heiden1,*
1Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA

*Correspondence: mvh@mit.edu
DOI 10.1016/j.cell.2010.11.010

Cancer cells metabolize glucose by aerobic glycolysis, a phenomenon known as the Warburg
effect. Fang et al. (2010) show that the endoplasmic reticulum enzyme ENTPD5 promotes ATP
consumption and favors aerobic glycolysis. The findings suggest that nutrient uptake in cancer
cells is limited by ATP and satisfies energy requirements other than ATP production.

Mounting evidence suggests that cancer Activation of the PI3K pathway in ENTPD5 leads to decreased levels of
cells engage in a unique metabolic pro- cancer can occur via genetic alterations several growth factor receptors, including
gram that allows for rapid cell prolifera- that allow growth factor-independent epidermal growth factor receptor (EGFR),
tion. Nonproliferating cells can use glycol- kinase activation or via the loss of PTEN, insulin-like growth factor receptor
ysis products to generate ATP for their a lipid phosphatase that attenuates PI3K b (IGFR-b), and Her2/ErbB2. Given that
energy needs. Such cells generally have signaling. Fang et al. now find that cell growth factor signaling plays an important
low rates of glycolysis followed by extracts from PTEN-deficient cells have role in increasing nutrient metabolism in
oxidation of pyruvate in the mitochondria, an enhanced ability to generate AMP rapidly proliferating cells (DeBerardinis
leading to efficient generation of ATP. from ATP. Subsequent purification and et al., 2008), these new findings suggest
Dividing cells, in contrast, also use glycol- biochemical characterization of this that cellular ATP levels can influence the
ysis intermediates for the synthesis of activity led to the identification of ectonu- folding and expression of growth factor
macromolecules and must therefore cleoside triphosphate diphosphohydro- receptors, perhaps ensuring that cells do
balance their ATP requirements and lase 5 (ENTPD5) as the enzyme associ- not attempt to grow when ATP is limiting.
biosynthetic needs (Vander Heiden et al., ated with the ATP hydrolysis activity. Furthermore, because glucose metabo-
2009). Metabolism of glucose by aerobic PI3K signaling leads to upregulation of lism by the hexosamine biosynthesis
glycolysis, a phenomenon known as the ENTPD5, a UDPase that promotes the pathway provides the carbon for these
Warburg effect, may help dividing cells N-glycosylation and folding of glycopro- receptor glycosylation events, the avail-
strike this balance. teins in the ER by hydrolyzing UDP to ability of glucose may provide a means
The phosphoinositide 3-kinase (PI3K) UMP (Trombetta and Helenius, 1999) to couple nutrient levels with growth
signaling pathway, which is activated in (Figure 1). UDP hydrolysis in the ER is factor receptor expression. These feed-
many cancers, regulates cell growth and a reaction necessary to promote protein backs may exist to prevent a metabolic
survival. PI3K signaling has been impli- folding via the calnexin/calreticulin catastrophe caused by activation of the
cated in the altered glucose metabolism pathway. It is linked to ATP hydrolysis in cell growth machinery when the supply
of cancer cells, and the serine/threonine the cytosol by a cycle of glucose and of nutrients or ATP is limiting.
kinase AKT, a major PI3K effector, phosphate transfer reactions. As part How does ENTPD5 regulate ATP
promotes glucose uptake and increases of this cycle, the UDP-glucose/UMP anti- levels? Fang et al. find that reconstitution
the activity of glycolytic enzymes (DeBer- porter exports UMP out of the ER in of the ATP consuming activity also
ardinis et al., 2008). In this issue of Cell, exchange for importing UDP-glucose requires the presence of UMP/CMP
Fang et al. (2010) report an important into the ER (Hirschberg et al., 1998). The kinase-1 and adenylate kinase-1. UMP/
mechanism by which AKT signaling leads UGGT enzyme then uses UDP-glucose CMP kinase-1 catalyzes the rephosphor-
to increased aerobic glycolysis. They to transfer glucose to proteins in the ER ylation of the UMP generated by ENTPD5
show that AKT activation promotes protein (Vembar and Brodsky, 2008). This into UDP (Figure 1), in the process con-
glycosylation in the endoplasmic retic- glucose addition to nascent glycoproteins verting ATP to ADP. Adenylate kinase-1
ulum, which elevates ATP consumption is necessary for their calnexin/calreticulin- then converts ADP molecules into ATP
and derepresses a rate-limiting enzyme mediated protein folding. Thus, disruption and AMP, thus allowing the cycle to
in glycolysis that is otherwise inhibited by of ENTPD5 in PTEN-deficient cells results continue. Surprisingly, this cycle involving
an elevated ratio of ATP to AMP. This in decreased protein N-glycosylation and ENTPD5 is a major source of ATP
work suggests how proliferating cells causes ER stress. consumption in PTEN-deficient cells.
may integrate growth signals with energy Cell surface proteins, including many Furthermore, these reactions directly
status to enable increased glucose uptake growth factor receptors, are N-glycosy- affect the cell’s glycolytic rate. Whereas
to support cell proliferation. lated. Fang et al. show that disruption of increased ENTPD5 expression has no

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 669


are dependent on activated PI3K, sug-
gesting that increased ENTPD5 activity
may not be a universal mechanism for
lowering ATP levels in tumors. However,
other enzymes involved in regulating
nucleotide pools in cells have also been
linked to cancer (Hartsough and Steeg,
2000), and there are additional homologs
of ENTPD5 whose functions are not well
understood. These enzymes may be
involved in analogous cycles of ATP con-
sumption, leading to enhanced glucose
metabolism in other genetic contexts.
Fang et al. also show that decreased
ENTPD5 expression inhibits tumor
growth, possibly via pleiotropic effects
involving induction of ER stress and
altered glucose metabolism. Consider-
ation of ENTPD5 as a potential thera-
peutic target in PI3K-driven cancer is
interesting given that pharmacological
inhibition of ENTPD5 is predicted to
Figure 1. ENTPD5 Promotes Glycolysis in Proliferating Cells
Fang et al. (2010) show that the endoplasmic reticulum (ER) UDPase ectonucleoside triphosphate decrease tumor ATP consumption.
diphosphohydrolase 5 (ENTPD5) is expressed in response to phosphoinositide 3-kinase (PI3K) signaling. Although counterintuitive, the resulting
Activation of PI3K results in FOXO phosphorylation by AKT and loss of ENTPD5 transcriptional repres- increase in ATP/AMP ratio might reduce
sion. This leads to increased ENTPD5 enzyme activity in the ER, promoting protein folding. ENTPD5
activity promotes the import of UDP-glucose into the ER, where UGGT transfers glucose to an unfolded
the entry of glucose into glycolysis and
N-glycoprotein and produces UDP. Properly folded N-glycoproteins, such as growth factor receptors, starve the cells of precursors necessary
exit the cycle, whereas unfolded proteins undergo further folding attempts or are degraded. ENTPD5 for biosynthesis. Successful efforts to
activity enables this process by hydrolyzing UDP to provide the UMP necessary for exchange with
target cancer metabolism will likely arise
UDP-glucose in the cytosol. The activities of UMP/CMP kinase-1 and adenylate kinase-1 couple the
energetic requirements of this cycle to the net conversion of ATP to AMP. Thus, increased ENTPD5 from understanding the feedbacks and
activity leads to a decrease in the cellular ATP/AMP ratio. Because this ratio is the major determinant complex regulation that are required for
of glucose flux through the phosphofructokinase (PFK) step in glycolysis, a lowered ATP/AMP ratio anabolic metabolism. The study by Fang
increases glycolysis, elevates lactate production, and provides glycolytic intermediates for biomass
production. et al. provides new insight by demon-
strating that ATP consumption serves to
increase glucose flux to satisfy the ener-
getic and biosynthetic demands of
effect on cellular respiration, it increases ATP is likely not the growth-limiting a rapidly proliferating cell.
lactate production, suggesting a link resource for most cells (Vander Heiden
between ATP consumption and in- et al., 2009). The concept that glucose ACKNOWLEDGMENTS
creased glycolytic flux. In a series of utilization by tumor cells may be limited
We thank Brooke Bevis for her help preparing the
experiments to determine how ENTPD5 by ATP consumption to prevent feedback
figure and editing the manuscript. M.G.V.H. is
increases glucose entry into glycolysis, inhibition of PFK has been suggested a consultant to Agios Pharmaceuticals regarding
Fang et al. find that the ratio of fructose- previously (Scholnick et al., 1973). This development of compounds targeting cancer
6-phosphate to fructose-1-6-bisphos- study finally provides a mechanism by metabolism and is a member of its Scientific Advi-
phate increases in cells following ENTPD5 which cells can increase ATP consump- sory Board.
knockdown, consistent with inhibition of tion to drive glucose uptake. An additional
this step in glycolysis. Phosphofructoki- mechanism has also recently been REFERENCES
nase (PFK), the enzyme that catalyzes described in which glucose incorporation
DeBerardinis, R.J., Lum, J.J., Hatzivassiliou, G.,
this reaction, is the major enzyme control- into biosynthetic pathways occurs
and Thompson, C.B. (2008). Cell Metab. 7, 11–20.
ling glucose commitment to the glycolytic without producing excess ATP (Vander
Dunaway, G.A. (1983). Mol. Cell. Biochem. 52,
pathway (Dunaway, 1983). A high ATP/ Heiden et al., 2010). Together, these 75–91.
AMP ratio in the cell inhibits both PFK studies support the notion that altered
Fang, M., Shen, Z., Huang, S., Zhao, L., Chen, S.,
activity and glucose metabolism by metabolism in cancer is not adapted to Mak, T.M., and Wang, X. (2010). Cell 143, this
glycolysis. In fact, the authors conclude support ATP production. issue, 711–724.
that increased ATP consumption by Fang et al. show that ENTPD5 expres- Hartsough, M.T., and Steeg, P.S. (2000). J. Bioen-
ENTPD5 increases glycolysis by lowering sion correlates with PI3K activation in erg. Biomembr. 32, 301–308.
the ATP/AMP ratio and relieving allosteric human prostate cancer cell lines and Hirschberg, C.B., Robbins, P.W., and Abeijon, C.
inhibition of PFK. tumor tissue samples. Not all cancer cells (1998). Annu. Rev. Biochem. 67, 49–69.

670 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


Scholnick, P., Lang, D., and Racker, E. (1973). Vander Heiden, M.G., Cantley, L.C., and Thomp- Asara, J.M., and Cantley, L.C. (2010). Science
J. Biol. Chem. 248, 5175. son, C.B. (2009). Science 324, 1029–1033. 329, 1492–1499.
Vander Heiden, M.G., Locasale, J.W., Swanson,
Trombetta, E.S., and Helenius, A. (1999). EMBO J. K.D., Sharfi, H., Heffron, G.J., Amador-Noguez, Vembar, S.S., and Brodsky, J.L. (2008). Nat. Rev.
18, 3282–3292. D., Christofk, H.R., Wagner, G., Rabinowitz, J.D., Mol. Cell Biol. 9, 944–957.

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 671


Leading Edge
Essay

Glycomics Hits the Big Time


Gerald W. Hart1,* and Ronald J. Copeland1
1Department of Biological Chemistry, School of Medicine, Johns Hopkins University, 725 North Wolfe Street, Baltimore, MD 21205-2185, USA

*Correspondence: gwhart@jhmi.edu
DOI 10.1016/j.cell.2010.11.008

Cells run on carbohydrates. Glycans, sequences of carbohydrates conjugated to proteins and


lipids, are arguably the most abundant and structurally diverse class of molecules in nature. Recent
advances in glycomics reveal the scope and scale of their functional roles and their impact on
human disease.

By analogy to the genome, transcriptome, O-GlcNAc (Hart et al., 2007). Even though Dynamic Structural Complexity
or proteome, the ‘‘glycome’’ is the the generic term ‘‘glycosylation’’ is often Underlies Glycan Functions
complete set of glycans and glycoconju- used to categorize and lump all glycan Glycoconjugates provide dynamic struc-
gates that are made by a cell or organism modifications of proteins into one bin, tural diversity to proteins and lipids that
under specific conditions. Therefore, side by side with other posttranslational is responsive to cellular phenotype, to
‘‘glycomics’’ refers to studies that attempt modifications such as phosphorylation, metabolic state, and to the developmental
to define or quantify the glycome of a cell, acetylation, ubiquitination, or methylation, stage of cells. Complex glycans play crit-
tissue, or organism (Bertozzi and Sasise- such a view is not only inaccurate, but ical roles in intercellular and intracellular
kharan, 2009). In eukaryotes, protein also is completely misleading. If one only processes, which are fundamentally
glycosylation generally involves the cova- considers the linkage of the first glycan important to the development of multicel-
lent attachment of glycans to serine, to the polypeptide in both prokaryotic lularity (Figure 1). Unlike nucleic acids and
threonine, or asparagine residues. Glyco- and eukaryotic organisms, there are at proteins, glycan structures are not hard-
proteins occur in all cellular compart- least 13 different monosaccharides and wired into the genome, depending upon
ments. Glycans are also attached to 8 different amino acids involved in glyco- a template for their synthesis. Rather,
lipids, often ceramide, which is comprised protein linkages, with a total of at least the glycan structures that end up on
of sphingosine, a hydrocarbon amino 41 different chemical bonds known to be a polypeptide or lipid result from the
alcohol and a fatty acid. Complex glycans linking the glycan to the protein (Spiro, concerted actions of highly specific gly-
are mainly attached to secreted or cell 2002). Importantly, each one of these cosyltransferases (Lairson et al., 2008),
surface proteins, and they do not cycle unique glycan:protein linkages is surely which in turn are dependent upon the
on and off of the polypeptide. In contrast, as different in both structure and function concentrations and localization of high-
the monosaccharide O-linked N-acetyl- as protein methylation is from acetylation. energy nucleotide sugar donors, such as
glucosamine (O-GlcNAc) cycles rapidly Of course, this modification is not only UDP-N-acetylglucosamine, the endpoint
on serine or threonine residues of many about a single linkage. When structural of the hexosamine biosynthetic pathway.
nuclear and cytoplasmic proteins. Identi- diversity of the additional oligosaccharide Therefore, the glycoforms of a glycopro-
fying the number, structure, and function branches of glycans and the added diver- tein depend upon many factors directly
of glycans in cellular biology is a daunting sity of complex terminal saccharides on tied to both gene expression and cellular
task but one that has been made easier in glycans, such as fucose or sialic acids metabolism.
recent years by advances in technology (about 50 different sialic acids are known There are at-least 250 glycosyltrans-
and by our growing appreciation of how [Schauer, 2009]), are taken into account, ferases in the human genome, and it has
integral glycans are to biology (Varki the molecular diversity and varied func- been estimated that about 2% of the
et al., 2009). tions of protein-bound glycans rapidly human genome encodes proteins
The scope of the glycomics challenge is increase exponentially. Just the ‘‘sia- involved in glycan biosynthesis, degrada-
immense. The covalent addition of lome’’ (Cohen and Varki, 2010) rivals or tion, or transport (Schachter and Freeze,
glycans to proteins and lipids represents exceeds many other posttranslational 2009). Biosynthesis of the nucleotide
not only the most abundant posttransla- modifications in abundance and struc- sugar donors is directly regulated by nu-
tional modification (PTM), but also by far tural/functional diversity. In addition, cleic acid, glucose, and energy metabo-
the most structurally diverse. Although it chemical modifications, such as phos- lism, and the compartmentalization of
is commonly stated that more than 50% phorylation, sulfation, and acetylation, these nucleotide sugar donors is highly
of all polypeptides are covalently modified increase the glycan structural/functional regulated by specific transporters. Protein
by glycans (Apweiler et al., 1999), even diversity even more. Thus, categorizing glycosylation is therefore controlled by
this estimate is far too low because it fails glycosylation as a single type of post- rates of polypeptide translation and
to include that myriad nuclear and translational modification is neither useful protein folding, localization of and compe-
cytoplasmic proteins are modified by nor at all reflective of reality. tition between glycosyltransferases,

672 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


it is estimated that the binding sites of
glycan-binding proteins (GBPs), such as
antibodies, lectins, receptors, toxins, mi-
crobial adhesions, or enzymes (Figure 1),
can accommodate only up to two to six
monosaccharides within a glycan struc-
ture (Cummings, 2009). Therefore, the
number of specific glycan substructures
that bind to biologically important GBPs
in a cell may be fewer than 10,000,
a number that is within the realm of
current analytical and, if targeted, chemi-
cal or enzymatic synthetic capabilities.
Until recently, the lack of tools and the
inherent complexity of glycans have
been major barriers preventing most biol-
ogists from embracing the importance of
glycans in biology. Recent technological
advances have significantly lowered these
barriers. Indeed, the tools of glycomics
and the subfields of glycoproteomics, gly-
colipidomics, and proteoglycomics have
all progressed substantially in recent
years (Krishnamoorthy and Mahal, 2009;
Laremore et al., 2010). Major technolog-
ical advances, many of which are shared
with proteomics, have recently allowed
Figure 1. Glycans Permeate Cellular Biology semiquantitative profiling of glycans and
Complex glycans at the cell surface are targets of microbes and viruses, regulate cell adhesion and devel- glycoproteins (Krishnamoorthy and
opment, influence metastasis of cancer cells, and regulate myriad receptor:ligand interactions. Glycans Mahal, 2009; Vanderschaeghe et al.,
within the secretory pathway regulate protein quality control, turnover, and trafficking of molecules to
organelles. Nucleocytoplasmic O-linked N-acetylglucosamine (O-GlcNAc) has extensive crosstalk with
2010). Some of these advances are the
phosphorylation to regulate signaling, cytoskeletal functions, and gene expression in response to nutrients result of the National Institute of General
and stress. Medical Science’s (NIGMS) support of
the Consortium for Functional Glycomics
cellular concentration and localization of gene expression of glycan-processing (CFG), which has served to focus and
nucleotide sugars, the localization of enzymes, by polypeptide structure at all assist more than 500 researchers on
glycosidases, and membrane trafficking. levels, and by cellular metabolism. issues related to glycomics (Paulson
Thus, individual glycosylation sites on the et al., 2006; Raman et al., 2006).
same polypeptide can contain different Technology of Glycomics Kobata and colleagues were among the
glycan structures that reflect both the A detailed understanding of cellular first to profile N-glycans, well before the
type and status of the cell in which they processes will require a detailed appreci- current concepts of glycomics were
are synthesized. For example, the glyco- ation of the glycans modulating proteins conceived. Despite the lack of many
forms of the membrane protein Thy-1 are and pathways. Although this ultimate modern methods, their pioneering work
very different in lymphocytes than they goal of glycomics is laudable, we are was characterized by a high level of rigor
are in brain, despite having the same poly- a very long way from having the tech- in defining the arrays of N-glycan struc-
peptide sequence (Rudd and Dwek, nology to completely characterize the gly- tures present in cells and tissues and on
1997). Conversely, even small changes in come of even a simple cell or tissue. Not specific proteins (Endo, 2010). Currently,
polypeptide sequence or structure will only is the glycome much more complex a wide variety of high-resolution and
alter the types of glycan structures than the genome, transcriptome, or pro- highly sensitive methods are available,
attached to a polypeptide. For example, teome, as noted above, it is also much including capillary electrophoresis (CE),
histocompatibility antigen polypeptides more dynamic, varying considerably not high-performance liquid chromatography
with more than 90% sequence homology only with cell type, but also with the (HPLC), and lectin microarrays.
contain different N-linked glycan profiles developmental stage and metabolic state Glycans are often profiled after their
at individual sites, reflective of their of a cell. Even very conservative esti- release from polypeptides, which results in
allelic type, even when they are synthe- mates indicate that there are well over the loss of any information about proteins
sized within the same cells (Swiedler a million different glycan structures in and sites to which they were attached.
et al., 1985). Thus, site-specific protein a mammalian cell’s glycome. However, Even though it is much more difficult, it is
glycosylation is highly regulated by upon considering ‘‘functional glycomics,’’ also much preferable to perform

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 673


glycopeptide profiling (glycoproteomics) to development of purified recombinant lec- custom-made DNA microarrays that
first identify attachment sites prior to tins, and better definition of the specific- represent glycosyltransferases and
detailed profiling or structural analysis of ities of many lectins (Gupta et al., 2010). glycan-binding proteins. The CFG also
the glycans present on a polypeptide. The Both matrix-assisted laser desorption has developed databases that present
ultimate goal of glycoproteomics, which is ionization (MALDI) and electrospray phenotypic and biochemical data on gly-
to define all of the molecular species (glyco- mass spectrometry have played a key cosyltransferase knockout mice. Even
forms) of glycoproteins in a cell or tissue, role in glycan profiling and in glycoproteo- though knocking out a single glycosyl-
has not yet been realized for any glycopro- mics (An et al., 2009; North et al., 2010; transferase gene often affects hundreds
tein with more than one glycan attachment Zaia, 2010). For biomarker discovery, of glycoconjugates and myriad biological
site. N-glycans are generally released from affinity enrichment approaches, based processes, these mutant mice have
proteins by peptide-N-glycosidase F upon chemical modification and solid- proven valuable in revealing the funda-
(PNGase F), which cleaves most, but not phase extraction of N-linked glycopro- mental biological importance of glycans.
all, N-glycans. Unfortunately, no such teins, have proven useful in profiling The microarrays and the databases
broadly specific enzyme exists for N-linked glycoprotein sites from serum- produced by the CFG member community
O-glycans, which are generally released or even from paraffin-embedded tissues at large are publically available on the CFG
by chemical methods, such as alkali- (Tian et al., 2009). Recently, using lectin website (http://www.functionalglycomics.
induced b elimination, or by hydrazinolysis. binding combined with advanced mass org) and have resulted in a profound
However, for relatively pure glycoproteins, spectrometric methods, thousands of increase in our understanding of the
so called ‘‘top-down’’ mass spectrometric N-glycan attachment sites have been binding specificities of GBPs, including
methods, which do not involve prior release mapped, a prerequisite for understanding lectins key to inflammation and immunity,
of the glycans, may eventually prove useful, their functions (Zielinska et al., 2010). and on infectious microbes or viruses.
as instrumentation and methods improve Given the structural diversity of However, a major barrier preventing
(Reid et al., 2002). glycans, all of these glycomic approaches glycan biology from being incorporated
Due to the small sample sizes involved, generate vast amounts of data. Glycan bi- more into the mainstream is the continued
most CE or HPLC separation methods oinformatics has made great strides failure by the community to adopt a univer-
require chemical modification of released within recent years with major efforts sally standard glycan structural format
glycans with fluorescent compounds. CE from several laboratories (Aoki-Kinoshita, and database that are easily accessed
and HPLC methods provide high-resolu- 2008). At least four major publicly worldwide. Most importantly, glycan data-
tion separation of glycans, and when available carbohydrate databases (Glyco- bases must eventually be incorporated
combined with laser-induced fluorescent sciences.de, KEGG GLYCAN, Euro- into standard interactive databases that
detection (LIF), tagged glycans can be de- carbDB, and CFG) are now maintained, are supported by public agencies (such
tected in the low femtomole range. High and efforts to structure them in a uniform as NCBI or EMBL) before glycan biology
pH anion-exchange chromatography format have been in progress for quite can be fully integrated into the wider
(HPAEC) with pulsed-amperometric some time. In addition, the Carbohy- research community.
detection separates glycans with high drate-Active EnZyme database (CAZy)
resolution and detects them with high has played a key role in providing a global From Glycomics to Biology
sensitivity without chemical modification, understanding of carbohydrate active Glycans are directly involved in almost
but the high alkalinity employed can be enzymes, documenting their evolutionary every biological process and certainly
problematic for some labile structures. relationships, providing a framework for play a major role in nearly every human
Lectins, which are defined as carbohy- elucidating common mechanisms, and disease (Figure 1). Genetic studies in
drate-binding proteins that are neither establishing the relationship between gly- tissue culture cells indicate that specific
antibodies nor enzymes, have a wide cogenomics and glycomes expressed by complex glycan structures are generally
range of glycan binding specificities, suit- cells (Cantarel et al., 2009). Moreover, not essential to a cell growing in culture,
able for partial characterization of a gly- recent advances in bioinformatic analysis indicating that most of the functions of
come. Lectin microarrays use methods tools for complex glycomic mass spec- complex glycans are at the multicellular
and equipment similar to that employed trometry data sets have allowed complex level. In contrast, the cycling monosac-
for nucleic acid arrays. Given the large data to be presented in formats useful to charide, O-GlcNAc, on nuclear and cyto-
number of different lectins available, lectin nonexperts in all fields of biology (Ceroni plasmic proteins, is essential even at the
microarrays can provide information et al., 2008; Goldberg et al., 2005). single cell level in mammals (Hart et al.,
about the glycome in a high-throughput Perhaps one of the most important 2007).
fashion, which is particularly useful in contributions to the field of functional The critical roles of glycans in mammals
profiling glycans produced by infectious glycomics has been the development of are now well established not only by the
organisms (Hsu et al., 2006). In the future, well-defined glycan microarrays, which dearth of mutations in glycan biosynthetic
it is highly likely that glycomics will play currently display more than 500 different enzymes that survive development, but
a central role in combating infectious glycan structures (Smith et al., 2010). also by the severe phenotypes generated
disease. However, many technical issues The NIGMS-supported Consortium for when such mutations are not lethal.
remain to be resolved, such as standard- Functional Glycomics (CFG) has gener- These severe phenotypes are clearly illus-
ization required for clinical use, the ated and made publicly available trated by the congenital disorders of

674 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


landscape, the pharmaceutical industry
and the US Food and Drug Administration
are rapidly realizing the critical importance,
in terms of both bioactivity and safety, of
carefully defining the glycoforms of any
therapeutics derived from glycoconju-
gates.

Glycoproteomics, Glycolipidomics,
and Biomarkers
Clinical cancer diagnostic markers are
often glycoproteins, but most current
diagnostic tests only measure the expres-
sion of the polypeptide. Clearly, given the
Figure 2. Glycomic Complexity Reflects Cellular Complexity long known alterations in glycans associ-
Given that glycan structures are regulated by metabolism and glyco-enzyme expression and glycans ated with cancer, it is highly likely that
modify both proteins and lipids, functional glycomics also requires the tools of genomics, proteomics, lip- cancer markers that detect specific glyco-
idomics, and metabolomics (modified after Packer et al., 2008).
forms of a protein will have much higher
sensitivity and specificity for early
glycosylation (CDGs) (Schachter and organization of receptors on the cell detection of cancer (Packer et al., 2008;
Freeze, 2009), which are associated with surface and play important roles in immu- Taniguchi, 2008). Thus, the convergence
severe mental and developmental abnor- nity, infections, development, and inflam- of glycomics and glycoproteomics is key
malities. Also, the severe muscular mation (Lajoie et al., 2009). Proteoglycans to the discovery of biomarkers for the early
dystrophy that results from defective and glycosaminoglycans play a key role in detection of cancer (Taylor et al., 2009).
O-glycosylation of a-dystroglycan (Yosh- the regulation of growth factors, in micro- Recently, the Food and Drug Administra-
ida-Moriguchi et al., 2010) further bial binding, in tissue morphogenesis, and tion has approved fucosylated a-fetopro-
illustrates how a mutation in a glycan in the etiology of cardiovascular disease. tein as a diagnostic marker of primary
biosynthetic enzyme results in a devas- Proteoglycans are perhaps the most hepatocarcinoma. In addition, fucosy-
tating disease. The interplay between complicated and information-rich mole- lated haptoglobin may be a much better
O-GlcNAcylation and phosphorylation on cules in biology, and progress in proteo- marker of pancreatic cancer than simply
nuclear and cytoplasmic proteins plays glycomics has begun to accelerate monitoring the expression of the hapto-
a key role in the etiology of diabetes, (Ly et al., 2010). Nearly all microbes and globin polypeptide. Indeed, The National
neurodegenerative disease, and cancer viruses that infect humans bind to cells Cancer Institute has begun an initiative to
(Hart et al., 2007; Zeidan and Hart, 2010). by attaching to specific cell surface discover, develop, and clinically validate
It has long been appreciated that alter- glycans. Glycomics and glycan arrays glycan biomarkers for cancer (http://
ations in cell surface glycans contribute to will have a substantial impact upon future glycomics.cancer.gov/). System biology
the metastatic and neoplastic properties research toward both diagnosing and analyses of the glycome to identify
of tumor cells (Taniguchi, 2008). The func- preventing infectious disease. biomarkers of human disease will, by
tions of many receptors are modulated by Some of the most important drugs on the necessity, also employ many of the same
their glycans, such as modulation of market are already the result of glycomics. methods used by genomics, proteomics,
Notch receptors by the action of specific The anti-flu virus drugs Relenza and Tami- metabolomics, and lipidomics (Figure 2)
glycosyltransferases (Moloney et al., flu are structural analogs of sialic acids that (Packer et al., 2008). Due to the critical
2000), which regulate Notch’s activation inhibit the flu virus neuraminidase and the roles of glycans in cardiovascular disease
by its ligands, affecting many develop- transmission of the virus. Natural heparin, and lung disease and in the functions of
mental events. Selectins, which specifi- a sulfated glycosaminoglycan, and chemi- blood cells, the National Heart Lung and
cally bind to a subset of fucosylated and cally defined synthetic heparin oligosac- Blood Institute (NHLBI) has recognized
sialylated glycans, play a critical role in charides have long been widely used in an acute need to train more researchers
leukocyte homing to sites of inflamma- the clinic as anticoagulants and for many in the area of glycosciences by creating
tion. Indeed, a selectin inhibitor is other clinical uses. Hyaluronic acid, a non- a ‘‘Program of Excellence in Glycoscien-
currently in phase two clinical trials for sulfated glycosaminoglycan, is used in the ces,’’ which will not only support collabo-
vaso-occlusive sickle cell disease (Chang treatment of arthritis. Many recombinant rative research, but will also provide
et al., 2010). Siglecs, which are a family of pharmaceuticals, including therapeutic hands-on laboratory training in the
cell surface sialic acid-binding lectins, monoclonal antibodies, are glycoproteins, methods of glycosciences to fellows.
play a fundamental role in regulating and their specific glycoforms are key to Thus, though our knowledge about the
lymphocyte functions and activation. their bioactivity and half lives in circulation biology of glycans and glycomics
Recent studies on galectins, a family of and to their possible induction of delete- continues to lag behind more mainstream
b-galactoside-binding lectins, have rious immune responses when they do fields of genomics and proteomics, tech-
shown that they play a critical role in the not contain the correct glycans. Given this nological advances in glycomics in the

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 675


last 5 years have begun to accelerate the Chang, J., Patton, J.T., Sarkar, A., Ernst, B., Mag- Raman, R., Venkataraman, M., Ramakrishnan, S.,
integration of glycobiology into the other nani, J.L., and Frenette, P.S. (2010). Blood 116, Lang, W., Raguram, S., and Sasisekharan, R.
1779–1786. (2006). Glycobiology 16, 82R–90R.
major fields of biomedical research. A
complete mechanistic understanding of Cohen, M., and Varki, A. (2010). OMICS 14, Reid, G.E., Stephenson, J.L., Jr., and McLuckey,
455–464. S.A. (2002). Anal. Chem. 74, 577–583.
the etiology of almost any disease will
depend upon the elucidation of the func- Cummings, R.D. (2009). Mol. Biosyst. 5, 1087– Rudd, P.M., and Dwek, R.A. (1997). Crit. Rev. Bio-
1104. chem. Mol. Biol. 32, 1–100.
tions of all posttranslational modifications
Endo, T. (2010). J. Biochem. 147, 9–17. Schachter, H., and Freeze, H.H. (2009). Biochim.
but will especially depend upon our
Goldberg, D., Sutton-Smith, M., Paulson, J., and Biophys. Acta 1792, 925–930.
understanding the many roles of glycans,
Dell, A. (2005). Proteomics 5, 865–875. Schauer, R. (2009). Curr. Opin. Struct. Biol. 19,
the most abundant and structurally
diverse type of posttranslational modifi- Gupta, G., Surolia, A., and Sampathkumar, S.G. 507–514.
(2010). OMICS 14, 419–436. Smith, D.F., Song, X., and Cummings, R.D. (2010).
cation.
Hart, G.W., Housley, M.P., and Slawson, C. (2007). Methods Enzymol. 480, 417–444.
Nature 446, 1017–1022.
Spiro, R.G. (2002). Glycobiology 12, 43R–56R.
ACKNOWLEDGMENTS Hsu, K.L., Pilobello, K.T., and Mahal, L.K. (2006).
Swiedler, S.J., Freed, J.H., Tarentino, A.L.,
Nat. Chem. Biol. 2, 153–157.
We thank Dr. Chad Slawson for helpful sugges- Plummer, T.H., Jr., and Hart, G.W. (1985). J. Biol.
Krishnamoorthy, L., and Mahal, L.K. (2009). ACS Chem. 260, 4046–4054.
tions. Original research in the author’s laboratory
Chem. Biol. 4, 715–732.
was supported by NIH grants R01CA42486, R01 Taniguchi, N. (2008). Mol. Cell. Proteomics 7,
DK61671, and R24 DK084949. Lairson, L.L., Henrissat, B., Davies, G.J., and 626–627.
Withers, S.G. (2008). Annu. Rev. Biochem. 77,
Taylor, A.D., Hancock, W.S., Hincapie, M., Tanigu-
521–555.
chi, N., and Hanash, S.M. (2009). Genome Med 1,
REFERENCES Lajoie, P., Goetz, J.G., Dennis, J.W., and Nabi, I.R. 57.
(2009). J. Cell Biol. 185, 381–385.
An, H.J., Froehlich, J.W., and Lebrilla, C.B. (2009). Tian, Y., Gurley, K., Meany, D.L., Kemp, C.J., and
Laremore, T.N., Leach, F.E., III, Solakyildirim, K., Zhang, H. (2009). J. Proteome Res. 8, 1657–1662.
Curr. Opin. Chem. Biol. 13, 421–426. Amster, I.J., and Linhardt, R.J. (2010). Methods
Aoki-Kinoshita, K.F. (2008). PLoS Comput. Biol. 4, Enzymol. 478, 79–108. Vanderschaeghe, D., Festjens, N., Delanghe, J.,
e1000075. and Callewaert, N. (2010). Biol. Chem. 391,
Ly, M., Laremore, T.N., and Linhardt, R.J. (2010).
149–161.
Apweiler, R., Hermjakob, H., and Sharon, N. OMICS 14, 389–399.
(1999). Biochim. Biophys. Acta 1473, 4–8. Varki, A., Cummings, R.D., Esko, J.D., Freeze,
Moloney, D.J., Panin, V.M., Johnston, S.H., Chen,
H.H., Stanley, P., Bertozzi, C.R., Hart, G.W., and
Bertozzi, C.R., and Sasisekharan, R. (2009). Glyco- J., Shao, L., Wilson, R., Wang, Y., Stanley, P.,
Etzler, M.E. (2009). Essentials of Glycobiology,
mics. In Essentials of Glycobiology, Second Irvine, K.D., Haltiwanger, R.S., and Vogt, T.F.
Second Edition (Cold Spring Harbor, NY: Cold
Edition, A. Varki, R.D. Cummings, J.D. Esko, H.H. (2000). Nature 406, 369–375.
Spring Harbor Laboratory Press).
Freeze, P. Stanley, C.R. Bertozzi, G.W. Hart, and North, S.J., Jang-Lee, J., Harrison, R., Canis, K.,
M.E. Etzler, eds. (Cold Spring Harbor, NY: Cold Yoshida-Moriguchi, T., Yu, L., Stalnaker, S.H.,
Ismail, M.N., Trollope, A., Antonopoulos, A.,
Spring Harbor Laboratory Press). Davis, S., Kunz, S., Madson, M., Oldstone, M.B.,
Pang, P.C., Grassi, P., Al-Chalabi, S., et al.
Schachter, H., Wells, L., and Campbell, K.P.
Cantarel, B.L., Coutinho, P.M., Rancurel, C., (2010). Methods Enzymol. 478, 27–77.
(2010). Science 327, 88–92.
Bernard, T., Lombard, V., and Henrissat, B. Packer, N.H., von der Lieth, C.W., Aoki-Kinoshita,
(2009). Nucleic Acids Res. 37(Database issue), Zaia, J. (2010). OMICS 14, 401–418.
K.F., Lebrilla, C.B., Paulson, J.C., Raman, R.,
D233–D238. Rudd, P., Sasisekharan, R., Taniguchi, N., and Zeidan, Q., and Hart, G.W. (2010). J. Cell Sci. 123,
Ceroni, A., Maass, K., Geyer, H., Geyer, R., Dell, A., York, W.S. (2008). Proteomics 8, 8–20. 13–22.
and Haslam, S.M. (2008). J. Proteome Res. 7, Paulson, J.C., Blixt, O., and Collins, B.E. (2006). Zielinska, D.F., Gnad, F., Wisniewski, J.R., and
1650–1659. Nat. Chem. Biol. 2, 238–248. Mann, M. (2010). Cell 141, 897–907.

676 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


Leading Edge
Essay

What Determines the Specificity


and Outcomes of Ubiquitin Signaling?
Fumiyo Ikeda,1 Nicola Crosetto,1 and Ivan Dikic1,*
1Frankfurt Institute for Molecular Life Sciences and Institute of Biochemistry II, Goethe University School of Medicine, Theodor-Stern-Kai 7,

D-60590 Frankfurt (Main), Germany


*Correspondence: ivan.dikic@biochem2.de
DOI 10.1016/j.cell.2010.10.026

Ubiquitin signals and ubiquitin-binding domains are implicated in almost every cellular process, but
how is their functionality achieved in cells? We assess recent advances in monitoring the dynamics
and specificity of ubiquitin networks in vivo and discuss challenges ahead.

Introduction of thousands of proteins involved in very (Virdee et al., 2010). The possibility of
A small protein modifier, ubiquitin, is the different cellular processes. Here we heterotypic ubiquitin chains (that is, with
building block of a repertoire of molecular summarize the most recent advances in mixed linkages) has been shown in vitro,
signals spanning from single ubiquitin understanding specificity determinants but their presence and biological func-
to ubiquitin chains of different linkage in ubiquitin signaling and discuss future tions in vivo remain to be confirmed. Alto-
used for posttranslational modification of challenges in the development of sensi- gether, monoubiquitin and homotypic
dozens of cellular proteins (Hershko and tive and reliable methods for monitoring polyubiquitin chains comprise no more
Ciechanover, 1998). The seven lysines spatial and temporal patterns of ubiquiti- than ten signal types. However, the ability
(K) of ubiquitin (K6, K11, K27, K29, K33, nation in vivo. to synthesize homotypic chains of various
K48, and K63) and the amino-terminal lengths indicates that the repertoire of
methionine (M1) are connected to the Diversity of Ubiquitin Signals ubiquitin signals in the cell may be larger
C-terminal glycine for chain assembly, Despite its relatively rigid globular struc- than expected.
generating polymers (Ikeda and Dikic, ture, ubiquitin is one of the most versatile
2008; Iwai and Tokunaga, 2009). Ubiquitin signaling molecules in the cell. Although Signals Decoders:
signals are recognized and processed the surface of ubiquitin is mostly com- Ubiquitin-Binding Domains
by specialized ubiquitin-binding domains posed of polar residues, it is through its Ubiquitin signals are read and processed
(UBDs) that form transient, noncovalent few hydrophobic patches that it interacts by UBDs that bind noncovalently to
interactions either with ubiquitin moieties with most UBDs (Dikic et al., 2009; Winget mono- or polyubiquitin chains. To date,
or with the linkage region in their chains. and Mayor, 2010). Moreover, the pres- five structural folds are known with
So far, roughly 200 intracellular proteins ence of seven lysine residues and the more than 20 UBDs identified overall
have been recognized to contain one N-terminal methionine within ubiquitin (Dikic et al., 2009). UBDs are commonly
or more UBDs (Dikic et al., 2009). Ubiqui- that can be fused to the C-terminal di- a-helical structures, zinc fingers, pleck-
tin-UBD interactions regulate almost glycine motif of another ubiquitin allows strin homology (PH) folds, or similar to
every aspect of cellular physiology, the formation of polymeric chains en- the ubiquitin-conjugating (Ubc) domain
including protein degradation, receptor dowed with flexibility, as in the case of present in E2 enzymes (Dikic et al.,
trafficking, DNA repair, cell-cycle pro- K63-linked or M1-linked chains (often 2009). In the majority of cases, isolated
gression, gene transcription, autophagy, referred to as linear) (Ikeda and Dikic, UBDs preferentially bind to monoubiquitin
and apoptosis (recently reviewed in 2008; Iwai and Tokunaga, 2009). K48- via a conserved hydrophobic patch sur-
Deshaies and Joazeiro, 2009; Kirkin linked and K11-linked chains adopt rounding isoleucine 44. The measured
et al., 2009; Raiborg and Stenmark, a more rigid conformation, in which ubiq- affinity of isolated UBDs for monoubiqui-
2009; Ulrich and Walden, 2010; Wickliffe uitin monomers are tightly packed against tin typically falls in the micromolar range
et al., 2009; Winget and Mayor, 2010). each other. This creates unique modules (Dikic et al., 2009; Winget and Mayor,
Yet, very little is known about the nature composed of aligned ubiquitin moieties 2010). In certain cases, monoubiquitin-
of ubiquitin signals and the dynamics in which the hydrophobic patch contain- UBD interactions have also been demon-
of their interpretation by UBDs in the ing isoleucine 44 is either embedded or strated in the context of endogenous full-
highly crowded molecular environment facing out toward the surface (Pickart size proteins. For example, UBDs present
of the cell. In particular, it remains unclear and Fushman, 2004; Bremm et al., 2010; in Y family polymerases performing DNA
how a relatively limited pool of signals Matsumoto et al., 2010). Conversely, translesion synthesis bind the monoubi-
(ubiquitin chains and UBDs) with partially K6-linked chains form an asymmetric quitinated sliding clamp PCNA (Bienko
overlapping biochemical properties can compact conformation distinct from any et al., 2005), and monoubiquitinated
orchestrate the localization and function other known type of ubiquitin chain transmembrane receptors are recognized

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 677


by endocytic sorting proteins containing regions and shows a 10-fold higher However, the importance of this regula-
diverse UBDs (Hicke and Dunn, 2003). affinity for diubiquitin over monoubiquitin tory mechanism in vivo remains to be
The affinity of UBD-containing proteins (Zhang et al., 2009). These observations further investigated. Therefore, the de-
for their monoubiquitinated targets in the suggest that the function of tandem UBD coding of ubiquitin signals might be per-
cellular environment, however, may be arrays is to increase the affinity for a given formed in vivo by different UBDs (not
different from that inferred from in vitro ubiquitinated substrate rather than simul- necessarily endowed with absolute selec-
studies. In fact, the way ubiquitin signals taneously binding multiple substrates. tivity toward monoubiquitin or a particular
are decoded in cells may be influenced chain type) embedded in key proteins
by multiple factors, including the pres- Specificity and Plasticity controlling a particular process. Although
ence of tandem copies of one UBD in of Ubiquitin Signaling this scenario could allow a certain degree
the same protein, oligomerization, and Historically, distinct ubiquitin signals have of plasticity in ubiquitin signaling, speci-
protein compartmentalization (reviewed been linked to specific cellular functions. ficity might be determined by the localiza-
in Dikic et al., 2009; Winget and Mayor, For example, K48-linked chains, also tion and assembly of UBD-containing
2010). known as ‘‘classical’’ ubiquitin chains, proteins and enzymes catalyzing ubiquiti-
In addition to monoubiquitin, many were originally described as the signal nation reactions.
UBDs display either relative or absolute that targets substrates for proteasomal
selectivity for certain types of chains degradation (Hershko and Ciechanover, Catching Ubiquitin Signaling
(Ikeda and Dikic, 2008; Dikic et al., 2009; 1998). Nonclassical linkage types, such in the Act
Winget and Mayor, 2010). For instance, as K63-, K11-, or M1-linked chains are The huge discrepancy between our
the Pru (Plextrin receptor for ubiquitin) instead associated with DNA repair current understanding of the ubiquitin
domain in the proteasome receptor regulation, cell-cycle progression, innate system from in vitro studies compared to
Rpn13 preferentially interacts with K48- immunity, and inflammation (Ikeda and in vivo models stems from the fact that
linked diubiquitin (Husnjak et al., 2008), Dikic, 2008; Iwai and Tokunaga, 2009; ubiquitination and its recognition and
and the NZF (Npl4 zinc finger) domain in Matsumoto et al., 2010; Wickliffe et al., cleavage occur in milliseconds (Pierce
TAK1-binding protein 2 (TAB2) binds 2009). Recent reports, however, have et al., 2009), therefore making it chal-
specifically to K63-linked ubiquitin (Kula- challenged the notion that distinct chain lenging to analyze these events in living
thu et al., 2009; Sato et al., 2009). In con- types exclusively regulate specific pro- systems. The first attempts to study ubiq-
trast, UBDs in NEMO and ABIN proteins cesses in the cell. For instance, nonclas- uitin signaling in vivo have used anti-
(UBAN) bind linear diubiquitin chains sical ubiquitin signals, such as K11 bodies against monoubiquitin, polyubi-
with approximately 100-fold higher affinity chains generated by the anaphase- quitin chains, or, more recently, selective
than K63 or K48 chains, and binding to promoting complex (APC/C), can also linkages, including K11, K48, K63, and
monoubiqutitin could not be detected target selected substrates for proteaso- linear chains (Matsumoto et al., 2010;
(Rahighi et al., 2009; Lo et al., 2009). The mal degradation (Jin et al., 2008). In yeast, Newton et al., 2008; Wang et al., 2008;
selectivity of UBAN for linear chains has cyclin B1 is modified by a mix of K48-, Tokunaga et al., 2009) (Figure 1A). Raising
been explained by the observation that K63-, and K11-linked chains rather than linkage-selective antibodies is not easy,
a NEMO dimer binds symmetrically to by K48 chains alone (Kirkpatrick et al., despite being urgently needed to provide
linear diubiquitin, involving direct interac- 2006). This heterogeneous pool is suffi- tools to discriminate between different
tions with residues exposed in the cient to bind to proteasomal ubiquitin chain types in the cell. These antibodies
glycine-methionine linkages (Rahighi receptors and drive cyclin B1 degradation were produced either by synthesizing
et al., 2009). In addition, the crystal struc- (Kirkpatrick et al., 2006). Furthermore, peptides resembling specific linkage
tures of the NZF domain of TAB2 and linear chains, initially discovered as acti- bonds (Wang et al., 2008; Tokunaga
TAB3 in complex with K63-linked diubi- vators of the NF-kB pathway (Tokunaga et al., 2009) or by using the phage-display
quitin have shown a two-sided ubiquitin- et al., 2009), can also trigger proteasomal method (Matsumoto et al., 2010; Newton
binding surface thanks to a flexible degradation when fused to artificial sub- et al., 2008). Although chain-selective
K-linkage positioned away from the strates (Zhao and Ulrich, 2010). antibodies have been used to demon-
interaction surface (Kulathu et al., 2009; So, how is functional specificity of ubiq- strate specific chain formation in several
Sato et al., 2009). Linkage selectivity can uitin signaling achieved in vivo? Even biological settings (such as the NF-kB
also result from multivalent interaction though evidence indicates that specific pathway and cell-cycle progression), their
between tandem UBD arrays in a given chain types control distinct molecular ability to monitor substrates with low
protein and ubiquitin monomers or link- processes, as clearly exemplified by abundance and the dynamics of chain
ages in a polyubiquitin chain. Tandem NF-kB signaling, we speculate that addi- (de)conjugation as well as their distribu-
ubiquitin-interacting motifs (UIMs) in the tional signals (monoubiquitin and chains tion in vivo are still very limited.
DNA double-strand break response pro- with different linkage and length) can Monoclonal antibodies recognizing di-
tein Rap80 are positioned to cross two control the same molecular process with glycine-modified lysines have been used
K63-linked monomers, whereas Ataxin-3 different kinetics and spatial constraints. in combination with mass spectrometry
UIMs display K48 avidity (Sims and It has also been speculated that unan- in efforts to increase the sensitivity of
Cohen, 2009). The proteasome receptor chored ubiquitin chains can regulate immune-based techniques (Xu et al.,
S5a has two UIMs separated by linker NF-kB activation (Xia et al., 2009). 2010) (Figure 1B). These antibodies enrich

678 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


for the C-terminal di-glycine mulate in the cell to facilitate
motif of ubiquitin attached to their detection, including the
the acceptor lysine following use of inhibitors of the pro-
proteolysis of ubiquitinated teasome and of deubiquiti-
proteins by trypsin (Fig- nating enzymes (DUBs). This
ure 1B). This method revealed has often led to the conclu-
more than 200 ubiquitinated sion that high-mobility ubiqui-
proteins from human embry- tin-positive smears observed
onic kidney 293 cells, the on immunoblots represent
majority of which were previ- the natural modification of
ously unknown targets (Xu substrates by very long ubiq-
et al., 2010). This strategy uitin chains. This, however,
can be coupled to stable can be misleading because
isotope labeling with amino the combination of different
acids in cell culture (SILAC) ubiquitin signals (monoubi-
to quantitatively explore pro- quitin or ubiquitin chains) on
tein ubiquitination in diverse the same type of substrate
biological settings. However, can also yield high-mobility
it needs to be noted that smears (Haglund et al.,
this approach can neither 2003; Huang et al., 2006),
detect short-lived proteins and inhibition of DUBs and
nor distinguish lysine modifi- the proteasome may cause
cation by NEDD8. an overrepresentation of
The AQUA (absolute quan- long ubiquitin chains that
tification) method developed might not naturally occur in
in the Gygi laboratory is the cell.
another promising approach The question of chain
to measure the dynamics of length is important given that
ubiquitin signaling in cells chains with different topology
(Kirkpatrick et al., 2005). and length may regulate dif-
AQUA relies on the use of Figure 1. Antibodies for Ubiquitin Signals ferent cellular functions. For
stable isotope-labeled inter- (A) Linkage-specific antibodies, such as a-lysine 11(K11)-, a-K48-, a-K63- instance, the length of K48-
nal standard peptides that linked ubiquitin chains and a-linear ubiquitin chains, can be applied for the linked tetraubiquitin chains
detection of endogenous ubiquitination of a specific linkage type.
mimic those produced during (B) After trypsin digestion of total cell extracts, immunoprecipitation of the is optimized for interaction
tryptic digestion of ubiquiti- samples by a specific antibody against glycine-glycine-lysine peptides with proteasomal receptors
nated proteins of interest. (a-GGK Ab) can enrich fragments with ubiquitinated K residues from both (Pickart and Fushman, 2004),
substrates and ubiquitin chains. Analysis by mass spectrometry enables the
In case of mono- or polyubi- identification of new target proteins as well as sites of ubiquitination.
as a ternary complex can
quitinated proteins, tryptic be formed between the ubiq-
digestion produces a series uitin chains and proteasomal
of unbranched and di-glycine- tandem mass spectrometers, makes receptors Rpn13 and S5a (Zhang et al.,
branched peptides. Initial analysis of AQUA the tool of choice for quantitatively 2009). Moreover, given that trimming
such mixtures allows identification of measuring ubiquitin modifications directly of ubiquitinated substrates occurs from
ubiquitination sites in the substrate and in cell lysates (Kirkpatrick et al., 2006). the distal end of the chains, it seems
the type of ubiquitin chain linkage (such that the length of K48-linked chains
as monoubiquitination or K63- or K48- What Is Known about Ubiquitin dictates the duration of proteasomal
ubiquitin chains). Based on this informa- Chain Length In Vivo? degradation (Lee et al., 2010).
tion, substrate-, site-, and linkage-specific The methods described above are pre- Monoubiquitination can also drive pro-
reference peptides are synthesized and dicted to provide quantitative information teins to proteasomal degradation (Shabek
used as quantitative internal standards, on the repertoire of ubiquitin signals and et al., 2009). These observations collec-
allowing for precise quantification of ubiquitinated proteins generated in dif- tively suggest that the ubiquitin chain
monoubiquitin and polyubiquitin chains ferent biological settings. However, these length required for proteasomal degrada-
by targeted proteomics approaches such methods cannot monitor the length of tion is determined by the substrate’s
as selective reaction monitoring. With ubiquitin chains in vivo. At present, all affinity for the proteasome and must be
this methodology, the stoichiometry of our knowledge on their length in vivo just high enough to allow processivity of
ubiquitin moieties on a protein of interest relies on nonquantitative analysis of the proteolytic process. This kind of
can be determined (Figure 2A). Its immunoblots. Several procedures have adjustment is most likely controlled by
simplicity and sensitivity, coupled with been designed to cause ubiquitin chains a proteasome-associated complex,
the current widespread availability of and polyubiquitinated substrates to accu- which is equipped with both ubiquitin

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 679


ligase (HUL5) and deubiquiti- UBDs. In such a scenario,
nating (UBP6) activities (Cro- short ubiquitin chains added
sas et al., 2006). to substrates will be preferen-
In the case of the NF-kB tially decoded by linkage-
pathway, distinct activation selective UBDs, whereas
steps involve K63, linear, long chains may be promis-
and K48 chains (Bianchi and cuously read by different
Meier, 2009), which are UBDs, possibly placing chain
further edited (in length and length next to chain linkage
topology) by ligases and type in determining ubiquitin-
DUBs (Wertz et al., 2004; UBD selectivity.
Newton et al., 2008). An initial
mechanism proposed for NF- Development of Sensors
kB activation implicated long Using Selective UBDs
K63-linked chains in the In order to measure the
recruitment of TAK1 and IKK dynamics of ubiquitin chain
kinases via their respective formation/disassembly and
adaptor proteins TAB2 and their length in vivo, functional
NEMO (reviewed in Bianchi ubiquitin sensors are needed
and Meier, 2009). This model (Figure 2B). A recently engi-
has been challenged by the neered sensor (TUBE, tan-
demonstration that cells ex- dem repeated ubiquitin enti-
pressing ubiquitin lacking ties) possesses four tandem
K63 have intact NF-kB UBA domains of either HR23
signaling via tumor necrosis or ubiquitin 1 (Hjerpe et al.,
factor-a receptors (Xu et al., 2009). The ubiquitin-binding
2009). Interestingly, based capacity of TUBE is markedly
on available structures it higher for ubiquitin tetramers
appears that chain-selective in comparison to monoubi-
UBDs in TAB2 and NEMO quitin. In addition, the affinity
interact with K63-linked or of the interaction of TUBE
linear diubiquitin chains, with either K63- or K48-tet-
respectively (Kulathu et al., raubiquitin chains is much
2009; Rahighi et al., 2009; greater than that of a single
Sato et al., 2009). Given that UBA domain (Hjerpe et al.,
no data are available on the 2009). An intriguing feature
precise length of ubiquitin of TUBE is its ability to pro-
chains in the NF-kB pathway, tect ubiquitin chains from
it is tempting to speculate cleavage by blocking acces-
that diubiquitin chains are sibility to DUBs.
the fundamental units recog- The design principle of
nized by selective UBDs. TUBE could be easily adap-
However, UBDs also show ted to other UBDs: for ex-
promiscuous binding with ample, a K63 chain-specific
lower affinities to other types sensor could be created by
of chains. Examples include fusing multiple NZF domains
Figure 2. Quantification and Detection of Ubiquitin Chains In Vivo
the NZF domain of TAB2, (A) The workflow for the AQUA (absolute quantification) method of quantitative of TAB2 in tandem, a K48-
which also binds K48 chains mass spectrometry is depicted. First, a representative tryptic peptide is selected specific sensor by merging
in solution (Kulathu et al., based on initial proteomic sequencing experiments and then synthesized with multiple Pru domains of
a stable isotope at one residue for identification. The tryptic peptide sequence
2009), and the UBAN domain for lysine 48 (K48)-linked ubiquitin chains is indicated (upper panel). AQUA peptide Rpn13, and a linear-specific
in NEMO, which interacts standards are added to the sample (cell lysates or immunocomplexes) prior to sensor by arraying several
with K63- and K48-linked trypsin digestion and targeted proteomic analysis is performed using selective copies of the UBAN domain
reaction monitoring. The amount of total protein and the extent of ubiquitination
chains longer than diubiquitin at that particular site can be determined by comparing the precise amounts of
of NEMO or ABINs. These
(Rahighi et al., 2009). We the unmodified and ubiquitinated versions of the peptide (lower panel). UBD-derived ubiquitin sen-
speculate that diubiquitin (B) Schematic models of ubiquitin sensors are shown. By using different ubiq- sors could be used to protect
uitin-binding domains (UBDs), the sensor can be applied for specific linkage
units in longer chains may and purify substrates deco-
type of ubiquitin chains (left), such as K48, K63, and linear chains. Tandem
amplify signals that can be UBDs may be used to determine the chain length (right). One UBD recognizes rated with endogenous ubiq-
recognized by nonselective 1 unit of diubiquitin. The tag chosen depends on the experimental purposes. uitin chains. They could also

680 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


be used to determine the predominant Programme (FP7/2007-2013)/ERC grant agree- Matsumoto, M.L., Wickliffe, K.E., Dong, K.C., Yu,
linkage type within these chains by ment n [250241-LineUb]. C., Bosanac, I., Bustos, D., Phu, L., Kirkpatrick,
D.S., Hymowitz, S.G., Rape, M., et al. (2010).
competition experiments and for
REFERENCES Mol. Cell 39, 477–484.
measuring the length of ubiquitin poly-
Newton, K., Matsumoto, M.L., Wertz, I.E., Kirkpa-
mers in their natural environment.
Bianchi, K., and Meier, P. (2009). Mol. Cell 36, trick, D.S., Lill, J.R., Tan, J., Dugger, D., Gordon,
A further critical challenge will be to 736–742. N., Sidhu, S.S., Fellouse, F.A., et al. (2008). Cell
evaluate chain-specific ubiquitin sensors 134, 668–678.
Bienko, M., Green, C.M., Crosetto, N., Rudolf, F.,
using advanced (high-throughput) single- Zapart, G., Coull, B., Kannouche, P., Wider, G., Pickart, C.M., and Fushman, D. (2004). Curr. Opin.
cell or -molecule microscopy. This might Peter, M., Lehmann, A.R., et al. (2005). Science Chem. Biol. 8, 610–616.
permit the qualitative and quantitative 310, 1821–1824. Pierce, N.W., Kleiger, G., Shan, S.O., and
assessment of ubiquitin chain formation Bremm, A., Freund, S.M., and Komander, D. Deshaies, R.J. (2009). Nature 462, 615–619.
and the interplay between different chain (2010). Nat. Struct. Mol. Biol. 17, 939–947. Rahighi, S., Ikeda, F., Kawasaki, M., Akutsu, M.,
types in vivo. Analyzing additional proper- Crosas, B., Hanna, J., Kirkpatrick, D.S., Zhang, Suzuki, N., Kato, R., Kensche, T., Uejima, T., Bloor,
ties, such as the spatial and temporal D.P., Tone, Y., Hathaway, N.A., Buecker, C., Leg- S., Komander, D., et al. (2009). Cell 136, 1098–
regulation of conjugation and deconjuga- gett, D.S., Schmidt, M., King, R.W., et al. (2006). 1109.
Cell 127, 1401–1413. Raiborg, C., and Stenmark, H. (2009). Nature 458,
tion of ubiquitin chains as well as their
Deshaies, R.J., and Joazeiro, C.A. (2009). Annu. 445–452.
length in vivo, could enable a high-
Rev. Biochem. 78, 399–434. Sato, Y., Yoshikawa, A., Yamashita, M., Yamagata,
resolution, systems-level analysis of the
Dikic, I., Wakatsuki, S., and Walters, K.J. (2009). A., and Fukai, S. (2009). EMBO J. 28, 3903–3909.
‘‘ubiquitinome.’’
Nat. Rev. Mol. Cell Biol. 10, 659–671. Shabek, N., Herman-Bachinsky, Y., and Ciechan-
Haglund, K., Sigismund, S., Polo, S., Szymkiewicz, over, A. (2009). Proc. Natl. Acad. Sci. USA 106,
Perspective I., Di Fiore, P.P., and Dikic, I. (2003). Nat. Cell Biol. 11907–11912.
Even though we have attained a sophisti- 5, 461–466. Sims, J.J., and Cohen, R.E. (2009). Mol. Cell 33,
cated mechanistic understanding of the Hershko, A., and Ciechanover, A. (1998). Annu. 775–783.
ubiquitin system, it has been more difficult Rev. Biochem. 67, 425–479. Tokunaga, F., Sakata, S., Saeki, Y., Satomi, Y., Kir-
to analyze the orchestration of its func- Hicke, L., and Dunn, R. (2003). Annu. Rev. Cell Dev. isako, T., Kamei, K., Nakagawa, T., Kato, M., Mur-
Biol. 19, 141–172. ata, S., Yamaoka, S., et al. (2009). Nat. Cell Biol. 11,
tions in vivo. Within the cellular environ-
Hjerpe, R., Aillet, F., Lopitz-Otsoa, F., Lang, V., 123–132.
ment, ubiquitin signals must select the
England, P., and Rodriguez, M.S. (2009). EMBO Ulrich, H.D., and Walden, H. (2010). Nat. Rev. Mol.
correct binding partner at the right place
Rep. 10, 1250–1258. Cell Biol. 11, 479–489.
and time, ensuring accurate signaling.
Huang, F., Kirkpatrick, D., Jiang, X., Gygi, S., and Virdee, S., Ye, Y., Nguyen, D.P., Komander, D.,
To understand the specificity and Sorkin, A. (2006). Mol. Cell 21, 737–748. and Chin, J.W. (2010). Nat. Chem. Biol. 6, 750–757.
dynamics of the ubiquitin system in its
Husnjak, K., Elsasser, S., Zhang, N., Chen, X., Ran- Wang, H., Matsuzawa, A., Brown, S.A., Zhou, J.,
biological context, it is critical that highly dles, L., Shi, Y., Hofmann, K., Walters, K.J., Finley, Guy, C.S., Tseng, P.H., Forbes, K., Nicholson,
sensitive methods, such as mass spec- D., and Dikic, I. (2008). Nature 453, 481–488. T.P., Sheppard, P.W., Hacker, H., et al. (2008).
trometry and advanced microscopy, are Ikeda, F., and Dikic, I. (2008). EMBO Rep. 9, Proc. Natl. Acad. Sci. USA 105, 20197–20202.
deployed to measure key parameters, 536–542. Wertz, I.E., O’Rourke, K.M., Zhou, H., Eby, M., Ara-
such as the amount of different ubiquitin Iwai, K., and Tokunaga, F. (2009). EMBO Rep. 10, vind, L., Seshagiri, S., Wu, P., Wiesmann, C.,
signals, the kinetics of UBD-ubiquitin 706–713. Baker, R., Boone, D.L., et al. (2004). Nature 430,
recognition, and the type and length of Jin, L., Williamson, A., Banerjee, S., Philipp, I., and 694–699.
ubiquitin chains attached onto substrates Rape, M. (2008). Cell 133, 653–665. Wickliffe, K., Williamson, A., Jin, L., and Rape, M.
in vivo. By shedding light onto these prop- Kirkin, V., McEwan, D.G., Novak, I., and Dikic, I. (2009). Chem. Rev. 109, 1537–1548.

erties, we will gain a deeper under- (2009). Mol. Cell 34, 259–269. Winget, J.M., and Mayor, T. (2010). Mol. Cell 38,
627–635.
standing of one of the most important Kirkpatrick, D.S., Weldon, S.F., Tsaprailis, G., Lie-
and widely used regulatory systems of bler, D.C., and Gandolfi, A.J. (2005). Proteomics Xia, Z.P., Sun, L., Chen, X., Pineda, G., Jiang, X.,
5, 2104–2111. Adhikari, A., Zeng, W., and Chen, Z.J. (2009).
cell physiology.
Kirkpatrick, D.S., Hathaway, N.A., Hanna, J., Nature 461, 114–119.
Elsasser, S., Rush, J., Finley, D., King, R.W., Xu, G., Paige, J.S., and Jaffrey, S.R. (2010). Nat.
ACKNOWLEDGMENTS and Gygi, S.P. (2006). Nat. Cell Biol. 8, 700–710. Biotechnol. 28, 868–873.
Kulathu, Y., Akutsu, M., Bremm, A., Hofmann, K., Xu, M., Skaug, B., Zeng, W., and Chen, Z.J. (2009).
We are grateful to C. Behrends, A. Ciechanover, K.
and Komander, D. (2009). Nat. Struct. Mol. Biol. Mol. Cell 36, 302–314.
Rittinger, and S. van Wijk for comments and
16, 1328–1330. Zhang, N., Wang, Q., Ehlinger, A., Randles, L.,
discussions. Research in the I.D. laboratory is sup-
ported by the Deutsche Forschungsgemeinschaft, Lee, M.J., Lee, B.H., Hanna, J., King, R.W., and Lary, J.W., Kang, Y., Haririnia, A., Storaska, A.J.,
the Cluster of Excellence ‘‘Macromolecular Finley, D. (2010). Mol. Cell. Proteomics 2010, 8. Cole, J.L., Fushman, D., et al. (2009). Mol. Cell
Complexes’’ of the Goethe University Frankfurt Lo, Y.C., Lin, S.C., Rospigliosi, C.C., Conze, D.B., 35, 280–290.
(EXC115), and the European Research Council Wu, C.J., Ashwell, J.D., Eliezer, D., and Wu, H. Zhao, S., and Ulrich, H.D. (2010). Proc. Natl. Acad.
under the European Union’s Seventh Framework (2009). Mol. Cell 33, 602–615. Sci. USA 107, 7704–7709.

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 681


Leading Edge
Minireview

Ubiquitin: Same Molecule,


Different Degradation Pathways
Michael J. Clague1,* and Sylvie Urbé1,*
1Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK

*Correspondence: clague@liv.ac.uk (M.J.C.), urbe@liv.ac.uk (S.U.)


DOI 10.1016/j.cell.2010.11.012

Ubiquitin is a common demoninator in the targeting of substrates to all three major protein degra-
dation pathways in mammalian cells: the proteasome, the lysosome, and the autophagosome. The
factors that direct a substrate toward a particular route of degradation likely include ubiquitin chain
length and linkage type, which may favor interaction with particular receptors or confer differential
susceptibility to deubiquitinase activities associated with each pathway.

The dynamic state of bodily proteins was established by a substrate dictates the chain linkage type. The human genome
analyzing the fate of stable isotope-labeled amino acids that encodes more than 20 different types of ubiquitin-binding
had been fed to mice. These classic experiments, conducted domains, and proof of principle for linkage specificity of binding
by Rudolf Schoenheimer in the late 1930s, presage modern has been established (see Essay by F. Ikeda, N. Crosetto, and
stable isotope labeling techniques (such as SILAC), which allow I. Dikic on page 677 of this issue). One means to achieve this is
determination of the turnover rate of hundreds to thousands of through the spatial arrangement of tandem ubiquitin-binding
individual proteins in a single mass spectrometry experiment domains (UBDs) either encoded in a single protein or by
(Kristensen et al., 2008). After its discovery, the lysosomal combining domains within a multimolecular complex, such that
compartment was considered the principal site of degradation simultaneous occupancy of two binding sites is restricted to
of cellular proteins, through the action of resident acid-depen- particular chain configurations.
dent proteases. However, this view perished with the demon-
stration that the half-lives of most cellular proteins are insensitive
Proteasomal Degradation
to alkalinization of the lysosomes. The subsequent discovery of
Early work suggested that proteasomal targeting requires
the ubiquitin-proteasome degradation system as the major route
a lysine 48 (K48)-linked ubiquitin chain consisting of at least
to protein degradation generated a new orthodoxy. Central to
four conjoined ubiquitin molecules. This was based first upon
this model is the idea that covalent modification of substrate
the biochemical analysis of chains formed on a model substrate,
proteins with a polypeptide ubiquitin tag targets them to the large
b-galactosidase, in a reticulocyte lysate system and second
(26S) proteolytic complex known as the proteasome.
upon studies showing that unique among lysine mutant versions
It came then as a surprise to discover that ubiquitin tagging also
of ubiquitin, K48R cannot serve as the sole source of ubiquitin in
provides a signal to route endocytosed receptors to the lyso-
yeast (Finley, 2009; Xu et al., 2009). The affinity of unanchored
somal degradation pathway and more recently to mark organ-
K48 polyubiquitin chains for the proteasome increases more
elles for disposal by the third major cellular degradative pathway
than 100-fold from di- to tetraubiquitin (170 nM) and less
of autophagocytosis. The role of ubiquitin in protein degradation
steeply thereafter (Thrower et al., 2000).
is more ubiquitous than once thought (Figure 1). In this Minire-
A body of work now suggests that in fact the proteasome
view, we consider how a ubiquitin tag selects for specific degra-
happily accepts other ubiquitin chain types. Indirect evidence
dation pathways and also highlight the interplay between these
for this comes from the observation that acute proteasome inhi-
pathways that a shared dependence on ubiquitin engenders.
bition does not lead to the selective accumulation of K48 chains.
Rather, all chain types with the exception of K63 are increased
General Considerations (Jacobson et al., 2009; Xu et al., 2009). During cell division, the
Substrate proteins are selected for modification of lysine resi- human anaphase-promoting complex (APC/C) recruits two E2
dues by ubiquitin through interaction with an E3 ligase protein ligases (UbcH10 and Ube2S), which combine to exclusively
that recruits an E2-enzyme charged with ubiquitin. This can generate K11-linked chains on substrates. Loss of this unit leads
result in transfer of a single ubiquitin molecule (monoubiquitina- to strong defects in mitotic progression due to failure of the
tion) or coupling of further ubiquitin molecules, through integral necessary degradation processes (Song and Rape, 2010).
lysine residues, to form a chain. The seven lysines of ubiquitin In vitro studies have even shown that K63-modified dihydrofo-
provide for the formation of different isopeptide chain linkages, late reductase provides an efficient proteasome substrate
which adopt different three-dimensional structures, and all of (Hofmann and Pickart, 1999).
which are represented in eukaryotic cells (Xu et al., 2009). The The proteasome is composed of a core (20S) particle contain-
specific combination of E2 and E3 enzymes recruited to ing multiple proteolytic sites and a 19S regulatory particle that

682 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


Figure 1. Ubiquitin Is a Common Denominator of Protein Degrada-
tion Pathways
Specific ubiquitin receptors are associated with each degradation pathway.
Autophagosomal and multivesicular body (MVB) pathways merge at the lyso-
some and share a dependence on v-ATPase activity (inhibited by bafilomycin).
Both pathways also share sensitivity to inhibitors of phosphoinositide 3-kinase
activity, such as wortmannin or 3-methyladenine, as the family member
hVPS34 is required both for recruitment of ESCRT (endosomal sorting
complex required for transport) components to MVBs and for expansion of
the double-membrane preautophagosomal structure. Proteasomal inhibitors
include lactacystin and epoxomicin.

governs access to the core. To enter the core, substrates must


be amenable to unfolding by a hexamer of ATPases associated
with the base of the regulatory particle. Other constituents of the
Figure 2. Ubiquitin Recognition by the Major Degradative Pathways
regulatory particle are implicated in the recruitment of substrates Depiction of the ‘‘ubiquitin receptors’’ associated with each degradative
(Finley, 2009). Rpn10 and Rpn13 interact with ubiquitinated pathway. The domain structures shown are for the human representatives of
substrates through UIM (ubiquitin-interacting motif) domains each protein family, except for yeast Ddi1, the human ortholog of which
and a Pru (pleckstrin-like receptor for ubiquitin) domain, respec- does not contain a UBA domain. CB: clathrin-binding motif; CC: coiled coil;
ESCRT: endosomal sorting complex required for transport; GGA: golgi-asso-
tively. The UBL/UBA family of proteins are substoichiometric ciated, gamma adaptin ear containing, ARF-binding protein; GAE: gamma
components of purified proteasomes that bind ubiquitin via their adaptin ear; GAT: GGA and TOM1; GLUE: GRAM-like ubiquitin-binding in
UBA (ubiquitin-associated) domain and the proteasome regula- Eap45; HRS: HGF receptor tyrosine kinase substrate; LIR: LC3-interacting
region; PB1: Phox and Bem1; PRU: Pleckstrin-like receptor for ubiquitin;
tory particle through its UBL (ubiquitin-like) domain. They are SH3: Src homology domain 3; STAM: signal transducing adaptor molecule;
proposed to remotely scavenge ubiquitinated substrates and TOM1: target of myb1; TSG101: tumor susceptibility gene 101; UBA: ubiqui-
present them to the proteasome (Figure 2). Particular protea- tin-associated domain; UBL: ubiquitin-like domain; UEV: ubiquitin E2 variant
domain; UIM: ubiquitin-interacting motif; VHS: Vps27, HRS, and STAM;
some-associated ubiquitin receptors have been linked with the
VPS36: vacuolar protein sorting 36; vWFA: von Willebrand Factor type A;
degradation of specific substrates (reviewed in Finley, 2009). ZZ: zinc finger. Note the following gene names and commonly used alternative
The mammalian regulatory particle has three associated deu- names also apply: p62; SQSTM1 (sequestosome), NDP52; CALCOCO2,
biquitinating enzymes (DUBs), POH1/PSMD14, USP14, and UBQLN1; PLIC1; DSK2. Domain annotation based on PFAM and UNIPROT.
UCH37 (Rpn11 and Ubp6 in budding yeast), which have distinct
specificities for different chain linkages (Finley, 2009). What is the protease activities (that is, USP14 and UCH37) account for all
function of these DUB activities? One important function is to activity directed toward K48-linked chains and also contribute
salvage ubiquitin in order to maintain the cellular ubiquitin pool. to K63-linked chain disassembly (Jacobson et al., 2009). One
The JAMM/MPN+ metalloprotease POH1 is thought to specifi- attractive notion is that the integration of these DUB activities
cally disassemble K63-linked chains, as well as cleave the may provide for a proof-reading mechanism, facilitating release
isopeptide bond that links the substrate and the proximal ubiqui- from the proteasome if commitment to degradation is not
tin, allowing for en bloc removal of an ubiquitin chain. It also accomplished within a given time window. For example, prefer-
governs entry into the central proteolytic chamber, thereby ential proteasomal DUB activity against K63-linked chains has
coupling substrate degradation to recycling of ubiquitin (Yao been proposed to select against these substrates for degrada-
and Cohen, 2002). Ubiquitin-aldehyde-sensitive cysteine tion (Jacobson et al., 2009). Also in line with this principal,

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 683


a specific chemical inhibitor of USP14 has recently been shown that are committed to degradation. Although deubiquitination is
to enhance the rate of protein degradation (Lee et al., 2010). not an obligate step for MVB sorting, proof-reading and ubiquitin
In yeast, a ubiquitin ligase, Hul5 (mammalian ortholog is recycling roles akin to those suggested for proteasomal DUBs
KIAA10/E3a), that is associated with proteasomes can oppose are consistent with available data (Clague and Urbé, 2006).
Ubp6 activity through chain elongation (E4) (Crosas et al.,
2006). Thus a balance between proteasome-associated ubiqui- Autophagy
tin ligase and DUB activity may determine receptor fate. The signature of autophagy is the capture of cytosol and organ-
elles through envelopment within a double-membrane compart-
Endolysosomal Degradation ment derived from the preautophagosomal structure. In
The lysosomal degradation pathway is the principle means by common with the MVB, the autophagosome can then directly
which a cell turns over plasma membrane proteins, such as recep- fuse with late endosomes or lysosomes to form the autolyso-
tors or channels. Its defining characteristic is a requirement for some, wherein the double-membrane structure is digested. It
organelle acidification, mediated by the v-ATPase. Endocytosed is well suited for the digestion of cytosolic entities, which are
proteins are either recycled to the plasma membrane or captured incompatible with unfolding by the proteasome, such as organ-
into lumenal vesicles of the multivesicular body (MVB) as it elles or protein aggregates.
matures from the sorting endosome, before fusing directly with Identification of autophagy (Atg) genes and subsequent
lysosomes. Some receptors use ubiquitin as an internalization biochemical characterization revealed two essential posttransla-
signal, but for other ubiquitinated receptors, such as epidermal tional modification pathways, which resemble ubiquitination.
growth factor receptor, this is secondary to, or redundant with, In one case, Atg12 is stably conjugated to Atg5 in a constitutive
other adaptor-binding motifs. Ubiquitination directs internalized fashion. In the second case, Atg8 is conjugated to the lipid phos-
proteins toward lysosomal degradation by engagement with en- phatidylethanolamine by transfer from an E2 enzyme following the
dosomal sorting complexes required for transport (ESCRTs) (re- onset of autophagy (for example, as induced by amino acid depra-
viewed in Clague and Urbé, 2006). Monoubiquitination, in the vation). This is a prerequisite for the expansion of the preautopha-
form of an irreversible linear fusion appended to various receptors, gosomal structure, perhaps by facilitating fusion between
is a sufficient signal for this sorting step. However, evidence membranes. In mammalian cells, Atg8 is known as LC3 and its lipi-
suggests K63 as the primary ubiquitin chain type involved in endo- dated form as LC3-II. In fact, there are six Atg8 homologs in the
somal sorting. Early studies in yeast cells, which suggested that human genome collectively known as the LC3/GABARAP family.
appendage of K63-linked diubiquitin enhances vacuolar sorting, Whereas autophagy is generally thought of as a nonselective
have been recently elaborated on with a detailed analysis of the degradation process, certain structures and organelles are selec-
downregulation of the Gap1 permease. These studies conclude tively removed by this pathway. For example, mitochondria are
that monoubiquitination is sufficient for initial internalization (at lost during reticulocyte maturation and as a consequence of un-
least so long as it is an irreversible linear fusion) but that efficient coupling (disconnecting the electron transport chain from ATP
sorting at the endosome by the ESCRT machinery requires K63- production) in cultured cells. Ribosomes, peroxisomes, and path-
linked polyubiquitin (Lauwers et al., 2009). Concordantly, studies ophysiological protein aggregates can also be degraded by
of the mammalian TrkA and MHC class I proteins reveal their utili- autophagy. Recent studies have led to the proposal of a common
zation of K63-linked polyubiquitination for routing to the lysosome principle involved in ‘‘selective autophagies’’ and once again ubiq-
(Duncan et al., 2006; Geetha and Wooten, 2008). uitin plays a critical role (Kirkin et al., 2009). In general if the body to
The first point of engagement of ubiquitinated cargo with the be cleared is ubiquitinated, then an adaptor molecule is required
MVB sorting machinery is proposed to be the ESCRT-0 complex, to couple this to the preautophagosomal membrane rich in
comprising HRS and STAM, both of which possess UIM and VHS Atg8/LC3. The prototypical adaptor of this class is p62/sequesto-
(Vps27, HRS, and STAM) domains, which can bind ubiquitin some 1, which contains both a ubiquitin-interacting domain (UBA)
(Figure 2). Intact ESCRT-0 binds 50 times more tightly to K63- and a LIR motif (LC3-interacting region), a domain structure
linked tetraubiquitin than to monoubiquitin, but only 2-fold more shared with Neighbor of BRCA1 gene 1 (NBR1) (Figure 2) (Pankiv
tightly than to K48-tetraubiquitin (Ren and Hurley, 2010). et al., 2007). p62 has been previously implicated in the clearing of
ESCRT-0 is recruited to endosomes through binding to phospha- protein aggregates, which are known to be concentrated in ubiq-
tidylinositol 3-phosphate but also binds to clathrin and the down- uitin. Recent data have indicated an essential role for ubiquitin
stream ESCRT-I complex. An alternative ESCRT-0 complex (K63 and K27 polyubiquitin chain linkages have been implicated)
comprising TOM1, Tollip, and Endofin possesses all these salient in the selective autophagy of depolarized mitochondria, which
features of the HRS-STAM complex. It is currently unclear become ubiquitinated following recruitment of the E3 ubiquitin
whether these two complexes are redundant or used to receive ligase Parkin (Geisler et al., 2010). Using a lysine-less mutant of
different cargoes. In a further striking parallel to the proteasomal ubiquitin fused with red fluorescent protein, Kim et al. established
system, the ESCRT machinery has known associations with at that irreversible monoubiquitination is sufficient to concentrate
least two DUB activities, AMSH and USP8 (UBPY), drawn from a soluble protein within autophagosomal structures in a p62-
the JAMM/MPN+ and USP families, respectively. In yeast, the dependent manner (Kim et al., 2008).
dominant endocytic E3 ligase activity Rsp5 can also associate A selective pathway requiring the Ubp3:Bre5 DUB complex in
with the STAM ortholog Hse1, providing a counterbalance to Saccharomyces cerevisiae operates in the removal of mature ribo-
Ubp2 and Ubp7 (Ren et al., 2007), while a third ESCRT-associ- somes (Kraft and Peter, 2008). In cells deficient in Ubp3, ribosomal
ated DUB Doa4 is required for ubiquitin recycling of receptors fractions are enriched with ubiquitin. Although an intimate

684 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


connection has been established, the exact role of ubiquitin in ri- substrate targeted to a particular pathway? Variable parameters
bophagy is unclear. One model posits that ubiquitin may be pro- include location, chain length, and linkage type. A clear bias of
tecting ribosomes from autophagy, which is then promoted by the endosomal pathway toward K63-linked chains has emerged.
Ubp3 activity. Alternatively, a dynamic modification with ubiquitin This may simply reflect the subcellular localization of specific
may be required, perhaps as an engulfment signal similar to that of E3 ligases in combination with a high local concentration of ubiq-
mitochondria. In support of this notion, a temperature-sensitive uitin-binding proteins, which couple to the ESCRT-machinery
defect in the E3 ligase Rsp5 shows a synthetic ribophagy defect rather than the proteasome. New techniques allow for the deter-
with loss of Ubp3 as compared with cells lacking Ubp3 alone (Kraft mination of individual protein turnover on a global scale (Kristen-
and Peter, 2008). If correct, then the principle of ensuring ubiquitin sen et al., 2008). This will enable the generation of a comprehen-
homeostasis through deubiquitination may be conserved by each sive annotation of turnover rates as a function of experimental
of the selective degradation pathways we have discussed. perturbations or disease states, opening the door to
a systems-level understanding of protein degradation.
The Interdependence of Degradation Pathways
The relative contribution of degradation pathways may vary ACKNOWLEDGMENTS
greatly between cell types. In most cases of cells cultured under
S.U. is a Cancer Research UK Senior Research Fellow.
stress-free conditions, proteasomal degradation predominates,
but in muscle cells, lysosomal pathways (principally autophagy) REFERENCES
can account for 40% of degradation of long-lived proteins. In
atrophying muscle cells, both pathways are proposed to be Carter, S., Urbé, S., and Clague, M.J. (2004). J. Biol. Chem. 279, 52835–52839.
co-ordinately upregulated under the transcriptional control of Clague, M.J., and Urbé, S. (2006). Trends Cell Biol. 16, 551–559.
FOXO3 (Zhao et al., 2007). However, the proteasome is itself Crosas, B., Hanna, J., Kirkpatrick, D.S., Zhang, D.P., Tone, Y., Hathaway, N.A.,
degraded by starvation-induced bulk autophagy (Kristensen Buecker, C., Leggett, D.S., Schmidt, M., King, R.W., et al. (2006). Cell 127,
et al., 2008). 1401–1413.
The reliance of three major cellular degradation pathways Duncan, L.M., Piper, S., Dodd, R.B., Saville, M.K., Sanderson, C.M., Luzio,
upon ubiquitination suggests that specific inhibition of any one J.P., and Lehner, P.J. (2006). EMBO J. 25, 1635–1645.
pathway may perturb the ubiquitin economy of the cell and Finley, D. (2009). Annu. Rev. Biochem. 78, 477–513.
hence indirectly affect other degradation events (Figure 1). Geetha, T., and Wooten, M.W. (2008). Traffic 9, 1146–1156.
A clear example of this is the activated Met receptor, for which Geisler, S., Holmström, K.M., Skujat, D., Fiesel, F.C., Rothfuss, O.C., Kahle,
its lysosomal degradation is exquisitely sensitive to the depletion P.J., and Springer, W. (2010). Nat. Cell Biol. 12, 119–131.
in free ubiquitin caused by proteasomal inhibition (Carter et al., Gorbea, C., Pratt, G., Ustrell, V., Bell, R., Sahasrabudhe, S., Hughes, R.E., and
2004). Proteasome inhibition may also induce autophagy as Rechsteiner, M. (2010). J. Biol. Chem. 285, 31616–31633.

a compensatory response. The autophagy adaptor protein p62 Hofmann, R.M., and Pickart, C.M. (1999). Cell 96, 645–653.
has also been implicated in proteasomal degradation, whereas Jacobson, A.D., Zhang, N.Y., Xu, P., Han, K.J., Noone, S., Peng, J., and Liu,
C.W. (2009). J. Biol. Chem. 284, 35485–35494.
the E3 ligase Parkin generates an autophagy tag on mitochon-
dria but elsewhere can target proteins to the proteasome. Kim, P.K., Hailey, D.W., Mullen, R.T., and Lippincott-Schwartz, J. (2008). Proc.
Natl. Acad. Sci. USA 105, 20567–20574.
VCP/p97 co-ordinates a number of ubiquitin-dependent
Kirkin, V., McEwan, D.G., Novak, I., and Dikic, I. (2009). Mol. Cell 34, 259–269.
processes that include the proteasome-dependent ERAD (endo-
Kraft, C., and Peter, M. (2008). Autophagy 4, 838–840.
plasmic reticulum-associated degradation) pathway but inter-
estingly has recently been identified as a necessary factor for Kristensen, A.R., Schandorff, S., Høyer-Hansen, M., Nielsen, M.O., Jäättelä,
M., Dengjel, J., and Andersen, J.S. (2008). Mol. Cell. Proteomics 7, 2419–2428.
autophagosome maturation under basal conditions and
Lauwers, E., Jacob, C., and André, B. (2009). J. Cell Biol. 185, 493–502.
following proteasome inhibition (Tresse et al., 2010).
Lee, B.H., Lee, M.J., Park, S., Oh, D.C., Elsasser, S., Chen, P.C., Gartner, C.,
The MVB and autophagy pathways merge at the late endo-
Dimova, N., Hanna, J., Gygi, S.P., et al. (2010). Nature 467, 179–184.
some/lysosome and are both sensitive to proton pump and
Pankiv, S., Clausen, T.H., Lamark, T., Brech, A., Bruun, J.A., Outzen, H., Øver-
phosphoinositide 3-kinase inhibitors. Autophagosome formation
vatn, A., Bjørkøy, G., and Johansen, T. (2007). J. Biol. Chem. 282, 24131–
is inherently sensitive to perturbations earlier in the endocytic 24145.
pathway, which change the character of later endosomal Ren, J., Kee, Y., Huibregtse, J.M., and Piper, R.C. (2007). Mol. Biol. Cell 18,
compartments (such as the composition of SNARE proteins). 324–335.
Occasionally, teleological distinctions between these systems Ren, X., and Hurley, J.H. (2010). EMBO J. 29, 1045–1054.
blur, such that some ubiquitinated cytosolic proteins may be Song, L., and Rape, M. (2010). Mol. Cell 38, 369–382.
degraded in the lysosome and cytoplasm-exposed domains of Thrower, J.S., Hoffman, L., Rechsteiner, M., and Pickart, C.M. (2000). EMBO J.
receptors may be nibbled by the proteasome. Mounting 19, 94–102.
evidence suggests that there is a proteasome pool associated Tresse, E., Salomons, F.A., Vesa, J., Bott, L.C., Kimonis, V., Yao, T.P.,
with endosomes that influences receptor sorting (Geetha and Dantuma, N.P., and Taylor, J.P. (2010). Autophagy 6, 217–227.
Wooten, 2008; Gorbea et al., 2010). Xu, P., Duong, D.M., Seyfried, N.T., Cheng, D., Xie, Y., Robert, J., Rush, J.,
Hochstrasser, M., Finley, D., and Peng, J. (2009). Cell 137, 133–145.
Concluding Remarks Yao, T., and Cohen, R.E. (2002). Nature 419, 403–407.
Ubiquitin tagging is common to the three major cellular pathways Zhao, J., Brault, J.J., Schild, A., Cao, P., Sandri, M., Schiaffino, S., Lecker,
for protein degradation. Herein lies a conundrum: how is a given S.H., and Goldberg, A.L. (2007). Cell Metab. 6, 472–483.

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 685


Leading Edge
Perspective

Will the Ubiquitin System Furnish as


Many Drug Targets as Protein Kinases?
Philip Cohen1,2,* and Marianna Tcherpakov3
1MRC Protein Phosphorylation Unit
2Scottish Institute for Cell Signalling
Sir James Black Centre, Dow Street, Dundee DD1 5EH, Scotland, UK
3BCC Research, 40 Washington Street, Suite 110, Wellesley, MA 02481, USA

*Correspondence: p.cohen@dundee.ac.uk
DOI 10.1016/j.cell.2010.11.016

Protein phosphorylation and protein ubiquitination regulate most aspects of cell life, and defects in
these control mechanisms cause cancer and many other diseases. In the past decade, protein
kinases have become one of the most important classes of drug targets for the pharmaceutical
industry. In contrast, drug discovery programs that target components of the ubiquitin system
have lagged behind. In this Perspective, we discuss the reasons for the delay in this pipeline, the
drugs targeting the ubiquitin system that have been developed, and new approaches that may
popularize this area of drug discovery in the future.

Protein Phosphorylation Drug Discovery undergoing Phase III clinical trials include Pfizer’s JAK3 inhibitor
It can take years, even decades, before a field of research rea- for rheumatoid arthritis (CP-690550) and Incyte Pharmaceuti-
ches the stage of maturity at which its discoveries can obviously cal’s JAK1/JAK2 inhibitor (INCB18424) for treating inflammatory
be exploited for the improvement of health. An excellent example diseases. If these drugs are approved, it will likely spark a new
of this paradigm is the regulation of protein function by reversible wave of interest in developing kinase inhibitors for the treatment
phosphorylation. Phosphorylation was identified in the mid of diseases other than cancer.
1950s as a mechanism for controlling glycogenolysis. Twenty- Even by the late 1970s and early 1980s researchers had shown
five years later, it was still largely thought of simply as a control that oncogenes, such as Src (sarcoma), are protein kinases;
switch for metabolism. Indeed, researchers finally realized that phorbol esters, which promote tumors, are kinase activators;
protein phosphorylation regulates most aspects of cell life only and, growth factor receptors, which have kinase domains, are
after many advances made throughout the 1980s and early overexpressed or mutated in human cancer (reviewed in Cohen,
1990s (Cohen, 2002a). 2002b). So why did it take so long for most pharmaceutical
Surprisingly, the idea that it would be possible to treat diseases companies to capitalize on the therapeutic potential of kinase
with drugs targeting protein kinases was even slower to take inhibitors? In retrospect, one realizes that many researchers
root. Indeed, as late as 1998, the Head of Research and Develop- believed that kinase inhibitors were bad drug targets because
ment at one major pharmaceutical company (which no longer they thought that it would be difficult to achieve the requisite
exists) told one of the authors that ‘‘there was absolutely no specificity and potency. Most protein kinase inhibitors target
future in kinase drug discovery.’’ Later that same year, the ATP-binding pockets of these enzymes, and the structural
researchers revealed the remarkable clinical efficacy of a tyrosine similarities of this site among many different kinases raised the
kinase inhibitor, called Gleevec, for treating chronic myeloge- suspicion that it would be impossible to develop drugs that in-
nous leukemia. Quite quickly, protein kinases then became one hibited only one type of protein kinase. Furthermore, the concen-
of the most popular classes of drug targets for the pharmaceu- tration of ATP in the cell is extremely high (i.e., millimolar), leading
tical industry, especially in the field of cancer treatment. researchers to doubt whether compounds could be developed
Over the past decade, 16 drugs targeting one or more protein with the potency needed to compete successfully with intracel-
kinases have been approved for clinical use in cancer, 12 taken lular ATP. These were, and indeed still are, challenging problems
orally as pills and 4 that are injected. As of 2009, 153 other for many developing kinase inhibitors, but they have proven to be
protein kinase inhibitors were undergoing clinical trials, and 23 quite surmountable.
of these drugs were in the most advanced stage of development, Indeed, considerable potency and specificity have been
termed Phase III (Table 1) (Lawler, 2009). The current global achieved by developing compounds that target not only the
market for kinase therapies is about US$15 billion per annum, ATP-binding site but also small hydrophobic pockets located
and this value is forecasted to double by 2020. Research on proximal to the ATP-binding site. Moreover, researchers are
protein kinases currently accounts for 30% of the drug identifying an increasing number of ‘‘allosteric’’ inhibitors that
discovery programs in the pharmaceutical industry and over bind to other regions of a kinase. These compounds induce
50% of cancer research and development. The kinase inhibitors conformational changes in the kinase, which either suppress

686 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


Table 1. Phosphorylation, Ubiquitination, and Drug Discovery
Phosphorylation Ubiquitination
First publication 1955a First publication 1978b
>500 protein kinases c
10 E1sf, 40 E2sf, >600 E3 ligasesf
140 protein phosphatasesc 90 deubiquitinasesc
Nobel Prize awarded 1992d Nobel Prize awarded 2004e
First drug approved in 2001 (Gleevec) First drug approved in 2003 (Bortezomib)
16 drugs approved, over 150 undergoing clinical trials One drug approved, 16 undergoing clinical trials
Current sales US$15 billion per year Current sales US$1.4 billion per year
30% of pharmaceutical research and development <1% of pharmaceutical research and development
a
Fischer and Krebs, 1955.
b
Ciechanover et al., 1978.
c
Encoded by the human genome.
d
Nobel Prize for Physiology or Medicine awarded to Edmond Fischer and Edwin Krebs.
e
Nobel Prize for Chemistry awarded to Aaron Ciechanover, Avram Hershko, and Irwin Rose.
f
Includes the E1s and E2s for ubiquitin-related modifiers such as Nedd8, SUMO, FAT10, and ISG15.

the enzyme’s activity directly or block its activation by another the E2 interacts with an E3 ligase, and the ubiquitin is then trans-
kinase in the same signaling cascade. ferred from the E2 enzyme to substrates, which also interact with
Furthermore, far from being a disadvantage, lack of specificity the E3 ligase. This last step can occur directly, as in the RING E3
can actually be an advantage. For example, Gleevec was devel- ligases, or it can occur indirectly with the ubiquitin first trans-
oped as an Abelson kinase inhibitor for the treatment of a specific ferred to a cysteine residue on the E3 ligase before being linked
type of leukemia. However, it is also an effective treatment for to the substrate, as in the HECT family of E3 ligases. Chains of
gastrointestinal stromal cancers because it inhibits the c-Kit ubiquitin are created by the same enzymatic process.
receptor and the platelet-derived growth factor (PDGF) receptor Similar to phosphorylation, ubiquitin can be linked covalently
tyrosine kinases, which are overexpressed or mutated in gastro- to only one or several amino acid residues on the same protein
intestinal cancers (Demetri et al., 2006). In addition, the efficacy (Figure 1). However, in contrast to protein phosphorylation, ubiq-
of several anticancer drugs depends on their combined inhibition uitin can also form polyubiquitin chains. Ubiquitin has seven
of several different kinases, and these drugs may be less prone lysine residues and an a-amino group; thus eight different types
to the development of drug resistance than ones that act on only of polyubiquitin chains can form (and probably more because
one specific kinase. Thus, some of the original prejudices against chains with ‘‘mixed’’ linkages are also present in cells).
protein kinases as drug targets have subsequently turned out to Even greater versatility is provided by ubiquitin-like proteins,
have little substance. such as Nedd8, SUMO (1, 2, and 3), FAT10, and ISG15, which
The beauty of targeting protein kinases for therapeutics and are also attached covalently to proteins in processes called ned-
the basis for their popularity is that the same technologies and dylation, SUMOylation, tenylation, and ISGylation, respectively.
small-molecule libraries can be used to develop inhibitors of The formation of polyubiquitin chains and the existence of these
many types of protein kinases in almost every therapeutic ‘‘ubiquitin-like modifiers’’ make the ubiquitin system a more
area. However, the vast amount of medicinal chemistry that complex and potentially more versatile control mechanism
has been carried out in recent years has meant that novel patent than phosphorylation.
space is becoming quite difficult to find. Plus, there is a growing, Like phosphorylation, ubiquitination is reversible. Isopepti-
but probably unfounded, concern that the most important drug dases, called deubiquitinases or DUBs, catalyze the cleavage
targets in this area have been fully exploited. Therefore, the phar- of the ubiquitin from proteins or ‘‘deubiquitination’’ (Figure 1).
maceutical industry has begun to wonder where they may find Interestingly, the number of deubiquitinases is comparable to
the next large set of drug targets that can be tackled in a manner the number of protein phosphatases, but taken together, the
analogous to protein kinases. In this Perspective, we discuss the number of E1-activating enzymes, E2-conjugating enzymes,
premise that components of the ubiquitin system are prime and E3 ligases encoded by the human genome exceeds the
candidates for these new targets. number of protein kinases.

Ubiquitination More Versatile than Phosphorylation? Ubiquitination and Phosphorylation: Analogous Control
Ubiquitination is the covalent attachment of a small protein, Mechanisms
ubiquitin (8.5 kDa), to other proteins. In the first step, a thioester For many years, the sole function of the ubiquitin system was
bond is formed between the C-terminal carboxylate group of thought to be the regulation of protein turnover inside the cell. At-
ubiquitin and the thiol or sulfhydryl group of a cysteine residue taching a chain of ubiquitins linked at lysine 48 (K48-linked poly-
on an E1-activating enzyme. Next, the ubiquitin is transferred ubiquitination) to a protein directs it to the 26S proteasome for
to a cysteine on an E2-conjugating enzyme. In the third step, destruction, and indeed, this is one of the key functions of the

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 687


Figure 1. Phosphorylation and Ubiquitina-
tion Regulate Most Aspects of Cell Life
Phosphorylation involves the covalent attachment
of phosphate to proteins, mainly to serine, threo-
nine, and tyrosine residues. Phosphorylation is
catalyzed by protein kinases and reversed by
protein phosphatases. Protein ubiquitination
involves the covalent attachment of ubiquitin,
a small protein with 76 amino acids, to other
proteins, predominantly to lysine residues. This
reaction is mediated by an E1-activating enzyme,
an E2-conjugating enzyme, and an E3 ligase; this
reaction is reversed by deubiquitinases.

ubiquitin system. However, other types of ubiquitination play K63-linked polyubiquitin chains attached to histone 2A and
distinct roles in the cell and regulate diverse areas of biology, histone 2AX by the E3 ligase RNF8 and the E2 -conjugating
as discussed in another article in this issue (Ikeda et al., 2010). enzyme Ubc13 (Kolas et al., 2007) recruit and assemble factors
For example, K63-linked polyubiquitination (Bhoj and Chen, that are essential for DNA repair, such as BRCA1 (breast cancer
2009; Zeng et al., 2010) and linear polyubiquitin chains 1), RAP80, and other proteins (Bennett and Harper, 2008).
(Tokunaga et al., 2009) regulate innate immunity; K11-linked pol- It is important to emphasize that protein phosphorylation and
yubiquitin chains, which are formed by the anaphase-promoting protein ubiquitination are not distinct and separate control mech-
complex (APC/C) and the E2-conjugating enzyme UbcH10, anisms because the interplay between them is critical for the
are critical for the regulation of mitosis (Garnett et al., 2009; regulation of many cellular processes. For example, phosphoryla-
Jin et al., 2008); and K29/33-linked polyubiquitination inhibits tion regulates a number of E3 ubiquitin ligases and deubiquiti-
certain members of a protein kinase subfamily (Al-Hakim et al., nases. Further, the E3 ligase Skp1-Cullin-F box (SCF) and some
2008). other E3 ligases contain an additional component bTRCP
Like phosphorylation, ubiquitination can also induce confor- (b-transducin repeat-containing protein), which recognizes partic-
mational changes that alter biological function. For example, ular phosphorylated sequence motifs that direct the SCFbTRCP
the response to the proinflammatory cytokine interleukin-1 (IL- complex to ubiquitinate these substrates. Finally, a number of
1) generates K63-linked polyubiquitin chains that interact with kinases can be activated or inhibited by interactions with polyubi-
a component of the TAK1 complex, inducing a conformational quitin chains or by polyubiquitination. Given the omnipresence of
change that allows this protein kinase to autoactivate (Xia protein phosphorylation and ubiquitination inside the cell, under-
et al., 2009). Similarly, monoubiquitination of the deubiquitinase standing the interplay between these two systems is likely to
Ataxin 3 (Todi et al., 2009) and dihydrofolate reductase (Maguire become increasingly more important over the next decade.
et al., 2008) enhances and suppresses their enzymatic activities,
respectively. In contrast, monoubiquitination of the tumor Developing Drugs that Target the Ubiquitin System
suppressor p53 induces a conformational change that exposes The Proteasome Inhibitor Bortezomib
a nuclear export signal. This leads to the translocation of p53 The protease inhibitor Bortezomib, originally called PS341 and
to the cytosol where it may promote apoptotic events (Carter then Velcade (Adams, 2002), was the first drug that targets
et al., 2007). Neddylation of the Cullin RING E3 ligases (CRLs) a component of the ubiquitin system to be approved for clinical
also induces conformational changes that bring the E2 active use in the United States. Developed by ProScript Inc in 1995,
site adjacent to the substrate, permitting the efficient ubiquitina- Bortezomib entered clinical trials in 1997 and was approved by
tion of the substrate by CRLs (Saha and Deshaies, 2008). the Federal Drug Administration in 2003. In 1999 ProScript was
Like phosphorylation, many effects of ubiquitination are medi- acquired by Leukosite, which in turn was acquired by Millenium
ated by interactions with ubiquitin-binding proteins. Different Pharmaceuticals later that same year. Bortezomib has been
polyubiquitin chains adopt distinct three-dimensional structures quite successful, with worldwide sales in 2009 of US$1.4 billion,
and hence interact with different polyubiquitin-binding proteins and this achievement led Takeda to acquire Millenium in 2008.
to regulate distinct processes. For example, proteins tagged Bortezomib was approved as a front-line treatment for B cell
with K48-linked polyubiquitin chains are targeted for destruction lymphoma found primarily in the bone marrow. It is also used
because these ubiquitin chains bind to particular components of for the treatment of mantle cell lymphoma in patients who have
the 26S proteasome. More than 20 different families of polyubi- already received other treatments. It is in Phase III clinical trials
quitin-binding proteins have been identified, and this area has for follicular non-Hodgkin’s lymphoma, Phase II trials for diffuse
become a large topic of research in its own right. large B cell lymphoma, and a great many other clinical trials (re-
Interactions through ubiquitin are also critical for DNA-damage viewed in Tcherpakov, 2010).
signaling and for certain DNA-repair pathways. For example, the Bortezomib, which is given by intravenous injection, has
monoubiquitinated form of FANCD2, a component of the Fanconi remarkable efficacy against multiple myeloma, but the molecular
Anemia Complex, interacts with the UBZ domain of the DNA mechanism underlying its effect is still unclear. Nevertheless, the
nuclease FAN1, and this interaction through ubiquitin is essential multiple myeloma cells that are particularly sensitive to protea-
for repair of DNA interstrand crosslinks (MacKay et al., 2010). some inhibitors express lower levels of proteasome particles

688 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


Table 2. Proteasome Inhibitors Approved or in Clinical Trials
Company Inhibitor Development Stage Disease
Millenium/Takeda Bortezomib/Velcade Approved Multiple myeloma and mantle cell lymphoma
Millenium/Takeda MLN9708 Phase I Multiple myeloma and other cancers
ONYX (Proteolix) Carfilzomib/PR171 Phase III Multiple myeloma and other cancers
ONYX (Proteolix) Onx 0912/PR047 Phase I Multiple myeloma and other cancers
Cephalon CEP18770 Phase I Multiple myeloma and other cancers
Nereus Pharmaceuticals Salinosporamid A/NPI0052 Phase I Multiple myeloma and leukemia

and have a higher proteasome workload than multiple myeloma bring the E2 active site adjacent to the lysine residue of its protein
cells that are relatively resistant to these drugs. Thus, the balance target substrates (Duda et al., 2008; Saha and Deshaies, 2008).
between proteasome workload and degradative capacity may be Millenium/Takeda has developed a relatively specific inhibitor
an important determinant of the sensitivity of a cancer cell to of the NAE-E1 enzyme (Table 3). This compound, MLN4924,
Bortezomib and other proteasome inhibitors (Bianchi et al., showed promise in mouse models of cancer and has entered
2009). Phase I clinical trials for the treatment of multiple myeloma and
A dipeptidyl boronic acid, Bortezomib binds noncovalently to non-Hodgkin’s lymphoma. MLN4924 seems to exert its effect
the 20S proteasome and primarily inhibits its chymotrypsin-like on these cancers by deregulating DNA synthesis during the S
activity (Kisselev et al., 2006). Its success has led to considerable phase of the cell division cycle. MLN4924 appears to stabilize
interest in developing improved ‘‘second generation’’ inhibitors, Cdt1, a DNA replication licensing factor normally ubiquitinated
and Millenium/Takeda has another proteasome inhibitor, by a Cullin RING E3 ligase and then degraded by the proteasome
MLN9708, which can be taken orally, in Phase 1 clinical trials. (Soucy et al., 2009).
Onyx Pharmaceuticals also has several orally active proteasome Inhibitors of Deubiquitinases
inhibitors in clinical trials, which they obtained through the acqui- Deubiquitinases comprise five separate gene families. Four
sition of Proteolix. These inhibitors include Carfilzomib, which families are cysteine proteinases (the USP, OTU, UCH, and
has recently entered Phase III trials according to the website MJD deubiquitinases), and the other one consists of metallo-
http://clinicaltrials.gov. Other proteasome inhibitors that are proteinases (the JAMM/MPN domain family). The E3 ligase
currently undergoing clinical development are listed in Table 2. HDM2 targets the tumor suppressor p53 for degradation. One
An Inhibitor of the E1 Enzyme for Neddylation of the cysteine protease deubiquitinases, USP7 (ubiquitin-spe-
The Nedd8 protein shares 60% sequence identity with ubiqui- cific protease 7), deubiquitinates HDM2, leading to increased
tin, and it is conjugated to its target proteins in a similar manner levels of HDM2 and decreased levels of p53. Therefore, two
to ubiquitin, with a specific E1-activating enzyme (NAE-E1) and companies, Progenra and Hybrigenics, have developed inhibi-
the E2-conjugating enzymes Ube2M and/or Ube2F. The primary tors of USP7 (i.e., P5091 and HBX 41108, respectively) (Colland
target for neddylation appears to be the Cullin components of et al., 2009), with the hope of promoting the proteasomal degra-
Cullin RING E3 ubiquitin ligases. The Cullin RING ligases are dation of HDM2 by enhancing its polyubiquitination. Reduced
the largest family of E3 ligases in the human genome with more expression of HDM2 would then be expected to increase the
than 100 members (Rabut and Peter, 2008). Neddylation permits level of p53.
efficient ubiquitination by Cullin RING ligases; neddylation Progenra is also developing inhibitors targeting USP20, and
induces a conformational change in the Cullin component to they are showing interest in agents for USP2a, USP33, and

Table 3. Inhibitors of E1-Activating Enzymes and E3 Ubiquitin Ligases Undergoing Clinical Trials
Company Inhibitor Target Stage Disease
b
Millenium/Takeda MLN4924 NAE-E1 Phase II Multiple myeloma and Hodgkin’s lymphoma
Roche Nutlin/R7112 E3-Hdm2 Phase I Blood cancers
and solid tumors
Johnson & Johnson JNJ26854165 E3-Hdm2 Phase I Multiple myeloma and solid tumors
Genentech/Roche GDC-0152 E3-IAP Phase I Metastatic malignancies
Novartis LCL161 E3-IAP Phase I Solid tumors
Ascenta Therapeutics AT-406 E3-IAP Phase I Solid tumors and lymphoma
a
Aegera Therapeutics AEG 35156 E3-IAP Phase II AML and liver cancer
Aegera Therapeutics AEG 40826 E3-IAP Phase I Lymphoid tumors
Tetralogics Pharma TL 32711 E3-IAP Phase I Solid tumors and lymphoma
Astellas Pharma YM155 E3-IAP Phase II Lung cancer
a
Antisense oligonucleotide.
b
The E1-activating enzyme for neddylation.

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 689


AMSH (associated molecule with the SH3 domain of STAM) muscle wasting. Nevertheless, several programs have made
(http://www.progenra.com/scientist.html, 2009). USP20, also good progress and a number of E3 ligase inhibitors have
called VDU2 (von Hippel-Lindau deubiquitinating enzyme 2), advanced to clinical trials (Table 3) (reviewed in Tcherpakov,
deubiquitinates and stabilizes hypoxia-inducible factor 1a (HIF- 2010). Moreover, several recent and unexpected developments
1a) (Li et al., 2005). HIF-1a is expressed at high levels in many in this area are likely to enhance future pharmaceutical interest in
human cancers because it is stabilized at the low concentration developing E3 ligase inhibitors.
of dissolved oxygen inside the tumor by high cytokine levels and Several companies have discovered compounds that disrupt
by specific genetic alterations. For example, in von Hippel-Lin- the interaction of the E3 ligase HDM2 and its substrate, the tumor
dau disease, in which individuals develop a variety of tumors, suppressor p53, with the aim of elevating p53 expression. One
mutations in the VHL gene compromise the ubiquitination and such compound, Nutlin 3/R7112, has entered clinical trials
degradation of HIF-1a, leading to the accumulation and overex- (Table 3). A second class of E3 ligases actively targeted by
pression of HIF-1a and its target genes. Therefore, inhibitors of a number of companies is the Inhibitors of Apoptosis Proteins
USP20 (VDU2) and/or USP33 (VDU1) may reduce levels of HIF- (IAPs), and seven antagonists of IAPs have even entered clinical
1a by enhancing its polyubiquitination. trials (Table 3). These drugs are small-molecule mimetics of
Novartis has patented compounds that inhibit the deubiquiti- Smac (also known as Diablo), a protein that antagonizes IAPs
nases USP2 and UCH-L3 (ubiquitin C-terminal hydrolase). by interacting with their BIR domains. Smac mimetics appear
USP2 is another deubiquitinase reported to target MDM2, the to induce the autoubiquitination and degradation of the IAPs,
mouse ortholog of HDM2 (Stevenson et al., 2007), whereas which then leads to the death of cancer cells by stimulating the
UCH-L3 probably plays a role in neurodegenerative disorders, TNF-a pathway (Wu et al., 2007). Destruction of IAPs through
such as Parkinson’s disease. Recently, researchers identified the Smac mimetics also suppresses the production of proinflam-
a small-molecule inhibitor of USP14, called IU1, which did not matory cytokines by Toll-like receptor agonists, suggesting that
inhibit eight other deubiquitylases tested, demonstrating the these drugs may be worth exploring as possible treatments for
feasibility of developing relatively specific inhibitors of these chronic inflammatory diseases (Tseng et al., 2010).
enzymes (Lee et al., 2010). USP14 is associated with the protea- Recently, Ito et al. (2010) surprisingly discovered that the drug
some, and treating cells with IU1 enhanced the degradation of thalidomide binds to cereblon (CRBN), a component of the Cullin
several proteasomal substrates that have been implicated in RING E3 ligase that is important for limb outgrowth and the
neurodegenerative diseases, such as Tau. Drugs that target expression of a fibroblast growth factor (FGF8) during embryonic
USP14 could, therefore, have a potential use in reducing or elim- development (Ito et al., 2010). This finding explained why thalid-
inating misfolded and aggregated proteins that accumulate in omide, originally prescribed as a sedative, caused multiple birth
neurodegenerative and other diseases. defects in pregnant women. Thalidomide is still used for the
Developing pharmaceutical agents that target deubiquitinases treatment of numerous conditions, including leprosy, skin sores,
is still in its infancy, and to our knowledge, no deubiquitinase and myelofibrosis. Therefore, pinpointing the molecular mecha-
inhibitor has yet entered clinical trials. However, as this field nism of the drug’s devastating side effects may facilitate the
progresses, it is clearly going to be essential to assess the development of new thalidomide derivatives that are free from
specificities of these inhibitors. Therefore, assembling compre- this problem.
hensive panels of deubiquitinases for testing specificity will be Arsenic is another drug that unexpectedly regulates an E3
critical, similar to how large panels of protein kinases have ligase. Arsenic is an effective and specific treatment for acute pro-
been of immense value in assessing the selectivity of kinase myelocytic leukemia. In this cancer, the promyelocytic leukemia
inhibitors. (PML) protein becomes fused to the retinoic acid receptor (RAR).
As with kinases, there are certainly going to be deubiquitinases Arsenic triggers the degradation of the PML-RAR fusion protein
for which inhibition needs to be avoided. For example, mutating by inducing the SUMOylation of PML. This modified version of
or deleting the A20 deubiquitinase causes or predisposes indi- PML recruits the SUMO-binding E3 ubiquitin ligase RNF4, which
viduals to inflammatory and autoimmune diseases (Musone catalyzes the polyubiquitination (K48-linked) and proteasomal
et al., 2008; Turer et al., 2008). Similarly, inactivating mutations degradation of the PML-RAR complex (Tatham et al., 2008).
in the deubiquitinase CYLD cause cylindromatosis, a type of Small-molecule inhibitors of several Cullin RING E3 ligases
skin cancer (Kovalenko et al., 2003; Trompouki et al., 2003). have also been identified. SCFskp2 is a Cullin RING E3 ligase
Targeting E3 Ubiquitin Ligases that is highly expressed in some human cancers. Decreased
The human genome encodes more E3 ubiquitin ligases than levels of p27kip1 are a poor prognosis factor in many malignan-
protein kinases (Table 1). Furthermore, the E3 ligase confers cies, and SCFskp2 ubiquitinates p27kip1, targeting it for protea-
specificity to ubiquitination when it transfers ubiquitin from an somal destruction (Cardozo and Pagano, 2007; Merlet et al.,
E2 to a particular substrate. For these reasons, E3 ubiquitin 2009). Researchers have identified one compound that prevents
ligases are attractive candidates as drug targets. In some cases, the incorporation of Skp2 into the SCFskp2 complex, which trig-
identifying compounds that disrupt the interaction of an E3 ligase gers cell death (i.e., autophagy) by stabilizing p27kip1 and
with its substrates has proven a frustrating experience for inducing G1/S cell-cycle arrest. This inhibitor synergizes with
several companies, and a number of programs have been Bortezomib and overcomes resistance to Bortezomib in models
unsuccessful. For example, we understand that several compa- of multiple myeloma. Moreover, the compound was active
nies have tried and failed to develop inhibitors of MuRF1, an E3 against aggressive leukemia cells (i.e., leukemia blasts) and
ligase involved in degrading myosin as a therapy for preventing plasma cells derived from patients (Chen et al., 2008).

690 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


SCFbTrCP1 is a Cullin RING E3 ligase that triggers the degradation for identifying inhibitors of many E3 ubiquitin ligases. This is
of IkBa, the inhibitory component of the proinflammatory tran- because, thus far, researchers have focused primarily on dis-
scription factor NF-kB. Therefore, drugs that target SCFbTrCP1 rupting the interaction between E3 ligases and their substrates,
may have potential as anti-inflammatory agents, and it is of great which is specific to particular E3 ligase-substrate pairs. More-
interest that an inhibitor of SCFbTrCP1 has been identified, which over, finding compounds to disrupt the interface of two proteins
prevents the polyubiquitination and degradation of IkBa (Nakajima can be intrinsically more difficult to achieve than searching for
et al., 2008). small molecules that block catalytic activity.
Researchers have also identified a small-molecule inhibitor of Surprisingly, little effort has been devoted to developing
Cdc4, the yeast ortholog of the mammalian Cullin RING E3 ligase compounds that disrupt the interactions between E2-conjugating
Fbw7 (F box and WD repeat domain-containing 7). A recent enzymes and E3 ligases. E2-E3 interactions are usually relatively
X-ray crystal structure (Orlicky et al., 2010) revealed that the weak (Ye and Rape, 2009) and may therefore be relatively easy
inhibitor inserts between two of the b strands of the WD40 to disrupt. Moreover, compounds that disturb the interaction
propeller domain of Cdc4, which are remote from the between an E2-conjugating enzyme and an E3 ligase could, in prin-
substrate-binding site. Binding of the inhibitor induces a long- ciple, exert their effects by binding to the E2, the E3, or the E2-E3
range conformational change that distorts the substrate-binding interface, creating the potential to identify three types of inhibitors
pocket and impedes recognition of the substrate. Thus, this from a single screen. There are 40 E2-conjugating enzymes en-
compound is one of the first allosteric inhibitors of an E3 ligase coded by the human genome; therefore, on average, each E2
to be identified and raises the possibility that other Cullin RING must interact productively with 15 E3 ligases. Compounds that
E3 ligases with WD40 domains may possess analogous pockets disrupt E2-E3 interactions by binding specifically to the E3 ligase
that could be targeted by inhibitors. A small-molecule inhibitor of could be identified by counterscreening with another E3 ligase
the SCFMet30 ligase was recently identified in a screen for small- that also forms a productive interaction with the same E2. Indeed,
molecule enhancers of the drug rapamycin (Aghajan et al., 2010). focusing efforts on large families of E3 ligases, such as the Cullin
To our knowledge, none of these compounds has yet entered RING ligases, may lead to the development of chemical libraries
clinical development, but they are proof-of-principle, demon- with the capability of disrupting many E2-E3 interactions.
strating that there is no particular fundamental barrier to identi- By analogy with kinases, perhaps the key to developing inhib-
fying inhibitors of the Cullin RING family of E3 ubiquitin ligases. itors of specific E2-E3 interactions is to find compounds that
bind to small hydrophobic pockets on E3 ligases located
The Future of Ubiquitin Drug Discovery proximal to the E2-E3 interface itself or to identify allosteric inhib-
There are striking parallels between the histories of protein phos- itors that disrupt the E2-E3 interaction by inducing long-range
phorylation and protein ubiquitination and their exploitation for conformational changes. The three-dimensional structure of an
the development of drugs to treat diseases (Table 1). Both bio- E2-ubiquitin thiol ester-E3 ligase complex has yet to be reported,
logical control mechanisms were identified many years ago, but such a structure might be extremely helpful in understanding
but interest in targeting them for drug discovery only started to how E2-E3 interactions could be disrupted. To crystallize such
take off in the 1990s. Indeed, the first compounds inhibiting a complex, it might be necessary to stabilize the E2-ubiquitin
components of these systems entered clinical trials at around thiol ester-E3 interactions by including a small molecule that
the same time (Bortezomib—1997, Gleevec—1998), and these inactivates E3 ligase function without affecting its ability to
drugs were among the fastest ever approved for clinical use bind to the E2-conjugating enzyme.
(Gleevec—2001, Bortezomib—2003). Both Gleevec and Borte- Another area where more effort will probably be fruitful is the
zomib subsequently achieved ‘‘blockbuster’’ status with current production of chemical libraries that target the different families
sales of about US$3 billion (Gleevec) and US$1.4 billion (Borte- of deubiquitinases. Although inhibitors of a few deubiquitinases
zomib) per annum. are under development, such as Usp2a, Usp7, Usp20, and
However, that is where their similarities end. Since the devel- Uch-L3, other deubiquitinases are also potentially rewarding
opment of Gleevec, 15 other drugs targeting a specific protein drug targets but seem to have attracted little attention so far.
kinase have been approved for clinical use, but no other drug tar- For example, Usp6 is an oncogene with transforming activity; re-
geting a particular component of the ubiquitin system has yet arrangements and fusions of this deubiquitinase are found in
been approved. In addition, kinase inhibitors currently under- a number of cancers (Oliveira et al., 2006). Moreover, the possi-
going clinical trials also outnumber the inhibitors of the ubiquitin bility of developing drugs that increase the expression and/or
system by more than ten to one (Table 1). activity of deubiquitinases also should not be ignored. For
Why has drug discovery in the ubiquitin system lagged so far example, the deubiquitinase BAP1 interacts with BRCA1, an
behind that of protein kinases, and what is needed to change E3 ligase frequently mutated in breast cancer. BAP1 enhances
this state of affairs in the future? In retrospect, one factor driving BRCA1-mediated inhibition of breast cancer cell growth and
the kinase field forward at such a rapid pace is the ease with may be a tumor suppressor gene that functions in the BRCA1
which large and varied chemical libraries can be synthesized growth control pathway (Jensen et al., 1998). Thus, drugs that
and exploited to develop inhibitors of many protein kinases. enhance the activity or expression of BAP1 could have thera-
Further, receptor tyrosine kinases have extracellular domains peutic potential for treating cancer.
that can also be targeted with therapeutic antibodies. In Experience with protein kinases has taught us that
contrast, although E3 ubiquitin ligases outnumber protein compounds developed as inhibitors of one protein kinase
kinases, researchers still have not developed a general approach commonly turn out to inhibit other protein kinases even more

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 691


potently (Bain et al., 2007) and thus can become leads in Predicting the future is notoriously difficult. However, given
completely different drug discovery programs. Sorafenib (also the diverse approaches and avenues that remain unexplored
called Nexavar), an approved drug for the treatment of renal in developing drugs targeted at the ubiquitin system, the
cell carcinoma, was originally developed as an inhibitor of authors of this article would be surprised if ubiquitin drug
a serine/threonine kinase Raf. However, now Sorafenib is discovery was not far more important in 10 years time than it
thought to exert its therapeutic benefit by inhibiting several tyro- is today. Nevertheless, only time will tell if ubiquitin drug
sine kinases, such as the PDGF receptor (Lierman et al., 2006). discovery will eventually rival in its importance that of kinase
Developing chemical libraries that target deubiquitinases is likely drug discovery.
to yield similar surprises and likely generate drug leads for
a number of these isopeptidases. ACKNOWLEDGMENTS
The success of Bortezomib and the advancement of the NAE-E1
We are grateful to Ron Hay, Thimo Kurz, and the reviewers of this Perspective
inhibitor MLN4924 into clinical trials suggest that there is vast
for making suggestions that improved the article. P.C. is a Royal Society
potential to develop more drugs targeted to general components Research Professor, and the work of his laboratory is supported by the UK
of the ubiquitin system. Drugs that block the same target by Medical Research Council, AstraZeneca, Boehringer Ingelheim, GlaxoSmithK-
distinct mechanisms can have strikingly different efficacies line, Merck-Serono, and Pfizer.
because their toxicities, half-lives in vivo, and pharmaco-dynamic
properties can vary substantially. Such targets might include other REFERENCES
E1-activating enzymes (e.g., the E1s for ubiquitination and SU-
MOylation) and other components of the proteasome. For Adams, J. (2002). Development of the proteasome inhibitor PS-341. Oncolo-
example, Bortezomib predominantly targets the chymotrypsin- gist 7, 9–16.

like activity of the proteasome, and drugs that inhibit the cas- Aghajan, M., Jonai, N., Flick, K., Fu, F., Luo, M., Cai, X., Ouni, I., Pierce, N.,
Tang, X., Lomenick, B., et al. (2010). Chemical genetics screen for enhancers
pase-like and trypsin-like activities of the proteasome may be
of rapamycin identifies a specific inhibitor of an SCF family E3 ubiquitin ligase.
more potent inhibitors or have different effects than Bortezomib. Nat. Biotechnol. 28, 738–742.
The 19S component of the proteasome is another underex-
Al-Hakim, A.K., Zagorska, A., Chapman, L., Deak, M., Peggie, M., and Alessi,
plored target. The 19S possesses ATPase activity, a polyubiqui- D.R. (2008). Control of AMPK-related kinases by USP9X and atypical Lys(29)/
tin-binding site, and deubiquitinase activities, all of which could Lys(33)-linked polyubiquitin chains. Biochem. J. 411, 249–260.
be targeted for drug development. Another possible target is Bain, J., Plater, L., Elliott, M., Shpiro, N., Hastie, C.J., McLauchlan, H., Kle-
p97/VCP, a protein that plays a key role in eliminating misfolded vernic, I., Arthur, J.S., Alessi, D.R., and Cohen, P. (2007). The selectivity of
proteins by the endoplasmic reticulum-associated degradation protein kinase inhibitors: a further update. Biochem. J. 408, 297–315.
pathway (ERAD). Indeed a small-molecule inhibitor of the Bennett, E.J., and Harper, J.W. (2008). DNA damage: ubiquitin marks the spot.
ATPase activity of p97/VCP has been discovered that blocks Nat. Struct. Mol. Biol. 15, 20–22.
proliferation of cancer cell lines (T.-F. Chou et al., 2008, FASEB Bhoj, V.G., and Chen, Z.J. (2009). Ubiquitylation in innate and adaptive immu-
J., abstract). Novel proteasome inhibitors might also be useful in nity. Nature 458, 430–437.
transplantation as a therapy for antibody- and cell-mediated acute Bianchi, G., Oliva, L., Cascio, P., Pengo, N., Fontana, F., Cerruti, F., Orsi, A.,
rejection (Everly et al., 2008). For example, Bortezomib has shown Pasqualetto, E., Mezghrani, A., Calbi, V., et al. (2009). The proteasome load
versus capacity balance determines apoptotic sensitivity of multiple myeloma
promise in reducing graft-versus-host disease and in reconstitut-
cells to proteasome inhibition. Blood 113, 3040–3049.
ing the immune system in some stem cell transplant patients.
Cardozo, T., and Pagano, M. (2007). Wrenches in the works: drug discovery
Inflammatory and autoimmune disorders may be treated with
targeting the SCF ubiquitin ligase and APC/C complexes. BMC Biochem. 8
selective inhibitors to a distinct class of proteasome, called the (Suppl 1), S9.
immunoproteasome. Expressed in monocytes and lympho- Carter, S., Bischof, O., Dejean, A., and Vousden, K.H. (2007). C-terminal modi-
cytes, the immunoproteasome regulates many facets of the fications regulate MDM2 dissociation and nuclear export of p53. Nat. Cell Biol.
immune response, in part by shaping the antigenic repertoire 9, 428–435.
presented on class I major histocompatibility complexes. The Chen, Q., Xie, W., Kuhn, D.J., Voorhees, P.M., Lopez-Girona, A., Mendy, D.,
immunoproteasome contains orthologs of the proteolytic activi- Corral, L.G., Krenitsky, V.P., Xu, W., Moutouh-de Parseval, L., et al. (2008).
ties associated with the ‘‘constitutive’’ 26S proteasome, Targeting the p27 E3 ligase SCF(Skp2) results in p27- and Skp2-mediated
including a component with chymotryptic-like activity, called cell-cycle arrest and activation of autophagy. Blood 111, 4690–4699.
LMP7. Recently, researchers developed a relatively selective Ciechanover, A., Hod, Y., and Hershko, A. (1978). A heat-stable polypeptide
component of an ATP-dependent proteolytic system from reticulocytes. Bio-
inhibitor of LMP7, which prevents the production of interleukin-
chem. Biophys. Res. Commun. 81, 1100–1105.
2 and interferon-g by activated T cells and interleukin-23 by acti-
Cohen, P. (2002a). The origins of protein phosphorylation. Nat. Cell Biol. 4,
vated monocytes. Furthermore, this inhibitor showed promise in
E127–E130.
treating arthritis in mouse models (Muchamuel et al., 2009).
Cohen, P. (2002b). Protein kinases—the major drug targets of the twenty-first
Finally, it is also worth noting that Mycobacterium tuberculosis
century? Nat. Rev. Drug Discov. 1, 309–315.
is the only bacterial pathogen known to have a proteasome.
Colland, F., Formstecher, E., Jacq, X., Reverdy, C., Planquette, C., Conrath,
Recently, one compound, oxathiazol-2-one, was identified with S., Trouplin, V., Bianchi, J., Aushev, V.N., Camonis, J., et al. (2009). Small-
preferential inhibition of the bacterial proteasome over the molecule inhibitor of USP7/HAUSP ubiquitin protease stabilizes and activates
human proteasome (Lin et al., 2009). Indeed, a selective inhibitor p53 in cells. Mol. Cancer Ther. 8, 2286–2295.
of this mycobacterial proteasome might be useful for treating Demetri, G.D., van Oosterom, A.T., Garrett, C.R., Blackstein, M.E., Shah, M.H.,
tuberculosis. Verweij, J., McArthur, G., Judson, I.R., Heinrich, M.C., Morgan, J.A., et al.

692 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


(2006). Efficacy and safety of sunitinib in patients with advanced gastrointes- Muchamuel, T., Basler, M., Aujay, M.A., Suzuki, E., Kalim, K.W., Lauer, C., Syl-
tinal stromal tumour after failure of imatinib: a randomised controlled trial. vain, C., Ring, E.R., Shields, J., Jiang, J., et al. (2009). A selective inhibitor of
Lancet 368, 1329–1338. the immunoproteasome subunit LMP7 blocks cytokine production and atten-
Duda, D.M., Borg, L.A., Scott, D.C., Hunt, H.W., Hammel, M., and Schulman, uates progression of experimental arthritis. Nat. Med. 15, 781–787.
B.A. (2008). Structural insights into NEDD8 activation of cullin-RING ligases: Musone, S.L., Taylor, K.E., Lu, T.T., Nititham, J., Ferreira, R.C., Ortmann, W.,
Conformational control of conjugation. Cell 134, 995–1006. Shifrin, N., Petri, M.A., Kamboh, M.I., Manzi, S., et al. (2008). Multiple polymor-
Everly, M.J., Everly, J.J., Susskind, B., Brailey, P., Arend, L.J., Alloway, R.R., phisms in the TNFAIP3 region are independently associated with systemic
Roy-Chaudhury, P., Govil, A., Mogilishetty, G., Rike, A.H., et al. (2008). Borte- lupus erythematosus. Nat. Genet. 40, 1062–1064.
zomib provides effective therapy for antibody- and cell-mediated acute rejec- Nakajima, H., Fujiwara, H., Furuichi, Y., Tanaka, K., and Shimbara, N. (2008). A
tion. Transplantation 86, 1754–1761. novel small-molecule inhibitor of NF-kappaB signaling. Biochem. Biophys.
Fischer, E.H., and Krebs, E.G. (1955). Conversion of phosphorylase b to phos- Res. Commun. 368, 1007–1013.
phorylase a in muscle extracts. J. Biol. Chem. 216, 121–132. Oliveira, A.M., Chou, M.M., Perez-Atayde, A.R., and Rosenberg, A.E. (2006).
Garnett, M.J., Mansfeld, J., Godwin, C., Matsusaka, T., Wu, J., Russell, P., Aneurysmal bone cyst: a neoplasm driven by upregulation of the USP6 onco-
Pines, J., and Venkitaraman, A.R. (2009). UBE2S elongates ubiquitin chains gene. J. Clin. Oncol. 24, e1–author reply e2.
on APC/C substrates to promote mitotic exit. Nat. Cell Biol. 11, 1363–1369. Orlicky, S., Tang, X., Neduva, V., Elowe, N., Brown, E.D., Sicheri, F., and Tyers,
Ikeda, F., Crosetto, N., and Dikic, I. (2010). What determines the specificity and M. (2010). An allosteric inhibitor of substrate recognition by the SCF(Cdc4)
outcomes of ubiquitin signaling? Cell 143, this issue, 677–681. ubiquitin ligase. Nat. Biotechnol. 28, 733–737.
Ito, T., Ando, H., Suzuki, T., Ogura, T., Hotta, K., Imamura, Y., Yamaguchi, Y., Rabut, G., and Peter, M. (2008). Function and regulation of protein neddylation.
and Handa, H. (2010). Identification of a primary target of thalidomide terato- ‘Protein modifications: beyond the usual suspects’ review series. EMBO Rep.
genicity. Science 327, 1345–1350. 9, 969–976.
Jensen, D.E., Proctor, M., Marquis, S.T., Gardner, H.P., Ha, S.I., Chodosh, L.A., Saha, A., and Deshaies, R.J. (2008). Multimodal activation of the ubiquitin
Ishov, A.M., Tommerup, N., Vissing, H., Sekido, Y., et al. (1998). BAP1: a novel ligase SCF by Nedd8 conjugation. Mol. Cell 32, 21–31.
ubiquitin hydrolase which binds to the BRCA1 RING finger and enhances Soucy, T.A., Smith, P.G., Milhollen, M.A., Berger, A.J., Gavin, J.M., Adhikari,
BRCA1-mediated cell growth suppression. Oncogene 16, 1097–1112. S., Brownell, J.E., Burke, K.E., Cardin, D.P., Critchley, S., et al. (2009). An inhib-
Jin, L., Williamson, A., Banerjee, S., Philipp, I., and Rape, M. (2008). Mecha- itor of NEDD8-activating enzyme as a new approach to treat cancer. Nature
nism of ubiquitin-chain formation by the human anaphase-promoting 458, 732–736.
complex. Cell 133, 653–665. Stevenson, L.F., Sparks, A., Allende-Vega, N., Xirodimas, D.P., Lane, D.P., and
Kisselev, A.F., Callard, A., and Goldberg, A.L. (2006). Importance of the Saville, M.K. (2007). The deubiquitinating enzyme USP2a regulates the p53
different proteolytic sites of the proteasome and the efficacy of inhibitors varies pathway by targeting Mdm2. EMBO J. 26, 976–986.
with the protein substrate. J. Biol. Chem. 281, 8582–8590. Tatham, M.H., Geoffroy, M.C., Shen, L., Plechanovova, A., Hattersley, N., Jaf-
Kolas, N.K., Chapman, J.R., Nakada, S., Ylanko, J., Chahwan, R., Sweeney, fray, E.G., Palvimo, J.J., and Hay, R.T. (2008). RNF4 is a poly-SUMO-specific
F.D., Panier, S., Mendez, M., Wildenhain, J., Thomson, T.M., et al. (2007). E3 ubiquitin ligase required for arsenic-induced PML degradation. Nat. Cell
Orchestration of the DNA-damage response by the RNF8 ubiquitin ligase. Biol. 10, 538–546.
Science 318, 1637–1640. Tcherpakov, M. (2010). Ubiquitin proteasome drug discovery (cancer and
Kovalenko, A., Chable-Bessia, C., Cantarella, G., Israël, A., Wallach, D., and other markets). BCC Research Report PHM066A.
Courtois, G. (2003). The tumour suppressor CYLD negatively regulates NF- Todi, S.V., Winborn, B.J., Scaglione, K.M., Blount, J.R., Travis, S.M., and Paul-
kappaB signalling by deubiquitination. Nature 424, 801–805. son, H.L. (2009). Ubiquitination directly enhances activity of the deubiquitinat-
Lawler, B. (2009). Kinase inhibitors. Global Markets BCC Market Research ing enzyme ataxin-3. EMBO J. 28, 372–382.
Report BIO053B. Tokunaga, F., Sakata, S., Saeki, Y., Satomi, Y., Kirisako, T., Kamei, K., Naka-
Lee, B.H., Lee, M.J., Park, S., Oh, D.C., Elsasser, S., Chen, P.C., Gartner, C., gawa, T., Kato, M., Murata, S., Yamaoka, S., et al. (2009). Involvement of linear
Dimova, N., Hanna, J., Gygi, S.P., et al. (2010). Enhancement of proteasome polyubiquitylation of NEMO in NF-kappaB activation. Nat. Cell Biol. 11, 123–132.
activity by a small-molecule inhibitor of USP14. Nature 467, 179–184. Trompouki, E., Hatzivassiliou, E., Tsichritzis, T., Farmer, H., Ashworth, A., and
Li, Z., Wang, D., Messing, E.M., and Wu, G. (2005). VHL protein-interacting Mosialos, G. (2003). CYLD is a deubiquitinating enzyme that negatively regu-
deubiquitinating enzyme 2 deubiquitinates and stabilizes HIF-1alpha. EMBO lates NF-kappaB activation by TNFR family members. Nature 424, 793–796.
Rep. 6, 373–378. Tseng, P.H., Matsuzawa, A., Zhang, W., Mino, T., Vignali, D.A., and Karin, M.
Lierman, E., Folens, C., Stover, E.H., Mentens, N., Van Miegroet, H., Scheers, (2010). Different modes of ubiquitination of the adaptor TRAF3 selectively acti-
W., Boogaerts, M., Vandenberghe, P., Marynen, P., and Cools, J. (2006). Sor- vate the expression of type I interferons and proinflammatory cytokines. Nat.
afenib is a potent inhibitor of FIP1L1-PDGFRalpha and the imatinib-resistant Immunol. 11, 70–75.
FIP1L1-PDGFRalpha T674I mutant. Blood 108, 1374–1376. Turer, E.E., Tavares, R.M., Mortier, E., Hitotsumatsu, O., Advincula, R., Lee, B.,
Lin, G., Li, D., de Carvalho, L.P., Deng, H., Tao, H., Vogt, G., Wu, K., Schneider, Shifrin, N., Malynn, B.A., and Ma, A. (2008). Homeostatic MyD88-dependent
J., Chidawanyika, T., Warren, J.D., et al. (2009). Inhibitors selective for myco- signals cause lethal inflamMation in the absence of A20. J. Exp. Med. 205,
bacterial versus human proteasomes. Nature 461, 621–626. 451–464.
MacKay, C., Déclais, A.C., Lundin, C., Agostinho, A., Deans, A.J., MacArtney, Wu, H., Tschopp, J., and Lin, S.C. (2007). Smac mimetics and TNFalpha: A
T.J., Hofmann, K., Gartner, A., West, S.C., Helleday, T., et al. (2010). Identifica- dangerous liaison? Cell 131, 655–658.
tion of KIAA1018/FAN1, a DNA repair nuclease recruited to DNA damage by Xia, Z.P., Sun, L., Chen, X., Pineda, G., Jiang, X., Adhikari, A., Zeng, W., and
monoubiquitinated FANCD2. Cell 142, 65–76. Chen, Z.J. (2009). Direct activation of protein kinases by unanchored polyubi-
ski, R.,
Maguire, M., Nield, P.C., Devling, T., Jenkins, R.E., Park, B.K., Polan quitin chains. Nature 461, 114–119.
Vlatkovic, N., and Boyd, M.T. (2008). MDM2 regulates dihydrofolate reductase Ye, Y., and Rape, M. (2009). Building ubiquitin chains: E2 enzymes at work.
activity through monoubiquitination. Cancer Res. 68, 3232–3242. Nat. Rev. Mol. Cell Biol. 10, 755–764.
Merlet, J., Burger, J., Gomes, J.E., and Pintard, L. (2009). Regulation of cullin- Zeng, W., Sun, L., Jiang, X., Chen, X., Hou, F., Adhikari, A., Xu, M., and Chen,
RING E3 ubiquitin-ligases by neddylation and dimerization. Cell. Mol. Life Sci. Z.J. (2010). Reconstitution of the RIG-I pathway reveals a signaling role of
66, 1924–1938. unanchored polyubiquitin chains in innate immunity. Cell 141, 315–330.

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 693


Leading Edge
Review

Pathogen-Mediated Posttranslational
Modifications: A Re-emerging Field
David Ribet1,2,3 and Pascale Cossart1,2,3,*
1Institut
Pasteur, Unité des Interactions Bactéries-Cellules, Département de Biologie Cellulaire et Infection, F-75015 Paris, France
2INSERM, U604, F-75015 Paris, France
3INRA, USC2020, F-75015 Paris, France

*Correspondence: pascale.cossart@pasteur.fr
DOI 10.1016/j.cell.2010.11.019

Posttranslational modifications are increasingly recognized as key strategies used by bacterial and
viral pathogens to modulate host factors critical for infection. A number of recent studies illustrate
how pathogens use these posttranslational modifications to target central signaling pathways in
the host cell, such as the NF-kB and MAP kinase pathways, which are essential for pathogens’ repli-
cation, propagation, and evasion from host immune responses. These discoveries open new
avenues for investigating the fundamental mechanisms of pathogen infection and the development
of new therapeutics.

Posttranslational modifications (PTMs) of proteins provide highly generally linked to the lysine residue of the target protein;
versatile tools and tricks used by both prokaryotic and eukary- however, a cysteine, serine, threonine, or N-terminal amino
otic cells to regulate the activity of key proteins. PTMs include group of a protein can also be modified. This conjugation
the addition of simple chemical groups, such as a phosphate, requires the successive activities of an E1-activating enzyme,
acetyl, methyl, or hydroxyl groups; more complex groups, an E2-conjugating enzyme, and then an E3 ligase. Ubiquitination
such as AMP, ADP-ribose, sugars, or lipids; and small polypep- is a fundamental PTM involved in many different cellular func-
tides, such as ubiquitin or ubiquitin-like proteins. They also tions, including the trafficking of membrane proteins, endocy-
include modifications of specific amino acid side chains (e.g., tosis, signal transduction, DNA repair, and transcription regula-
deamidation of glutamine residues) and the cleavage of a peptide tion. Ubiquitin itself contains seven lysines, K6, K11, K27, K29,
bond (i.e., proteolysis). K33, K48, and K63. Therefore, chains of ubiquitin can be formed
PTMs represent efficient strategies to modify activities, half- by attaching additional ubiquitin molecules to a lysine residue of
lives, or the intracellular localization of host proteins that are the previously attached ubiquitin.
critical for infection. The first report that a pathogen could K48-linked polyubiquitin chains play a fundamental role in
mediate a PTM occurred 40 years ago with the discovery that protein degradation by targeting proteins to the proteasome. In
diphtheria toxin, produced by Corynebacterium diphtheriae, contrast, K63-linked polyubiquitin chains are involved in nonpro-
ADP-ribosylates and thus inhibits the host Elongation Factor-2 teolytic processes, such as DNA repair and vesicular trafficking.
(EF-2) (Collier and Cole, 1969). This modification blocks transla- In addition to these ‘‘homotypic’’ K48- or K63-linked chains, in
tion in the intoxicated cells and thereby leads to cell death. which only one type of ubiquitin linkage is involved, mixed
Since then, a considerable number of host PTMs mediated, K11/K63-linked chains have also recently been described
induced, or counteracted by different pathogen-encoded virulence (Boname et al., 2010). The discovery of these ‘‘mixed’’ chains
factors have been reported (for reviews, see Ribet and Cossart, highlights that ubiquitin chains are probably more diverse and
2010; Randow and Lehner, 2009). In this Review, we discuss new complex than appreciated until now.
discoveries in the modulation of PTMs by pathogens. In the first Ubiquitination is reversible because eukaryotic cells encode
part, we focus on ubiquitin and ubiquitin-like proteins, which have proteases that are specific for ubiquitin. These proteases, called
emerged as central regulating modules targeted by both viral and deubiquitinases (DUBs), remove ubiquitin from their targets or
bacterial pathogens. We then discuss two recently identified cleave the bond between two linked ubiquitins.
PTMs catalyzed by bacterial pathogens, AMPylation and eliminyla- Ubiquitination constitutes an attractive target for a wide range
tion. In the third part, we describe how pathogens hijack certain of pathogens because it regulates many pathways in eukaryotic
PTMs to preferentially target specific host pathways to promote cells. Indeed, viruses and pathogenic bacteria can modulate the
their replication, propagation, and escape from the immune system. ubiquitination level of host proteins by inducing their monoubi-
quitination, their polyubiquitination with K48-linked chains
Ubiquitin and Ubiquitin-like Modifications Targeted (which then triggers their degradation), their polyubiquitination
by Pathogens with other types of ubiquitin chains, or their deubiquitination (re-
Ubiquitination viewed in Ribet and Cossart, 2010; Randow and Lehner, 2009).
Ubiquitination is the covalent attachment of ubiquitin, a small Some pathogen-encoded effectors display E3 ubiquitin ligase
polypeptide of 76 amino acids, to a target protein. Ubiquitin is activities. An important fraction of these viral or bacterial E3

694 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


Figure 1. Posttranslational Modification of
Host Proteins during Infection
Yersinia (blue) is an extracellular pathogen that
injects effectors into the host cell’s cytoplasm
using a specialized type III secretion system
(T3SS). Salmonella (red) triggers its own entry
into host cells and replicates in a remodeled
vacuole. It also secretes T3SS-dependent effec-
tors. After cell invasion, Listeria (green) escapes
from vacuoles and resides free in the cytoplasm,
where it replicates and starts moving using the
host cell’s actin. Interactions with host factors
are mediated by bacterial surface or secreted
proteins. Effectors from all three of these bacteria
(blue for Yersinia effectors, red for Salmonella
effectors, and green for Listeria effectors) alter
posttranslational modifications of host proteins
(purple) to facilitate pathogens’ replication, propa-
gation, and evasion from host immune responses .

rium (S. Typhimurium) is an invasive path-


ogen of the small intestine that, in mice,
causes a disease similar to human
typhoid fever. SseL, an effector secreted
by this bacterium, displays deubiquitinat-
ing activity in vitro. It suppresses ubiquiti-
nation and degradation of IkBa, a central
ligases shares structural homologies with eukaryotic E3 ligases, regulator of the NF-kB pathway (see below) (Figure 1) (Le Ne-
which are classically divided into HECT and RING E3s depend- grate et al., 2008). Infection with a strain of S. Typhimurium lack-
ing on their structures and mechanistic properties (reviewed in ing sseL leads to the accumulation of ubiquitinated proteins at
Kerscher et al., 2006). HECT E3 ligases transiently bind ubiquitin the site of replicating intracellular bacteria (Rytkönen
before transferring it to the target protein. In contrast, RING E3 et al., 2007). Strikingly, the decoration of intracytosolic bacteria
ligases do not link ubiquitin directly but rather facilitate ubiquiti- with polyubiquitinated proteins has recently been proposed as
nation by binding simultaneously to the charged E2 enzyme and a signal used by host cells to sense intracellular invaders
the protein target. (Figure 1). This signal triggers cytosolic defense pathways,
Recent studies have identified a new family of bacterial E3 such as autophagy, although the nature of ubiquitinated proteins
ligases with a structural domain completely distinct from the is unknown (Perrin et al., 2004; Thurston et al., 2009). Bacterial
eukaryotic RING and HECT domains (Hicks and Galán, 2010). DUBs may decrease this accumulation of polyubiquitinated
Studies have also identified viral E3 ligases structurally distinct proteins and thus might represent a strategy developed by intra-
from eukaryotic ones (Randow and Lehner, 2009). Whether cellular bacteria to escape these specific host defense systems.
these new E3 ligases also exist in eukaryotes is still unknown. Interestingly, pathogen-encoded proteins can also be directly
Whereas pathogens may have acquired eukaryotic-like E3 ubiquitinated by the host cell machinery. A striking example in
ligases by horizontal transfer from diverse eukaryotic sources, which PTMs by the host cell strongly alter the behavior of bacte-
the noneukaryotic E3 ligases may represent novel structures rial effectors is the Salmonella SopE and SptP proteins. These
evolved by pathogens to mimic the function of these essential two effectors contribute to the transient remodeling of the host
enzymes of the host cell. cell’s cytoskeleton during bacterial entry into the cell. SopE
In addition to encoding their own E3 ligases, some pathogens acts as a GEF (guanine nucleotide exchange factor) and
may encode adaptor proteins that bind host E3 enzymes and activates host Rho-GTPases, resulting in actin cytoskeleton
redirect them to specific targets. For example, two decades rearrangement, membrane ruffling, and subsequent bacterial
ago, a study found that this strategy is used by some human uptake. In contrast, SptP acts as a GAP (GTPase-activating
papillomaviruses (HPVs), which are associated with the develop- protein) to deactivate Rho-GTPases and allow the recovery of
ment of uterine cervix cancer. The E6 oncoproteins of HPV sero- the actin cytoskeleton’s normal architecture a few hours after
type 16 and 18 recruit a host E3 ligase to induce the degradation infection. Although SopE and SptP are codelivered by Salmo-
of the p53 tumor suppressor, thereby facilitating transformation nella, they exhibit different half-lives. SopE is rapidly polyubiqui-
of the infected cells (Scheffner et al., 1990). tinated and degraded by the host proteasome, whereas SptP
In addition to E3 ubiquitin ligases, pathogens also encode exhibits much slower degradation kinetics (Kubori and Galán,
DUB-like proteins. A few viral DUBs have been identified, but 2003). Recent studies found that Salmonella also hijacks the
their roles in vivo, as well as their host targets, are unknown. ubiquitination machinery to control one of its effectors, SopB,
In contrast, several DUB-like proteins have been characterized which displays two different activities depending on whether
in pathogenic bacteria. Salmonella enterica serovar Typhimu- the protein is ubiquitinated or not (Patel et al., 2009; Knodler

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 695


et al., 2009). Thus, by actively co-opting the ubiquitination O triggers the degradation of Ubc9, as well as the degradation
machinery of the host cell, Salmonella regulates the half-lives of some SUMOylated host proteins (Ribet et al., 2010). In
and activities of some of its key virulence factors. contrast to the ubiquitin system, which includes dozens of E2
SUMOylation enzymes in humans, the SUMO system has only one E2 enzyme.
In addition to ubiquitin, other polypeptides can be covalently Therefore, this degradation of Ubc9 leads to a blockade of the
linked to cellular proteins to modify their fate and functions. SUMOylation machinery and to a global decrease in the level
These polypeptides, which belong to the ubiquitin-like protein of SUMO-conjugated host proteins in infected cells. Thus, by
family, share high structural homology with ubiquitin, ranging decreasing SUMOylation in infected cells, Listeria may alter the
from 15% to 50% sequence similarity with it. SUMO (small activities of host factors critical for infection (Ribet et al., 2010).
ubiquitin-like modifier) belongs to the ubiquitin-like protein family Pathogen-encoded deSUMOylases can also cause a decrease
and is ubiquitous in the eukaryotic kingdom. The human genome in the SUMOylation level of host proteins. Indeed, this is the case
encodes three functional SUMO isoforms that can be linked to for XopD, a protein injected by the plant pathogen Xanthomonas
hundreds of different targets. Similar to the ubiquitin system, campestris into the cytoplasm of plant cells. This protein is
the conjugation of SUMO onto the lysine of a target protein a SUMO-specific protease, which induces deSUMOylation of
requires an E1, an E2, and an E3 SUMO enzyme. In parallel, several host factors when it is expressed in plant cells (Hotson
deSUMOylases regulate the SUMOylation level of cellular et al., 2003). XopD is known to alter host transcription, to promote
proteins by removing SUMO from its targets. pathogen multiplication, and to delay the onset of leaf chlorosis
SUMOylation is a fundamental PTM involved in transcription and necrosis. However, the exact roles of deSUMOylation in
regulation, intracellular transport, stress responses, the mainte- XopD’s effects are unknown (Kim et al., 2008).
nance of genome integrity, and many other biological processes. In addition to the induction or inhibition of SUMOylation of host
Although SUMOylation was first thought not to play a role in proteins, viral proteins can be SUMOylated themselves.
protein degradation, recent findings show that SUMO can trigger However, the role that these modifications play in virulence is
the recruitment of ubiquitin E3 ligases, such as RNF4 (RING unknown in most cases (Boggio and Chiocca, 2006). Surpris-
finger protein 4), leading to the ubiquitination and proteasomal ingly, examples of bacterial factors directly SUMOylated by
degradation of some SUMOylated proteins (Lallemand-Breiten- host enzymes have not been identified. It is, however, likely
bach et al., 2008; Tatham et al., 2008). that future studies will unveil the existence of such modifications,
As with the ubiquitin system, several bacterial and viral factors as well as their role in bacterial infection or in antibacterial
target or mimic components of the SUMOylation machinery, defenses.
thereby increasing or decreasing the SUMOylation level of host Neddylation
proteins (reviewed in Boggio and Chiocca, 2006; Ribet and Neddylation is another PTM that pathogens target during infec-
Cossart, 2010). For example, KSHV (Kaposi’s sarcoma-associ- tion. Nedd8, which is a member of the ubiquitin-like protein
ated herpes virus), a herpes virus responsible for Kaposi’s family, can be linked to cellular proteins in a fashion similar to
sarcoma development, encodes an enzyme, K-bZip, which ubiquitin (reviewed in Rabut and Peter, 2008). The major class
displays E3 SUMO ligase activity. This protein directly partici- of currently known Nedd8 substrates is Cullins. Cullins act as
pates in catalyzing SUMO conjugation to host targets, such as scaffolding proteins in the assembly of multisubunit RING E3
p53 and Retinoblastoma (Rb) protein (Chang et al., 2010). These ubiquitin enzymes, called Cullin RING ligases (CRLs). Neddyla-
modifications are proposed to play a role in modulating host tion of Cullins controls the activity of CRLs and thereby the ubiq-
genes expression in the early stage of viral infection (Chang uitination and degradation kinetics of CRLs substrates. As with
et al., 2010). ubiquitin, Nedd8 can be deconjugated from its targets by dened-
VP35, a protein encoded by Ebola virus, does not display dylases.
E3-like activity, but it binds to the host E3 SUMO enzyme Bacterial and viral pathogens can interfere with the neddyla-
PIAS1 (protein inhibitor of activated STAT 1) and increases the tion of host proteins. For example, the Epstein-Barr virus
SUMOylation level of IRF7 (interferon regulatory factor 7) (Chang encodes a protein BPLF1, which displays deneddylase activity
et al., 2009). This SUMOylation of IRF7 downregulates interferon (Gastaldello et al., 2010). During infection, BPLF1 deneddylates
transcription and may contribute to the dampening of the anti- Cullins, thereby inhibiting the activity of CRLs and stabilizing
viral response induced upon infection of Ebola virus (Chang several CRL substrates. In particular, this leads to the deregula-
et al., 2009). tion of the cell cycle and the establishment of an S-phase-like
Gam1, a protein encoded by an avian adenovirus, has an cellular environment, which is required for efficient replication
opposite effect on SUMOylation; it targets the host E1 SUMO of virus DNA (Gastaldello et al., 2010).
enzyme to proteasomal degradation, thereby inhibiting the A recent study also reported that Cif (cycle-inhibiting factor),
SUMOylation machinery and altering host transcription (Boggio a cyclomodulin translocated into cells by enteropathogenic
et al., 2004). Degradation of the SUMOylation machinery is and enterohemorrhagic Escherichia coli, binds to Nedd8-conju-
a strategy also used by Listeria monocytogenes, a food-borne gated CRLs of the host. This interaction inhibits the activity of the
bacterial pathogen responsible for listeriosis. Indeed, infection CRLs, leading to a deregulation of the host cell cycle (Jubelin
by L. monocytogenes leads to the degradation of Ubc9, the et al., 2010). Proteins with in vitro deneddylase activity have
human E2 SUMO enzyme (Ribet et al., 2010). Listeriolysin O is also been described in Chlamydia trachomatis, an obligate intra-
a pore-forming toxin secreted by this bacterium, which plays cellular bacterial pathogen. However, the role these deneddy-
a fundamental role in bacterial virulence (Figure 1). Listeriolysin lases play in infection remains unknown (Misaghi et al., 2006).

696 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


ISGylation ably rely on the ISGylation of cellular proteins rather than bacte-
ISG15 (interferon stimulated gene 15) is an ubiquitin-like protein rial proteins because the latter are not translated by the host cell
with two ubiquitin domains. The expression of ISG15 is induced machinery. Nevertheless, investigating the role of ISG15 in infec-
in response to type I interferons (IFN), a family of cytokines tions by bacterial pathogens will undoubtedly provide exciting
involved in the antiviral response. Consistent with this induction insights into the field of host-pathogens interactions.
in response to IFN, a growing number of studies are now high-
lighting the roles ISG15 plays in antiviral defense against several AMPylation and Eliminylation, New PTMs Mediated
types of viruses (reviewed in Skaug and Chen, 2010; Jeon et al., by Bacteria
2010). Conjugation of ISG15 to target proteins requires the AMPylation
activity of E1, E2, and E3 enzymes, which are also induced by AMPylation is the addition of an adenosine monophosphate
IFN. In contrast to the ubiquitin system, which includes hundreds (AMP) group onto a threonine, tyrosine, or, possibly, serine
of E3 enzymes, one unique E3 ISG15 enzyme, namely HERC5, residue of a protein. The AMPylation of host proteins by bacterial
modifies the vast majority of ISG15 substrates in human cells. pathogens was recently detected in cells during an infection with
Like with other ubiquitin-like modifications, ISGylation is revers- Vibrio parahaemolyticus, a human pathogen causing acute
ible; specific proteases, called deISGylases, remove ISG15 from gastroenteritis, and Histophilus somni, a pathogen responsible
its targets. for respiratory diseases and septicemia in cattle. Two virulence
The antiviral activity of ISG15 can be due to either the factors produced by these extracellular bacteria, namely VopS
ISGylation of host proteins critical for infection or the direct and IbpA, are able to reach the cytoplasm of host cells during
ISGylation of viral proteins (Skaug and Chen, 2010; Jeon et al., infection, where they use ATP to transfer an AMP moiety to
2010). This latter case has been described for the NS1 protein host Rho-GTPases (Figure 2) (Yarbrough et al., 2009; Worby
of influenza A virus (NS1A), which is ISGylated during infection. et al., 2009). This AMPylation alters the activity of Rho-GTPases,
This modification of NS1A was linked to an impairment of influ- which regulate the dynamics of the cell cytoskeleton.
enza replication, although the precise effect of the ISG15 addi- The catalytic domain responsible for AMPylation was mapped
tion on NS1A remains to be determined (Zhao et al., 2010; to the Fic domain (filamention induced by cAMP) of VopS and
Tang et al., 2010). IbpA. Fic domains are defined by a core sequence of nine amino
Interestingly, recent studies also proposed that the ISG15 acids containing an invariant histidine residue that is essential for
conjugation system may modify broadly, and somehow nonspe- the AMPylation (Yarbrough et al., 2009). Interestingly, proteins
cifically, newly synthesized proteins in a cotranslational manner containing Fic domains are found not only in prokaryotes but
(Durfee et al., 2010). This implies that, in the context of an inter- also in eukaryotes, and the existence of eukaryotic proteins
feron response, viral proteins, rather than cellular proteins, may able to catalyze AMPylation has been proposed (Worby et al.,
be the principal targets of ISGylation (Durfee et al., 2010). 2009; Kinch et al., 2009). Thus, AMPylation might represent
Although only a small fraction of viral proteins might be a new and important posttranslational modification in eukaryotic
ISGylated, it was proposed that ISGylation of viruses’ structural cells.
proteins, which precisely assemble into high-order structures, Legionella pneumophila is a human pathogen of the respira-
might impair the production of infectious viral particles. Indeed, tory tract responsible for a severe form of pneumonia, called
this was demonstrated for the human papillomavirus HPV16. Legionnaire’s disease. L. pneumophila encodes a factor, DrrA,
ISGylation of a small proportion of its structural protein L1 was which AMPylates the host protein Rab1b, a small GTPase
sufficient to have a dominant-negative effect on virus infectivity involved in intracellular vesicular transport (Muller et al., 2010).
(Durfee et al., 2010). The authors postulated that the ISGylation AMPylation of Rab1b leads to its constitutive activation, which
of host proteins could thus only be a side effect of the cell’s effort not only alters vesicular transport in infected cells but also
to target viral proteins. contributes to the formation of Legionella intracellular vacuoles
Consistent with the role of ISG15 in antiviral defense, several and aids bacterial replication.
viruses have evolved strategies to impair ISGylation (Skaug Interestingly, the catalytic domain of DrrA is distinct from the
and Chen, 2010; Jeon et al., 2010). In particular, studies Fic domains observed in VopS and IbpA (Muller et al., 2010).
have identified several viral proteins that can either mimic Thus, a wide diversity of both prokaryotic and eukaryotic
deISGylases or interfere with the ISGylation machinery of the enzymes may catalyze AMPylation, a posttranslational modifica-
infected cell. Indeed, the papain-like protease of SARS corona- tion that might represent an unsuspected way of regulating
virus and the ovarian tumor domain-containing proteases of various signaling pathways in the cell.
nairo- and arteriviruses all display ISG15-deconjugating activi- Eliminylation
ties (Lindner et al., 2005; Frias-Staheli et al., 2007). On the other Phosphorylation was the first covalent protein modification
hand, NS1 protein of influenza B virus binds to ISG15 and inhibits described. Since its discovery in the late 1950s, phosphorylation
its conjugation to target proteins (Yuan and Krug, 2001). By has emerged as a common and fundamental PTM. Phosphory-
inhibiting ISG15 conjugation or increasing ISG15 deconjugation, lation involves the reversible attachment of a phosphate group
all these effector proteins were proposed to decrease the to target proteins by forming a phosphoester bond. This addition
potential antiviral effect of ISGylation. generally occurs on hydroxyl groups of serine, threonine, or tyro-
The role of ISG15 in bacterial infections remains completely sine residues. Phosphorylation is reversible; phosphatases can
unknown. According to the study by Durfee et al. (2010), the hydrolyze the phosphoester bond to release the phosphate
participation of ISG15 in antibacterial defenses, if any, will prob- group and restore the amino acid in its unphosphorylated form.

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 697


Figure 2. Pathogen-Mediated PTMs Target
the Cytoskeleton and Immunoreceptors
Bacteria effector proteins (green) control the
dynamics of the host cell’s actin cytoskeleton by
posttranslationally modifying Rho-GTPases (left).
Viral effector proteins (blue) regulate posttransla-
tional modification of immunoreceptors, such as
the major histocompatibility complex class I
(MHC I) and the CD4 (cluster of differentiation 4)
molecules (right), thereby decreasing their expres-
sion at the cell surface and dampening immune
responses.

PTMs. In this section, we will focus on


several key cellular pathways that are
preferentially targeted by pathogens
through these PTMs.
Regulation of the Cytoskeleton
Dynamics by PTMs
The niches occupied by pathogens within
their hosts are quite diverse. Whereas
some bacterial pathogens remain strictly
extracellular, other bacteria, as well as
viruses, invade host cells and replicate
therein. For viruses, entry into host cell
is strictly required for the synthesis of viral
proteins and the production of new infec-
tious viral particles. Bacteria take refuge
inside host cells to escape humoral
immune response and to replicate in
a well-protected environment. To enter
Interestingly, a previously unknown enzymatic activity, called the cell and create such niches requires extensive remodeling
phosphothreonine lyase, was recently identified in three different of the host cell cytoskeleton, a multiprotein assembly of struc-
bacterial factors (Li et al., 2007; Mazurkiewicz et al., 2008; Zhang tural and regulatory elements. Indeed, many pathogen-induced
et al., 2007). These enzymes remove the phosphate group from PTMs target structural or regulatory components of the host
a threonine residue but, in contrast to classical phosphatases, cell’s cytoskeleton.
do not regenerate the hydroxyl group. Instead, this reaction, Listeria monocytogenes is a bacterium that can induce its own
nicknamed eliminylation, modifies threonine into dehydrobutyr- entry into a wide range of cells that are normally nonphagocytic.
ine, a residue that can no longer be phosphorylated (Brennan This internalization requires interactions between surface
and Barford, 2009). proteins of Listeria and host receptors. After successive PTMs,
The first factor identified with such activity is OspF, a protein these interactions trigger the recruitment of host factors and
produced by Shigella flexneri, the causative agent of bacillary the remodeling of host cell cytoskeleton required for internaliza-
dysentery in humans (Li et al., 2007). During infection, bacteria tion of the bacteria (Figure 1). For example, the interaction
directly secrete OspF into the host cell cytoplasm, where OspF between the Listeria surface protein InlA and its cellular receptor
helps to dampen the host immune responses by irreversibly E-cadherin promotes Listeria’s invasion into epithelial cells of the
dephosphorylating host MAP (mitogen-activated protein) intestine. Activation of E-cadherin by InlA leads to phosphoryla-
kinases (Figure 3) (Li et al., 2007; Arbibe et al., 2007). Phospho- tion and ubiquitination of E-cadherin by the Src kinase and
threonine lyases have been described only in S. flexneri, S. Typhi- the Hakai E3 ligase, respectively. These PTMs trigger the recruit-
murium, and the plant pathogen Pseudomonas syringae, and ment of the host’s clathrin-mediated endocytic machinery
MAP kinases are the only known targets of this PTM. However, followed by rearrangements of the actin cytoskeleton and inter-
we can expect that, as with AMPylation, some eukaryotic nalization of the bacteria (Bonazzi et al., 2008).
enzymes may also display this activity and that eliminylation In contrast, entry of Listeria into cells that do not express
might regulate numerous signaling pathways in eukaryotic cells. E-cadherin is mediated by another surface protein, InlB, which
interacts with and activates Met, the hepatocyte growth factor
Signaling Pathways Preferentially Targeted by (HGF) receptor (Figure 1). Similar to HGF activation, Met activa-
Pathogens by Alteration of Host PTMs tion by InlB induces its autophosphorylation and subsequent
Some pathogens produce several effectors that modulate the monoubiquitination by the host E3 ligase Cbl. This leads to
activity of host cell proteins by stimulating or counteracting their the recruitment of the host’s clathrin-dependent endocytic

698 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


Figure 3. Pathogen-Mediated PTMs Target
the MAP Kinase and NF-kB Signaling
Pathways
The MAP kinase (left) and NF-kB (right) signaling
cascades trigger immune responses in the host
cell during infections. Both bacterial (green) and
viral (blue) effectors weaken these immune
responses by inducing or counteracting post-
translational modifications of key components in
these critical pathways.

and cytokine expression (reviewed in Ak-


tories and Barbieri, 2005).
Inhibition of the NF-kB Pathway
The NF-kB pathway is an example of
a pathway tightly regulated by ubiquitina-
tion (Figure 3). The NF-kB pathway plays
a central role in inflammation and in the
establishment of both innate and immune
responses. Specific signals, such as
cytokines or microbial signatures, acti-
vate this pathway by switching on the
IkB kinase (IKK) complex. This leads to
the phosphorylation of IkBa, an inhibitor
protein that sequesters transcription
factors of the NF-kB family in the cyto-
plasm. Phosphorylated IkBa is then
recognized by specific ubiquitin E3
ligases, polyubiquitinated with K48-
linked chains, and targeted to the protea-
some for degradation. Destroying IkBa
leads to the release of NF-kB transcrip-
tion factors, allowing them to translocate
into the nucleus and initiate transcription
machinery, actin rearrangements, and ultimately, the internaliza- of various genes involved in host immune responses. Because
tion of the bacteria (Veiga and Cossart, 2005; Veiga et al., 2007). the NF-kB pathway plays a central role in immune responses,
To avoid being killed, pathogens can also actively inhibit their there is a strong evolutionary pressure on pathogens to prevent
engulfment by professional phagocytes. The mechanisms activation of this pathway during infection.
involved in this process may also require various pathogen effec- One possibility for dampening this pathway is to block the
tors to regulate the PTMs of host proteins (Figure 1). Pathogenic ubiquitination of IkBa, thereby inhibiting its proteasomal degra-
Yersinia species are involved in human diseases, ranging from dation and the translocation of NF-kB factors into the nucleus
enteric disorders to the plague. One virulence factor secreted (Figure 3). In numerous cases, factors achieve this goal by inter-
by Yersinia, YopH, displays potent phosphatase activity. It fering with the host ubiquitination machinery. For example,
decreases phosphorylation levels of host proteins involved in S. flexneri secretes the effector OspG into the host cell’s
focal adhesion complexes and impairs the cytoskeleton rear- cytoplasm, where it binds to and inhibits UbcH5, a host E2
rangements required for bacterial uptake. Another factor of ubiquitin enzyme involved in IkBa ubiquitination (Kim et al.,
Yersinia, YopT, is a protease that cleaves the membrane- 2005). The accessory protein Vpu (viral protein U) of HIV1 also
anchoring domain of host Rho-GTPases, leading to their irre- interferes with IkBa ubiquitination by inhibiting the E3 ubiquitin
versible detachment from the plasma membrane and their inac- ligase involved in IkBa’s modification (Bour et al., 2001). The
tivation (Figure 2 and Figure 1) (Shao et al., 2002). Thus, YopT DUB-like SseL factor produced by S. Typhimurium inhibits
contributes to the inhibition of bacterial phagocytosis by pre- IkBa ubiquitination in response to the TNF-a cytokine, suggest-
venting rearrangements of the actin cytoskeleton. ing that SseL acts directly by removing the K48-linked chains of
Finally, some bacterial pathogens, such as Clostridium diffi- IkBa (Le Negrate et al., 2008).
cile, secrete several toxins that posttranslationally modify host Numerous factors also target the IKK complex directly
Rho-GTPases, leading to their constitutive activation, inactiva- (Figure 3). For example, in addition to producing OspG, S. flex-
tion, or degradation (Figure 2). This alteration of Rho-GTPases neri also secretes IpaH9.8, an effector with E3 ubiquitin ligase
is widespread and allows bacteria to regulate the host cell’s activity. IpaH9.8 polyubiquitinates the NEMO/IKKg protein of
cytoskeleton in numerous ways, as well as gene transcription the IKK complex and targets it to the proteasome, thereby

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 699


impairing the phosphorylation and subsequent degradation of quitin chains in the control of immune responses (Boname
IkBa (Rohde et al., 2007; Ashida et al., 2010). L. monocytogenes et al., 2010). Some herpesvirus E3 ubiquitin ligases downregu-
intracellularly secretes InlC, which directly interacts with the late MHC I molecules by triggering their degradation by the
IKKa protein to block the phosphorylation of IkBa (Gouin et al., ERAD (endoplasmic reticulum-associated protein degradation)
2010). Similarly, YopJ/P, an effector produced by pathogenic pathway (reviewed in Randow and Lehner, 2009). Some viral
Yersinia species, mediates the acetylation of the IKKa and proteins, such as HIV Vpu accessory protein, can act as adap-
b proteins, which prevents their activation and subsequent tors of host E3 ubiquitin ligases to induce the proteasomal
IkBa phosphorylation (Mittal et al., 2006). degradation of other types of host immunoreceptors, such as
Interestingly, commensal bacteria of the human intestine can CD4 (cluster of differentiation 4) receptor on T cells (Schubert
also act on the NF-kB pathway. Indeed, some bacterial fermen- et al., 1998). Finally, bacterial pathogens, such as Salmonella,
tation products, such as butyrate or other short-chained fatty can decrease the expression of MHC class II molecules at the
acids, can stimulate the local production of reactive oxygen cell surface by modulating their ubiquitination, which also leads
species in intestinal epithelial cells. This leads to the inactivation to the dampening of host immune responses (Lapaque et al.,
of some redox-sensitive enzymes, such as E2 Nedd8 enzyme, 2009).
and therefore a decrease in the neddylation level of host
proteins. In this context, reduced neddylation levels, in particular Conclusion
the decrease in Cullin-1 neddylation, have been associated with Researchers have known for decades that pathogens interfere
a downregulation of the NF-kB pathway and hypothesized to with the host’s PTMs. However, the current ‘‘re-emergence’’ of
contribute to the inflammatory tolerance of the intestinal epithe- this field of research reflects the importance of controlling
lium toward commensal bacteria (Kumar et al., 2009). PTMs during infection and the complexity of these processes
Targeting of MAP Kinase Pathway in host-pathogen interactions. In this Review, we focused on
Similar to the NF-kB pathway, the MAP kinase pathway is how pathogens manipulate host PTMs and how they use these
another central signaling cascade that is essential for the activa- PTMs to solve their own biological needs.
tion of host innate immune responses. Therefore, not surpris- It should be stressed that pathogens may also actively co-opt
ingly, pathogens often target the MAP kinase pathway in order or be the passive targets of the host cell’s PTM machinery. As
to facilitate their infection (Figure 3). One effector protein mentioned above, pathogen-encoded proteins can indeed be
secreted intracellularly by Shigella is OspF, which possesses ubiquitinated, SUMOylated, or ISGylated, and like with host
phosphothreonine lyase activity. OspF irreversibly dephosphor- proteins, PTMs of pathogen-encoded proteins regulate these
ylates host MAP kinases and, therefore, was proposed to partic- factors’ half-lives, activities, intracellular localization, or binding
ipate in the dampening of host immune responses (Li et al., 2007; to other host- or pathogen-encoded factors. Therefore, it is
Arbibe et al., 2007). Interestingly, other bacterial virulence tempting to speculate that the diversity of known PTMs affecting
factors, such as SpvC from S. Typhimurium or HopAI1 from pathogen-encoded proteins will greatly increase in the near
the plant pathogen P. syringae, possess the same phospho- future.
threonine lyase activity as OspF and also target MAP kinases As the number of studies reporting crosstalk between different
of their hosts (Mazurkiewicz et al., 2008; Zhang et al., 2007). In PTMs increases, an emerging idea is that PTMs are more
addition to these factors, the Yersinia YopJ/P effector can inac- complex than originally anticipated. For example, in the NF-kB
tivate host MAP kinases by catalyzing their acetylation (Mittal signaling pathway alone, phosphorylation, SUMOylation, K63-
et al., 2006; Mukherjee et al., 2006). Finally, the anthrax lethal polyubiquitination, and K48-polyubiquitination act in synergy
factor, a subunit of the Anthrax toxin encoded by Bacillus anthra- to regulate the activation or the inhibition of transcriptional
cis, cleaves host MAP kinases, leading to their irreversible inac- responses. Targeting of these pathways by pathogens, there-
tivation (reviewed in Turk, 2007). fore, often requires a tightly controlled orchestration of multiple
Regulation of Cellular Immunoreceptors levels of PTMs.
To avoid detection by the immune system, some pathogens Studies on pathogen interference with host protein PTMs has
restrict the surface expression of fundamental molecules of the provided numerous insights into cell biology over the years. In
immune system by subverting host ubiquitination (Figure 2). particular, some pathogen effectors serve as invaluable tools
For example, KSHV encodes two E3 ubiquitin ligases, K3 and to study particular aspects of cell biology. For example, the
K5, which both target the host protein’s major histocompatibility C3 exoenzyme from Clostridium ADP-ribosylates and inhibits
complex class I (MHC I). An essential player of the immune multiple Rho-GTPases. Therefore, the C3 protein has been used
response, MHC I alerts the immune system to intracellular path- successfully to highlight the specific role of the Rho-GTPase in
ogens by sampling the protein repertoire of host cells and then stress fiber formation and to study the regulation of the actin
presenting peptides to cytotoxic T lymphocytes. K3 rapidly cytoskeleton dynamics in eukaryotic cells (Ridley and Hall,
mediates the polyubiquitination of MHC I molecules at the 1992; Ridley et al., 1992).
surface of the cell with K63-linked chains, leading to their endo- Finally, the development of new technologies, such as
cytosis and degradation. Interestingly, K5 also mediates polyu- improvements in mass spectrometry (especially the SILAC
biquitination of MHC I but with mixed K63 and K11 chains, [stable isotope labeling of amino acids in cell culture] technique;
instead of homotypic chains. Indeed, these mixed chains are Mann, 2006), will undoubtedly increase the list of currently
required for the internalization of MHC I by K5, thus highlighting, known PTMs and facilitate the understanding of their roles in
for the first time, the putative importance of such mixed polyubi- host-pathogen interactions. Identifying pathogen-encoded

700 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


enzymes that catalyze specific PTMs critical for infection will Epstein-Barr virus promotes viral DNA replication by regulating the activity
provide valuable new targets for drug development. Indeed, of cullin-RING ligases. Nat. Cell Biol. 12, 351–361.
the selective inhibition of these enzymes may constitute a prom- Gouin, E., Adib-Conquy, M., Balestrino, D., Nahori, M.A., Villiers, V., Colland,
ising strategy to counter these insidious invaders. F., Dramsi, S., Dussurget, O., and Cossart, P. (2010). The Listeria monocyto-
genes InlC protein interferes with innate immune responses by targeting
the IkappaB kinase subunit IKKalpha. Proc. Natl. Acad. Sci. USA 107,
ACKNOWLEDGMENTS 17333–17338.
Hicks, S.W., and Galán, J.E. (2010). Hijacking the host ubiquitin pathway:
We apologize to authors whose work could not be included because of space
structural strategies of bacterial E3 ubiquitin ligases. Curr. Opin. Microbiol.
constraints. Work in P.C.’s laboratory receives financial support from Institut
13, 41–46.
Pasteur, Inserm, INRA, ERC (advanced grant 233348), the Fondation le
Roch Les Mousquetaires, and the Fondation Louis-Jeantet. D.R. is supported Hotson, A., Chosed, R., Shu, H., Orth, K., and Mudgett, M.B. (2003). Xantho-
by a fellowship from the Association pour la Recherche sur le Cancer. P.C. is monas type III effector XopD targets SUMO-conjugated proteins in planta.
an international research scholar of the Howard Hughes Medical Institute. Mol. Microbiol. 50, 377–389.
Jeon, Y.J., Yoo, H.M., and Chung, C.H. (2010). ISG15 and immune diseases.
Biochim. Biophys. Acta 1802, 485–496.
REFERENCES
Jubelin, G., Taieb, F., Duda, D.M., Hsu, Y., Samba-Louaka, A., Nobe, R.,
Aktories, K., and Barbieri, J.T. (2005). Bacterial cytotoxins: targeting eukary- Penary, M., Watrin, C., Nougayrède, J.P., Schulman, B.A., et al. (2010). Path-
otic switches. Nat. Rev. Microbiol. 3, 397–410. ogenic bacteria target NEDD8-conjugated cullins to hijack host-cell signaling
pathways. PLoS Pathog. 6, e1001128.
Arbibe, L., Kim, D.W., Batsche, E., Pedron, T., Mateescu, B., Muchardt, C.,
Parsot, C., and Sansonetti, P.J. (2007). An injected bacterial effector targets Kerscher, O., Felberbaum, R., and Hochstrasser, M. (2006). Modification of
chromatin access for transcription factor NF-kappaB to alter transcription of proteins by ubiquitin and ubiquitin-like proteins. Annu. Rev. Cell Dev. Biol.
host genes involved in immune responses. Nat. Immunol. 8, 47–56. 22, 159–180.

Ashida, H., Kim, M., Schmidt-Supprian, M., Ma, A., Ogawa, M., and Sasa- Kim, D.W., Lenzen, G., Page, A.L., Legrain, P., Sansonetti, P.J., and Parsot, C.
kawa, C. (2010). A bacterial E3 ubiquitin ligase IpaH9.8 targets NEMO/IKK- (2005). The Shigella flexneri effector OspG interferes with innate immune
gamma to dampen the host NF-kappaB-mediated inflammatory response. responses by targeting ubiquitin-conjugating enzymes. Proc. Natl. Acad.
Nat. Cell Biol. 12, 66–73, 1–9. Sci. USA 102, 14046–14051.
Kim, J.G., Taylor, K.W., Hotson, A., Keegan, M., Schmelz, E.A., and Mudgett,
Boggio, R., and Chiocca, S. (2006). Viruses and sumoylation: recent highlights.
M.B. (2008). XopD SUMO protease affects host transcription, promotes path-
Curr. Opin. Microbiol. 9, 430–436.
ogen growth, and delays symptom development in xanthomonas-infected
Boggio, R., Colombo, R., Hay, R.T., Draetta, G.F., and Chiocca, S. (2004). tomato leaves. Plant Cell 20, 1915–1929.
A mechanism for inhibiting the SUMO pathway. Mol. Cell 16, 549–561.
Kinch, L.N., Yarbrough, M.L., Orth, K., and Grishin, N.V. (2009). Fido, a novel
Boname, J.M., Thomas, M., Stagg, H.R., Xu, P., Peng, J., and Lehner, P.J. AMPylation domain common to fic, doc, and AvrB. PLoS ONE 4, e5818.
(2010). Efficient internalization of MHC I requires lysine-11 and lysine-63 mixed
Knodler, L.A., Winfree, S., Drecktrah, D., Ireland, R., and Steele-Mortimer, O.
linkage polyubiquitin chains. Traffic 11, 210–220.
(2009). Ubiquitination of the bacterial inositol phosphatase, SopB, regulates its
Bonazzi, M., Veiga, E., Pizarro-Cerdá, J., and Cossart, P. (2008). Successive biological activity at the plasma membrane. Cell. Microbiol. 11, 1652–1670.
post-translational modifications of E-cadherin are required for InlA-mediated
Kubori, T., and Galán, J.E. (2003). Temporal regulation of salmonella virulence
internalization of Listeria monocytogenes. Cell. Microbiol. 10, 2208–2222.
effector function by proteasome-dependent protein degradation. Cell 115,
Bour, S., Perrin, C., Akari, H., and Strebel, K. (2001). The human immunodefi- 333–342.
ciency virus type 1 Vpu protein inhibits NF-kappa B activation by interfering
Kumar, A., Wu, H., Collier-Hyams, L.S., Kwon, Y.M., Hanson, J.M., and Neish,
with beta TrCP-mediated degradation of Ikappa B. J. Biol. Chem. 276,
A.S. (2009). The bacterial fermentation product butyrate influences epithelial
15920–15928.
signaling via reactive oxygen species-mediated changes in cullin-1 neddyla-
Brennan, D.F., and Barford, D. (2009). Eliminylation: a post-translational tion. J. Immunol. 182, 538–546.
modification catalyzed by phosphothreonine lyases. Trends Biochem. Sci.
Lallemand-Breitenbach, V., Jeanne, M., Benhenda, S., Nasr, R., Lei, M., Peres,
34, 108–114.
L., Zhou, J., Zhu, J., Raught, B., and de Thé, H. (2008). Arsenic degrades PML
Chang, P.C., Izumiya, Y., Wu, C.Y., Fitzgerald, L.D., Campbell, M., Ellison, T.J., or PML-RARalpha through a SUMO-triggered RNF4/ubiquitin-mediated
Lam, K.S., Luciw, P.A., and Kung, H.J. (2010). Kaposi’s sarcoma-associated pathway. Nat. Cell Biol. 10, 547–555.
herpesvirus (KSHV) encodes a SUMO E3 ligase that is SIM-dependent and
Lapaque, N., Hutchinson, J.L., Jones, D.C., Méresse, S., Holden, D.W.,
SUMO-2/3-specific. J. Biol. Chem. 285, 5266–5273.
Trowsdale, J., and Kelly, A.P. (2009). Salmonella regulates polyubiquitination
Chang, T.H., Kubota, T., Matsuoka, M., Jones, S., Bradfute, S.B., Bray, M., and surface expression of MHC class II antigens. Proc. Natl. Acad. Sci. USA
and Ozato, K. (2009). Ebola Zaire virus blocks type I interferon production by 106, 14052–14057.
exploiting the host SUMO modification machinery. PLoS Pathog. 5, e1000493.
Le Negrate, G., Faustin, B., Welsh, K., Loeffler, M., Krajewska, M., Hasegawa,
Collier, R.J., and Cole, H.A. (1969). Diphtheria toxin subunit active in vitro. P., Mukherjee, S., Orth, K., Krajewski, S., Godzik, A., et al. (2008). Salmonella
Science 164, 1179–1181. secreted factor L deubiquitinase of Salmonella typhimurium inhibits
Durfee, L.A., Lyon, N., Seo, K., and Huibregtse, J.M. (2010). The ISG15 conju- NF-kappaB, suppresses IkappaBalpha ubiquitination and modulates innate
gation system broadly targets newly synthesized proteins: Implications for the immune responses. J. Immunol. 180, 5045–5056.
antiviral function of ISG15. Mol. Cell 38, 722–732. Li, H., Xu, H., Zhou, Y., Zhang, J., Long, C., Li, S., Chen, S., Zhou, J.M., and
Frias-Staheli, N., Giannakopoulos, N.V., Kikkert, M., Taylor, S.L., Bridgen, A., Shao, F. (2007). The phosphothreonine lyase activity of a bacterial type III
Paragas, J., Richt, J.A., Rowland, R.R., Schmaljohn, C.S., Lenschow, D.J., effector family. Science 315, 1000–1003.
et al. (2007). Ovarian tumor domain-containing viral proteases evade ubiquitin- Lindner, H.A., Fotouhi-Ardakani, N., Lytvyn, V., Lachance, P., Sulea, T., and
and ISG15-dependent innate immune responses. Cell Host Microbe 2, Ménard, R. (2005). The papain-like protease from the severe acute respiratory
404–416. syndrome coronavirus is a deubiquitinating enzyme. J. Virol. 79, 15199–15208.
Gastaldello, S., Hildebrand, S., Faridani, O., Callegari, S., Palmkvist, M., Mann, M. (2006). Functional and quantitative proteomics using SILAC. Nat.
Di Guglielmo, C., and Masucci, M.G. (2010). A deneddylase encoded by Rev. Mol. Cell Biol. 7, 952–958.

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 701


Mazurkiewicz, P., Thomas, J., Thompson, J.A., Liu, M., Arbibe, L., Sansonetti, Schubert, U., Antón, L.C., Bacı́k, I., Cox, J.H., Bour, S., Bennink, J.R., Orlow-
P., and Holden, D.W. (2008). SpvC is a Salmonella effector with phosphothreo- ski, M., Strebel, K., and Yewdell, J.W. (1998). CD4 glycoprotein degradation
nine lyase activity on host mitogen-activated protein kinases. Mol. Microbiol. induced by human immunodeficiency virus type 1 Vpu protein requires the
67, 1371–1383. function of proteasomes and the ubiquitin-conjugating pathway. J. Virol. 72,
Misaghi, S., Balsara, Z.R., Catic, A., Spooner, E., Ploegh, H.L., and Starnbach, 2280–2288.
M.N. (2006). Chlamydia trachomatis-derived deubiquitinating enzymes in Shao, F., Merritt, P.M., Bao, Z., Innes, R.W., and Dixon, J.E. (2002). A Yersinia
mammalian cells during infection. Mol. Microbiol. 61, 142–150. effector and a Pseudomonas avirulence protein define a family of cysteine
Mittal, R., Peak-Chew, S.Y., and McMahon, H.T. (2006). Acetylation of MEK2 proteases functioning in bacterial pathogenesis. Cell 109, 575–588.
and I kappa B kinase (IKK) activation loop residues by YopJ inhibits signaling. Skaug, B., and Chen, Z.J. (2010). Emerging role of ISG15 in antiviral immunity.
Proc. Natl. Acad. Sci. USA 103, 18574–18579. Cell 143, 187–190.
Mukherjee, S., Keitany, G., Li, Y., Wang, Y., Ball, H.L., Goldsmith, E.J., and Tang, Y., Zhong, G., Zhu, L., Liu, X., Shan, Y., Feng, H., Bu, Z., Chen, H., and
Orth, K. (2006). Yersinia YopJ acetylates and inhibits kinase activation by Wang, C. (2010). Herc5 attenuates influenza A virus by catalyzing ISGylation of
blocking phosphorylation. Science 312, 1211–1214. viral NS1 protein. J. Immunol. 184, 5777–5790.
Muller, M.P., Peters, H., Blumer, J., Blankenfeldt, W., Goody, R.S., and Itzen, Tatham, M.H., Geoffroy, M.C., Shen, L., Plechanovova, A., Hattersley, N.,
A. (2010). The Legionella effector protein DrrA AMPylates the membrane traffic Jaffray, E.G., Palvimo, J.J., and Hay, R.T. (2008). RNF4 is a poly-SUMO-
regulator Rab1b. Science 329, 964–969. specific E3 ubiquitin ligase required for arsenic-induced PML degradation.
Patel, J.C., Hueffer, K., Lam, T.T., and Galán, J.E. (2009). Diversification of Nat. Cell Biol. 10, 538–546.
a Salmonella virulence protein function by ubiquitin-dependent differential Thurston, T.L., Ryzhakov, G., Bloor, S., von Muhlinen, N., and Randow, F.
localization. Cell 137, 283–294. (2009). The TBK1 adaptor and autophagy receptor NDP52 restricts the prolif-
Perrin, A.J., Jiang, X., Birmingham, C.L., So, N.S., and Brumell, J.H. (2004). eration of ubiquitin-coated bacteria. Nat. Immunol. 10, 1215–1221.
Recognition of bacteria in the cytosol of mammalian cells by the ubiquitin Turk, B.E. (2007). Manipulation of host signalling pathways by anthrax toxins.
system. Curr. Biol. 14, 806–811. Biochem. J. 402, 405–417.
Rabut, G., and Peter, M. (2008). Function and regulation of protein neddylation. Veiga, E., and Cossart, P. (2005). Listeria hijacks the clathrin-dependent endo-
‘Protein modifications: beyond the usual suspects’ review series. EMBO Rep. cytic machinery to invade mammalian cells. Nat. Cell Biol. 7, 894–900.
9, 969–976. Veiga, E., Guttman, J.A., Bonazzi, M., Boucrot, E., Toledo-Arana, A., Lin, A.E.,
Randow, F., and Lehner, P.J. (2009). Viral avoidance and exploitation of the Enninga, J., Pizarro-Cerdá, J., Finlay, B.B., Kirchhausen, T., and Cossart, P.
ubiquitin system. Nat. Cell Biol. 11, 527–534. (2007). Invasive and adherent bacterial pathogens co-opt host clathrin for
Ribet, D., and Cossart, P. (2010). Post-translational modifications in host cells infection. Cell Host Microbe 2, 340–351.
during bacterial infection. FEBS Lett. 584, 2748–2758. Worby, C.A., Mattoo, S., Kruger, R.P., Corbeil, L.B., Koller, A., Mendez, J.C.,
Ribet, D., Hamon, M., Gouin, E., Nahori, M.A., Impens, F., Neyret-Kahn, H., Zekarias, B., Lazar, C., and Dixon, J.E. (2009). The fic domain: regulation of
Gevaert, K., Vandekerckhove, J., Dejean, A., and Cossart, P. (2010). Listeria cell signaling by adenylylation. Mol. Cell 34, 93–103.
monocytogenes impairs SUMOylation for efficient infection. Nature 464, Yarbrough, M.L., Li, Y., Kinch, L.N., Grishin, N.V., Ball, H.L., and Orth, K.
1192–1195. (2009). AMPylation of Rho GTPases by Vibrio VopS disrupts effector binding
Ridley, A.J., and Hall, A. (1992). The small GTP-binding protein rho regulates and downstream signaling. Science 323, 269–272.
the assembly of focal adhesions and actin stress fibers in response to growth Yuan, W., and Krug, R.M. (2001). Influenza B virus NS1 protein inhibits conju-
factors. Cell 70, 389–399. gation of the interferon (IFN)-induced ubiquitin-like ISG15 protein. EMBO J. 20,
Ridley, A.J., Paterson, H.F., Johnston, C.L., Diekmann, D., and Hall, A. (1992). 362–371.
The small GTP-binding protein rac regulates growth factor-induced Zhang, J., Shao, F., Li, Y., Cui, H., Chen, L., Li, H., Zou, Y., Long, C., Lan, L.,
membrane ruffling. Cell 70, 401–410. Chai, J., et al. (2007). A Pseudomonas syringae effector inactivates MAPKs
Rohde, J.R., Breitkreutz, A., Chenal, A., Sansonetti, P.J., and Parsot, C. (2007). to suppress PAMP-induced immunity in plants. Cell Host Microbe 1, 175–185.
Type III secretion effectors of the IpaH family are E3 ubiquitin ligases. Cell Host Zhao, C., Hsiang, T.Y., Kuo, R.L., and Krug, R.M. (2010). ISG15 conjugation
Microbe 1, 77–83. system targets the viral NS1 protein in influenza A virus-infected cells. Proc.
Rytkönen, A., Poh, J., Garmendia, J., Boyle, C., Thompson, A., Liu, M., Natl. Acad. Sci. USA 107, 2253–2258.
Freemont, P., Hinton, J.C., and Holden, D.W. (2007). SseL, a Salmonella deu-
biquitinase required for macrophage killing and virulence. Proc. Natl. Acad. Note Added in Proof
Sci. USA 104, 3502–3507. It came recently to our attention that AMPylation is also called adenylylation
Scheffner, M., Werness, B.A., Huibregtse, J.M., Levine, A.J., and Howley, P.M. and was first discovered in the late sixties as regulating the glutamine synthe-
(1990). The E6 oncoprotein encoded by human papillomavirus types 16 and 18 tase of E. coli by modifying a tyrosine residue (Stadtman, E.R., 2001, The story
promotes the degradation of p53. Cell 63, 1129–1136. of glutamine synthetase regulation. J. Biol. Chem. 276, 44357–44364).

702 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


Leading Edge
Review

Modifications of Small RNAs


and Their Associated Proteins
Young-Kook Kim,1 Inha Heo,1 and V. Narry Kim1,*
1School of Biological Sciences and Center for National Creative Research, Seoul National University, Seoul 151-742, Korea

*Correspondence: narrykim@snu.ac.kr
DOI 10.1016/j.cell.2010.11.018

Small regulatory RNAs and their associated proteins are subject to diverse modifications that can
impinge on their abundance and function. Some of the modifications are under the influence of
cellular signaling, thus contributing to the dynamic regulation of RNA silencing.

Introduction temporal pattern of expression. An miRNA originates from a long


The past decade has witnessed an explosion of research on primary transcript (pri-miRNA) containing a local hairpin struc-
small regulatory RNAs that has yielded a basic understanding ture (Kim et al., 2009). In animals, the nuclear RNase III Drosha
of the many types of small RNAs in diverse eukaryotic species, liberates the hairpin-shaped precursor miRNA (pre-miRNA)
the protein factors involved, and the functions of key factors (Figure 1). The cytoplasmic RNase III Dicer removes the terminal
along the RNA silencing pathways. Much more remains to be loop to produce a small RNA duplex, consisting of the functional
learned, however, with recent studies unveiling interesting new miRNA strand and the passenger (*) strand (miRNA/miRNA*).
layers of regulation and complexity associated with small The duplex then binds to the Argonaute loading complex
RNAs. We now know that both small RNAs and their associated (comprised of Dicer, TRBP, and Ago), whose action leads to
protein factors can be modified at multiple steps in their biogen- the incorporation of the functional miRNA strand (mature miRNA)
esis and effector pathways. into Ago. The plant miRNA system differs from its animal coun-
Insight into modifications of small RNAs came initially from terparts in several aspects (Figure 2). The plant homolog of Dicer,
sequencing efforts, which made it clear that most microRNA Dicer-like 1 (DCL1), cleaves both pri-miRNA and pre-miRNA in
(miRNA) loci generate multiple isoforms (called isomiRs) apart the nucleus. Plant miRNAs generally show extensive comple-
from the reference sequence (Morin et al., 2008). Alternative/ mentary to their target mRNAs and induce endonucleolytic
inaccurate processing partly explains the heterogeneity, but cleavage of the targets.
a substantial portion of the variation is due to RNA modifications. Endogenous siRNAs (endo-siRNAs, 21 nt) are similar to
Small RNAs are modified either internally or externally by untem- miRNAs in their binding to the Ago subfamily proteins, in their
plated nucleotide addition, exonucleolytic trimming, 20 -O-methyl dependence on Dicer for biogenesis, and in exerting their regu-
transfer, and RNA editing. Protein factors in RNA silencing path- latory effects posttranscriptionally (Kim et al., 2009). But unlike
ways are also subject to various posttranslational modifications, miRNAs, endo-siRNAs originate from long double-stranded
including phosphorylation, hydroxylation, ubiquitination, and RNA precursors (dsRNAs), and their biogenesis does not require
methylation. In this Review, we focus on the recent develop- processing by Drosha. Endo-siRNAs are abundant in lower
ments in the modifications of RNAs and proteins in RNA silencing eukaryotes and in plants, whereas in mammals, they are found
pathways. in restricted tissues such as the ovary.
Piwi-interacting RNAs (piRNAs, 21–30 nt) associate with the
Small RNA Biogenesis Piwi subfamily of Argonaute proteins. piRNAs mediate the
RNA silencing is a widespread mechanism of gene regulation in silencing of repetitive elements in gonads via transcriptional
eukaryotes. At the core of all RNA silencing pathways lie small and posttranscriptional silencing mechanisms. Production of
RNAs (20–30 nt in length) associated with the Argonaute family piRNAs is not dependent on RNase III nucleases, and the steps
proteins (Kim et al., 2009). Small RNAs provide the specificity and factors involved in their biogenesis remain largely unknown.
of regulation by base-pairing to the target nucleic acids while
the Argonaute proteins execute the silencing effects. The Argo-
naute (Ago) proteins are grouped into Ago and Piwi subfamilies, Modifications of Small RNAs
and in animals, three types of small RNAs have been described: 30 End Modifications: Uridylation, Adenylation,
microRNAs (miRNAs), small interfering RNAs (siRNAs), and Piwi- and 20 -O-Methylation
interacting RNAs (piRNAs). The 30 ends of mature miRNAs are highly heterogeneous,
miRNAs (22 nt) induce mRNA degradation and/or transla- whereas the 50 ends are relatively invariable. The patterns
tional repression. Nucleotides 2–7, from the 50 end of the miRNA, and sources of heterogeneity seem to vary depending on the
are referred to as the ‘‘seed’’ and are critical for hybridization to miRNA species and the cell types. The 30 end often contains
the targets (Bartel, 2009). As a class, miRNAs are found in all extra 1–3 nucleotides that do not match the genomic DNA
tissues, although each miRNA species displays a unique spatio- sequences. These untemplated nucleotides are added by

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 703


Figure 1. Modifications in the Animal
MicroRNA Pathway
(Left) MicroRNAs (miRNAs) are subject to diverse
modifications. Pri-miRNAs are edited by ADARs,
which convert adenosine to inosine (I). RNA editing
inhibits processing and/or alters target specificity.
Pre-let-7 is regulated through uridylation. Lin28
recognizes pre-let-7 and, in turn, recruits a nucleo-
tidyl transferase TUT4 (mammal) or PUP-2
(worms), which adds an oligo-uridine tail at the 30
end of RNA. The uridylated pre-miRNA is resistant
to Dicer processing and subject to decay. TUT4
also uridylates mature miRNA (miR-26), which
reduces miRNA activity. Another nucleotidyl trans-
ferase GLD-2 adenylates mature miRNAs, which
reduces the activity of miRNA and/or increases
the stability of specific miRNAs (such as miR-122).
(Bottom) Mature miRNAs are degraded through
several mechanisms. In worms, a 50 /30 exonu-
clease XRN-2 degrades miRNAs that are released
from Ago. In flies and humans, extensive pairing
between miRNA/siRNA and target RNA triggers
tailing as well as 30 /50 trimming of miRNA/siRNA.
(Right) Protein factors, which are involved in the
miRNA pathway, are also subject to various post-
translational modifications. Human Drosha is
phosphorylated at two serine residues, S300/
S302, by an unknown kinase. Phosphorylation
localizes Drosha to the nucleus, where the pri-
miRNA processing occurs. MAP kinases Erk1/2
phosphorylate human TRBP at S142, S152,
S283, and S286, which increases the protein
stability of TRBP and Dicer. Ago2 is regulated by
multiple modifications. A prolyl hydroxylase
C-P4H(I) hydroxylates P700 in human Ago2, which
enhances stability of Ago2 and increases P body
localization. Phosphorylation of human Ago2 at
S387 by MAPKAPK2, which is induced by p38
pathway, also promotes P body localization of
Ago2. However, the biological significance of
P body localization of Ago2 remains unclear. In
mice, a stem cell-specific E3 ligase, mLin41, ubiq-
uitinates Ago2 and targets it for proteosome-
dependent degradation.

terminal nucleotidyl transferases that preferentially introduce that piRNA levels are reduced in hen1 mutant germ cells (Kam-
uridyl or adenyl residues to the 30 terminus of RNA. minga et al., 2010). In flies and mice, piRNAs are methylated
The first indication of 30 end modification of small RNA came by HEN1 orthologs, but the connection to stability control
from a hen1 mutant of Arabidopsis (Li et al., 2005). HEN1 is remains unclear (Horwich et al., 2007; Kirino and Mourelatos,
a methyl transferase that adds a methyl group to the 20 -OH at 2007; Ohara et al., 2007; Saito et al., 2007). In flies, dAgo2-bound
the 30 end of RNA (Yu et al., 2005). In hen1 mutants, miRNAs RNAs (mostly siRNAs) are protected by 20 -O-methylation from
are reduced in abundance and become heterogeneous in size being uridylated/adenylated, which in turn induces 30
due to uridylation at the 30 end. Because U tailing correlates exonucleolytic trimming (Ameres et al., 2010). In nematode
with the exonucleolytic degradation of mRNAs (Shen and worms, the role of 20 -O-methylation has yet to be determined.
Goodman, 2004), it was postulated that uridylation induces However, a subset of endo-siRNAs associated with an Ago
degradation of plant miRNAs and that the 20 -O-methyl moiety homolog CSR-1 is uridylated at the 30 end, and the uridyl trans-
is required to protect small RNAs from uridylation and decay ferase CDE-1 (also known as CID-1 or PUP-1) negatively regu-
(see below). Consistent with this notion, in green algae Chlamy- lates these siRNAs, indicating that uridylation serves as a trigger
domonas, a nucleotidyl transferase, MUT68, uridylates the 30 for decay (van Wolfswinkel et al., 2009).
end of small RNA, and the RRP6 exosome subunit facilitates Although mature miRNAs lack methylation in animals, uridyla-
small RNA decay in a manner dependent on MUT68 in vitro (Ibra- tion plays a significant role in the control of miRNA biogenesis.
him et al., 2010). Deletion of MUT68 results in elevated miRNA In mammalian embryonic stem cells, let-7 biogenesis is sup-
and siRNA levels, indicating that MUT68 and RRP6 collaborate pressed by the Lin28 protein that binds to the terminal loop of
in the turnover of mature small RNAs in plants. the let-7 precursors (Heo et al., 2008; Newman et al., 2008;
Similar links between 20 -O-methylation, uridylation, and decay Rybak et al., 2008; Viswanathan et al., 2008). Of interest, Lin28
appear to exist in animals. A recent study on the zebrafish Hen1 induces 30 uridylation of pre-let-7 by recruiting the terminal
homolog shows that piRNAs are uridylated and adenylated and nucleotidyl transferase TUT4 (also known as ZCCHC11) (Hagan

704 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


However, a recent study indicates that GLD-2 adenylates
most miRNAs, and the adenylation may affect their activity rather
than stability (Burroughs et al., 2010). Deep sequencing of Ago-
associated small RNAs shows that adenylated miRNAs are
relatively depleted in the Ago2 and Ago3 complexes, suggesting
that adenylation may interfere with Ago loading. Similarly, it
has been reported that uridylation of mature miR-26 by TUT4
results in the reduction of miR-26’s activity without altering the
miRNA levels (Jones et al., 2009). Therefore, it remains an inter-
esting but yet unresolved issue whether or not uridylation/
adenylation affects the stability of miRNAs in animals. One may
speculate that 30 modified miRNAs enter the silencing complex
with altered frequencies, which in turn affects the small RNA’s
sensitivity to nucleases. Further examination is needed to iden-
tify the players involved in these processes, particularly the
nucleases that recognize a U/A tail, and to dissect their action
mechanisms.
miRNA Decay
Several nucleases degrade small RNAs (Figures 1 and 2). An
Arabidopsis enzyme SDN1 (small RNA degrading nuclease,
a 30 -to-50 exonuclease) degrades single-stranded miRNAs
in vitro (Ramachandran and Chen, 2008). miRNAs accumulate
in a mutant lacking SDN1 and its related nucleases SDN2 and
SDN3, indicating that the SDN proteins may act redundantly to
Figure 2. RNA Modifications in the Plant miRNA Pathway degrade plant miRNAs. The 20 -O-methyl group at the 30 end of
In plants, both pri-miRNA and pre-miRNA are cleaved by DCL1/HYL1
complex. After cleavage, 30 ends of miRNA duplex are 20 -O-methylated by miRNAs, which is a general feature of plant miRNAs, has
a methyl transferase HEN1. The methylation protects miRNAs from uridylation a protective effect against SDN1 in in vitro assays. Of note, uridy-
and exonucleolytic degradation. In the green algae Chlamydomonas, the nu- lation causes a small but detectable protective effect in the same
cleotidyl transferase MUT68 attaches uridine residues at the 30 end of mature
in vitro assay, indicating that SDN1 is unlikely to be the nuclease
miRNA lacking a methyl group. Then, the RRP6 exosome subunit, a 30 -to-50
exonuclease, degrades the uridylated miRNAs. In Arabidopsis, a 30 /50 responsible for U-tail-promoted degradation. Given that RRP6
exonuclease SDN1 is reported to degrade mature miRNAs. (a 30 -to-50 exonuclease) facilitates decay of small RNAs in a
MUT68-dependent manner in Chlamydomonas extracts, multi-
et al., 2009; Heo et al., 2009). The oligo U-tail added by TUT4 ple enzymes may be involved in small RNA decay in plants,
blocks Dicer processing and facilitates the decay of pre-let-7. playing partially overlapping but differential roles (Ibrahim et al.,
The homologs of TUT4 may have related functions in other 2010).
organisms. In nematode worms, PUP-2 uridylates pre-let-7 In C. elegans, XRN-2 (a 50 -to-30 exonuclease) is involved in the
in vitro and suppresses the let-7 function in vivo (Lehrbach degradation of mature miRNAs (Chatterjee and Grosshans,
et al., 2009). 2009). Because miRNAs are tightly bound to and protected by
Let-7 is unlikely to be the only miRNA uridylated at the pre- Ago, it is unclear how XRN-2 accesses the 50 end of an miRNA
miRNA level. In support of this notion, untemplated 30 uridine is for decay. Of interest, larval lysate promotes efficient release of
frequently found in other mature miRNAs originating from the miRNA in vitro, implicating an as yet unknown factor that assists
30 arm of pre-miRNAs (but significantly less frequently in those the release of miRNA from the otherwise tightly associated Argo-
from the 50 arm) (Burroughs et al., 2010; Chiang et al., 2010). naute protein (Chatterjee and Grosshans, 2009). In Arabidopsis,
Because untemplated uridylation is observed in cells lacking two XRN-2 homologs, XRN2 and XRN3, degrade the loop of
Lin28, it will be interesting to determine which pre-miRNAs other miRNA precursor following processing, but they do not affect
than pre-let-7 are controlled by uridylation and to identify addi- mature miRNA levels (Gy et al., 2007).
tional factors required for pre-miRNA uridylation. In mammals, a general nuclease for miRNAs has yet to be
Although uridylation is generally thought to induce the decay identified. Knockdown of XRN-1 or an exosome subunit in
of small RNAs, adenylation may have the opposite conse- human cells results in only partial upregulation of miR-382, and
quence. In cottonwood P. trichoacarpa, many miRNA families XRN-2 depletion does not have a significant effect (Bail et al.,
are adenylated at their 30 ends, and adenylation prevents miRNA 2010). Thus, it awaits further investigation whether or not there
degradation in in vitro decay assay (Lu et al., 2009). In the case of is one major conserved pathway for miRNA decay in mammals.
mammalian miR-122, which is adenylated by cytoplasmic poly There have been intriguing reports of regulated decay of
(A) polymerase GLD-2 (or TUTase2), 30 end adenylation is also miRNAs. For instance, miR-29b is degraded in dividing cells
implicated in its stabilization (Katoh et al., 2009). In the liver of more rapidly than in mitotically arrested cells (Hwang et al.,
Gld-2 knockout mice, the steady-state level of mature miR-122 2007). In the central nervous system of Aplysia, the levels of
is reduced, and the abundance of target mRNAs of miR-122 miR-124 and miR-184 decrease in 1 hr after treatment with
increases. the neurotransmitter serotonin (Rajasethupathy et al., 2009).

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 705


Figure 3. Modifications in the Endo-siRNA
and piRNA Pathways
(A) Endogenous small interfering RNAs (endo-
siRNAs) are processed from long dsRNAs in
a Dicer-dependent manner and are loaded onto
Ago proteins. High-throughput sequencing data
show that the adenosine-to-inosine (I) editing
occurs in fly endo-siRNAs, likely by ADAR,
although the role of RNA editing is unknown. Fly
endo-siRNAs bound to dAgo2 are 20 -O-methyl-
ated by HEN1 homolog, which protects RNAs
from uridyl/adenyl tailing and degradation. In
worms, a subset of endo-siRNAs, which are asso-
ciated with an Ago homolog CSR-1, is uridylated
at the 30 end by the nucleotidyl transferase CDE-1.
(B) piRNAs are generated from single-stranded
RNA precursors that are processed by primary
processing and/or secondary processing (ping-
pong amplification cycle). piRNAs are associated
with Piwi subfamily proteins (PIWI). Animal piRNAs
are 20 -O-methylated by HEN1 orthologs. In zebra-
fish, depletion of hen1 induces uridylation of
piRNAs and facilitates decay, suggesting that
methylation stabilizes piRNAs. However, the phys-
iological significance of piRNA methylation in flies
and mammals remains unclear. PIWI proteins are methylated at arginine residues (sDMA, symmetrical dimethyl arginine) at their N termini by orthologs of the
methyl transferase PRMT5. In flies and mice, TDRD proteins interact with PIWI proteins through sDMA and may play important roles in piRNA metabolism.

Because U0126, an inhibitor of mitogen-activated protein kinase of small RNAs, which is influenced by the interaction with the
(MAPK), blocks the reduction of miR-124, the decay process target RNA.
may be dependent on the MAPK pathway. Of interest, a study miRNA Editing
on mammalian neuronal cells shows that most miRNAs turn Adenosine deaminases acting on RNAs (ADARs) convert
over more rapidly in neurons than in other cell types (Krol et al., adenosine to inosine on the dsRNA region of small RNA precur-
2010). Neuronal activation accelerates decay of the miRNAs, sors (Figure 1 and Figure 3A). Because inosine (I) pairs with
whereas blocking neuronal activity stabilizes the miRNAs. It cytosine instead of uridine, such edits could alter the structure
will be exciting to discover the nuclease(s) and the upstream of small RNA precursor, thereby interfering with processing.
signals for miRNA degradation in these systems. For instance, editing of pri-miR-142 by ADAR1 and ADAR2
Recently it has been shown that a polynucleotide phosphory- suppresses Drosha processing (Yang et al., 2006), whereas
lase (PNPase, a type I interferon-inducible 30 -to-50 exonuclease) that of pre-miR-151 by ADAR1 interferes with Dicer processing
binds specifically to several miRNAs (miR-221, miR-222, and (Kawahara et al., 2007a). Because hyperedited dsRNAs can be
miR-106b) and induces rapid turnover in a human melanoma targeted by the nuclease Tudor-SN, RNA editing may also desta-
cell line (Das et al., 2010). Because there is no apparent bilize small RNA precursors (Scadden, 2005). In rare cases, RNA
commonality in terms of the sequences, it is unclear how editing occurs in the seed sequence of miRNA, changing the
PNPase recognizes the miRNAs specifically. targeting specificity. In the brain, where ADAR is abundant,
As mentioned above, there is substantial evidence linking uri- miR-376 cluster miRNAs are frequently edited in the seed
dylation/adenylation and exonucleolytic attack on small RNAs. region and are redirected to repress a different set of mRNAs
A recent study provides evidence that extensive complemen- (Kawahara et al., 2007b). High-throughput sequencing of the
tarity between a small RNA and its target RNA triggers uridyl/ fly endo-siRNA pool also reveals evidence for RNA editing
adenyl tailing as well as 30 /50 trimming in flies and humans (Kawamura et al., 2008). The precursors of endo-siRNAs (long
(Figure 1) (Ameres et al., 2010). Animal small RNAs with high hairpins and sense-antisense pairs) may be targeted by ADARs,
complementarity to the targets, such as piRNAs and fly endo- although the functional significance of this siRNA modification is
siRNAs, appear to be generally protected by 20 -O-methylation unknown.
at the 30 end like plant small RNAs. It has been postulated that
animal miRNAs, which do not carry methylation, maintain only Posttranslational Protein Modifications
partial complementarity with their targets so as to avoid tailing Phosphorylation of RNase III Enzymes
and trimming of miRNAs. Of note, viruses seem to exploit a Human Dicer interacts with two related dsRNA-binding proteins,
related miRNA decay pathway to invade host cells more effec- TRBP and PACT. Although they do not influence Dicer process-
tively. Herpesvirus saimiri, a family of primate-infecting herpesvi- ing itself, TRBP and PACT stabilize Dicer and may also function
ruses, expresses viral noncoding RNAs called HSURs (H. saimiri in RISC assembly (Chendrimada et al., 2005; Haase et al., 2005;
U-rich RNAs). A recent report reveals that HSURs rapidly down- Lee et al., 2006). A recent study indicates that four serine
regulate host miR-27 and that base-pairing between HSUR and residues of human TRBP (S142, S152, S283, and S286) are
miR-27 is required for the degradation (Cazalla et al., 2010). phosphorylated by the MAP kinase Erk, which controls cell
These discoveries imply an additional layer of stability control proliferation, survival, and differentiation (Figure 1) (Paroo et al.,

706 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


2009). Phosphorylation enhances protein stability of TRBP, studies have revealed that PIWI proteins carry symmetrical
consequently elevating Dicer protein levels. Intriguingly, TRBP dimethyl arginine (sDMA) at their N termini. Arginine methylation
phosphorylation preferentially increases growth-promoting of PIWI is mediated by a methyl transferase PRMT5 (dPRMT5/
miRNAs such as miR-17, whereas tumor-suppressive let-7 is capsuleen [csul]/dart5 in Drosophila) (Figure 3B) (Heo and Kim,
reduced. The mechanism of selective downregulation of let-7 2009; Siomi et al., 2010). sDMA is recognized by Tudor
is unclear, but it may be an indirect effect. An interesting implica- domain-containing proteins (TDRDs), which are critical for germ-
tion of these findings is that the MAPK/Erk pathway exerts its line development. In both flies and mice, deletion of TDRDs alters
effects, in part, by regulating miRNA biogenesis. piRNA abundance and/or composition, indicating that TDRDs
Drosha, a nuclear enzyme for pri-miRNA processing (Lee play important roles in the piRNA metabolism through specific
et al., 2003), has recently been shown to be a direct target of binding to the sDMAs of PIWI proteins. How TDRDs act in the
posttranslational modification (Tang et al., 2010). Mass spec- piRNA pathway at a molecular level awaits further investigation.
trometry and mutagenesis studies reveal that human Drosha is
phosphorylated at serine 300 (S300) and serine 302 (S302) Perspectives
(Figure 1). Phosphorylation of these residues is essential for As we delve deeper and wider into the small RNA world, the
the nuclear localization of Drosha and is required for pri-miRNA emerging landscape becomes ever more complex on both the
processing. Because both endogenous and overexpressed RNA and protein sides. High-throughput analyses have uncov-
Drosha localize to the nucleus constitutively, it is unclear whether ered a considerable heterogeneity in small RNA populations.
or not the phosphorylation at S300/S302 is a regulated process. Some isomiRs are expressed differentially in certain tissues,
Understanding the physiological significance of this regulation suggesting that these variations may be associated with specific
will require the identification of the kinase that phosphorylates regulatory functions (Chiang et al., 2010). Biochemical and
Drosha. genetic studies also provide substantial evidence for the regula-
Argonaute2 Is a Target of Multiple Modifications tory roles of the modifications discussed in this Review. Thus, it
Ago2 is subject to multiple posttranslational modifications is likely that at least some of the observed heterogeneity reflects
(Figure 1). Human Ago2 binds to the type I collagen prolyl-4- multiple layers of regulation. We should be cautious, however, in
hydroxylase (C-P4H(I)) that hydroxylates Ago2 at proline 700 extrapolating the current evidence because it is unclear how
(Qi et al., 2008). Depletion of C-P4H(I) reduces the stability of much fraction of the small RNA and protein modifications trans-
the Ago2 protein and, accordingly, downregulates siRNA-medi- late into functional consequences and whether certain modifica-
ated silencing. Furthermore, hydroxylation is required for Ago2 tions simply reflect the noise of RNA metabolism.
localization to the processing body (P body), a cytoplasmic In addition to the functionality issue, a number of key questions
granule that is thought to be a site for RNA storage and degrada- remain to be answered. Are there conserved pathways and
tion. P body localization of Ago2 is also enhanced by phosphor- enzymes for RNA and protein modifications? If so, what are
ylation at serine 387, which is mediated by the p38 MAPK the similarities and differences? 20 -O-methylation is applied to
pathway (Zeng et al., 2008). However, given the controversy many small RNA pathways, but the details differ significantly in
over the direct role of P body in small RNA-mediated silencing, different systems. For instance, plant HEN1 acts on dsRNA
the biological significance of P body localization of Ago2 remains duplexes, whereas animal HEN1 homologs methylate ssRNA
unclear. loaded on Argonaute proteins. Uridylation/adenylation is carried
Ubiquitination also plays a part in the control of Ago2. Mouse out by a family of ribonucleotidyl transferases. How each
Lin41 (mLin41 or Trim71), a stem cell-specific Trim-NHL protein, member selectively recognizes its substrates is largely unknown.
inhibits the miRNA pathway (Rybak et al., 2009). As an E3 ubiq- RNA stability is likely to play important roles in RNA silencing
uitin ligase, mLin41 ubiquitinates Ago2 and targets it for protea- pathways. Decay pathways of small RNA are beginning to be
some-dependent degradation. Of interest, mLin41 is a target of unraveled, but there is no consensus between different species
let-7 miRNA, suggesting that mLin41 and let-7 may be engaged as yet. One possibility is that multiple enzymes act in parallel as
in a reciprocal negative feedback loop. Recently, other Trim-NHL in the mRNA decay pathway, which involves several 30 exonucle-
proteins have been reported to associate with the Argonaute ases, 50 exonucleases, and endonucleases. Some of the decay
proteins and affect miRNA pathway. Mei-P26 (fly) inhibits miRNA enzymes may function redundantly, and it remains one of the
biogenesis, whereas TRIM32 (mouse) and NHL-2 (worm) acti- major challenges in the field to identify them. Protein modifica-
vate the miRNA pathway (Hammell et al., 2009; Neumüller tion is also emerging as one of the key regulatory layers.
et al., 2008; Schwamborn et al., 2009). Their mechanism of Outstanding questions include which enzymes are involved,
action appears to be different than that of mLin41 because the what the in vivo significance of such modifications is, and
E3 ligase activity of Mei-P26 and TRIM32 is dispensable for their whether the protein modifications are developmentally regu-
effects and because NHL-2 enhances miRNA activity without lated. Future studies will reveal new types of modifications, addi-
a change in miRNA levels. tional regulatory factors, and their biological relevance.
Tudor Regulates PIWI Proteins The RNA silencing machinery should respond accurately to
The PIWI (P element-induced wimpy testis) clade proteins bind developmental and environmental cues. Most signaling path-
to Piwi-interacting RNAs (piRNAs) and silence transposable ways are thought to be connected to RNA silencing, but we
elements in gonads. Mouse has three PIWI homologs (MILI, are just beginning to understand the molecular links between
MIWI, and MIWI2), and there are three PIWI proteins in flies RNA silencing and cell signaling. What the upstream signals
(Aubergine [Aub], AGO3, and Piwi) (Kim et al., 2009). Recent are, how certain RNAs and proteins get specifically recognized,

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 707


and what the downstream effects of the modifications are await Hagan, J.P., Piskounova, E., and Gregory, R.I. (2009). Lin28 recruits the
elucidation. We also need to understand the interplay between TUTase Zcchc11 to inhibit let-7 maturation in mouse embryonic stem cells.
Nat. Struct. Mol. Biol. 16, 1021–1025.
different modifications. There appears to be a crosstalk between
certain modifications of RNA (such as methylation, uridylation, Hammell, C.M., Lubin, I., Boag, P.R., Blackwell, T.K., and Ambros, V. (2009).
nhl-2 Modulates microRNA activity in Caenorhabditis elegans. Cell 136,
and decay), which may influence their fate and function. It is likely
926–938.
that there is a crosstalk between the different posttranslational
modifications in the proteins involved in the biogenesis and Heo, I., and Kim, V.N. (2009). Regulating the regulators: posttranslational
modifications of RNA silencing factors. Cell 139, 28–31.
effector functions of small RNA silencing pathways. Under-
standing these networks will undoubtedly provide ample oppor- Heo, I., Joo, C., Cho, J., Ha, M., Han, J., and Kim, V.N. (2008). Lin28 mediates
the terminal uridylation of let-7 precursor MicroRNA. Mol. Cell 32, 276–284.
tunities to manipulate RNA silencing and will reveal new lessons
about gene regulation. Heo, I., Joo, C., Kim, Y.K., Ha, M., Yoon, M.J., Cho, J., Yeom, K.H., Han, J.,
and Kim, V.N. (2009). TUT4 in concert with Lin28 suppresses microRNA
biogenesis through pre-microRNA uridylation. Cell 138, 696–708.
ACKNOWLEDGMENTS Horwich, M.D., Li, C., Matranga, C., Vagin, V., Farley, G., Wang, P., and
Zamore, P.D. (2007). The Drosophila RNA methyltransferase, DmHen1,
We thank members of V.N.K.’s laboratory for helpful discussions and modifies germline piRNAs and single-stranded siRNAs in RISC. Curr. Biol.
comments. This work was supported by the Creative Research Initiatives 17, 1265–1272.
Program (20100000021) and the National Honor Scientist Program
(20100020415) through the National Research Foundation of Korea (NRF) Hwang, H.W., Wentzel, E.A., and Mendell, J.T. (2007). A hexanucleotide
and the BK21 Research Fellowships (I.H.) from the Ministry of Education, element directs microRNA nuclear import. Science 315, 97–100.
Science and Technology of Korea. We apologize to authors whose work has Ibrahim, F., Rymarquis, L.A., Kim, E.J., Becker, J., Balassa, E., Green, P.J.,
not been covered because of space limitations. and Cerutti, H. (2010). Uridylation of mature miRNAs and siRNAs by the
MUT68 nucleotidyltransferase promotes their degradation in Chlamydomo-
nas. Proc. Natl. Acad. Sci. USA 107, 3906–3911.
REFERENCES
Jones, M.R., Quinton, L.J., Blahna, M.T., Neilson, J.R., Fu, S., Ivanov, A.R.,
Wolf, D.A., and Mizgerd, J.P. (2009). Zcchc11-dependent uridylation of micro-
Ameres, S.L., Horwich, M.D., Hung, J.H., Xu, J., Ghildiyal, M., Weng, Z., and
RNA directs cytokine expression. Nat. Cell Biol. 11, 1157–1163.
Zamore, P.D. (2010). Target RNA-directed trimming and tailing of small
silencing RNAs. Science 328, 1534–1539. Kamminga, L.M., Luteijn, M.J., den Broeder, M.J., Redl, S., Kaaij, L.J.,
Roovers, E.F., Ladurner, P., Berezikov, E., and Ketting, R.F. (2010). Hen1 is
Bail, S., Swerdel, M., Liu, H., Jiao, X., Goff, L.A., Hart, R.P., and Kiledjian, M.
required for oocyte development and piRNA stability in zebrafish. EMBO J.
(2010). Differential regulation of microRNA stability. RNA 16, 1032–1039.
29, 3688–3700.
Bartel, D.P. (2009). MicroRNAs: target recognition and regulatory functions.
Katoh, T., Sakaguchi, Y., Miyauchi, K., Suzuki, T., Kashiwabara, S., Baba, T.,
Cell 136, 215–233.
and Suzuki, T. (2009). Selective stabilization of mammalian microRNAs by 30
Burroughs, A.M., Ando, Y., de Hoon, M.J., Tomaru, Y., Nishibu, T., Ukekawa, adenylation mediated by the cytoplasmic poly(A) polymerase GLD-2. Genes
R., Funakoshi, T., Kurokawa, T., Suzuki, H., Hayashizaki, Y., and Daub, C.O. Dev. 23, 433–438.
(2010). A comprehensive survey of 30 animal miRNA modification events and
Kawahara, Y., Zinshteyn, B., Chendrimada, T.P., Shiekhattar, R., and Nishi-
a possible role for 30 adenylation in modulating miRNA targeting effectiveness.
kura, K. (2007a). RNA editing of the microRNA-151 precursor blocks cleavage
Genome Res. 20, 1398–1410.
by the Dicer-TRBP complex. EMBO Rep. 8, 763–769.
Cazalla, D., Yario, T., Steitz, J.A., and Steitz, J. (2010). Down-regulation of
Kawahara, Y., Zinshteyn, B., Sethupathy, P., Iizasa, H., Hatzigeorgiou, A.G.,
a host microRNA by a Herpesvirus saimiri noncoding RNA. Science 328,
and Nishikura, K. (2007b). Redirection of silencing targets by adenosine-to-
1563–1566.
inosine editing of miRNAs. Science 315, 1137–1140.
Chatterjee, S., and Grosshans, H. (2009). Active turnover modulates mature
Kawamura, Y., Saito, K., Kin, T., Ono, Y., Asai, K., Sunohara, T., Okada, T.N.,
microRNA activity in Caenorhabditis elegans. Nature 461, 546–549.
Siomi, M.C., and Siomi, H. (2008). Drosophila endogenous small RNAs bind to
Chendrimada, T.P., Gregory, R.I., Kumaraswamy, E., Norman, J., Cooch, N., Argonaute 2 in somatic cells. Nature 453, 793–797.
Nishikura, K., and Shiekhattar, R. (2005). TRBP recruits the Dicer complex to
Kim, V.N., Han, J., and Siomi, M.C. (2009). Biogenesis of small RNAs in
Ago2 for microRNA processing and gene silencing. Nature 436, 740–744.
animals. Nat. Rev. Mol. Cell Biol. 10, 126–139.
Chiang, H.R., Schoenfeld, L.W., Ruby, J.G., Auyeung, V.C., Spies, N., Baek,
Kirino, Y., and Mourelatos, Z. (2007). The mouse homolog of HEN1 is a poten-
D., Johnston, W.K., Russ, C., Luo, S., Babiarz, J.E., et al. (2010). Mammalian
tial methylase for Piwi-interacting RNAs. RNA 13, 1397–1401.
microRNAs: experimental evaluation of novel and previously annotated genes.
Genes Dev. 24, 992–1009. Krol, J., Busskamp, V., Markiewicz, I., Stadler, M.B., Ribi, S., Richter, J.,
Duebel, J., Bicker, S., Fehling, H.J., Schübeler, D., et al. (2010). Characterizing
Das, S.K., Sokhi, U.K., Bhutia, S.K., Azab, B., Su, Z.Z., Sarkar, D., and Fisher,
light-regulated retinal microRNAs reveals rapid turnover as a common
P.B. (2010). Human polynucleotide phosphorylase selectively and preferen-
property of neuronal microRNAs. Cell 141, 618–631.
tially degrades microRNA-221 in human melanoma cells. Proc. Natl. Acad.
Sci. USA 107, 11948–11953. Lee, Y., Ahn, C., Han, J., Choi, H., Kim, J., Yim, J., Lee, J., Provost, P.,
Rådmark, O., Kim, S., and Kim, V.N. (2003). The nuclear RNase III Drosha initi-
Gy, I., Gasciolli, V., Lauressergues, D., Morel, J.B., Gombert, J., Proux, F.,
ates microRNA processing. Nature 425, 415–419.
Proux, C., Vaucheret, H., and Mallory, A.C. (2007). Arabidopsis FIERY1,
XRN2, and XRN3 are endogenous RNA silencing suppressors. Plant Cell 19, Lee, Y., Hur, I., Park, S.Y., Kim, Y.K., Suh, M.R., and Kim, V.N. (2006). The role
3451–3461. of PACT in the RNA silencing pathway. EMBO J. 25, 522–532.
Haase, A.D., Jaskiewicz, L., Zhang, H., Lainé, S., Sack, R., Gatignol, A., and Lehrbach, N.J., Armisen, J., Lightfoot, H.L., Murfitt, K.J., Bugaut, A., Balasu-
Filipowicz, W. (2005). TRBP, a regulator of cellular PKR and HIV-1 virus bramanian, S., and Miska, E.A. (2009). LIN-28 and the poly(U) polymerase
expression, interacts with Dicer and functions in RNA silencing. EMBO Rep. PUP-2 regulate let-7 microRNA processing in Caenorhabditis elegans. Nat.
6, 961–967. Struct. Mol. Biol. 16, 1016–1020.

708 Cell 143, November 24, 2010 ª2010 Elsevier Inc.


Li, J., Yang, Z., Yu, B., Liu, J., and Chen, X. (2005). Methylation protects miR- Rybak, A., Fuchs, H., Hadian, K., Smirnova, L., Wulczyn, E.A., Michel, G.,
NAs and siRNAs from a 30 -end uridylation activity in Arabidopsis. Curr. Biol. 15, Nitsch, R., Krappmann, D., and Wulczyn, F.G. (2009). The let-7 target gene
1501–1507. mouse lin-41 is a stem cell specific E3 ubiquitin ligase for the miRNA pathway
Lu, S., Sun, Y.H., and Chiang, V.L. (2009). Adenylation of plant miRNAs. protein Ago2. Nat. Cell Biol. 11, 1411–1420.
Nucleic Acids Res. 37, 1878–1885. Saito, K., Sakaguchi, Y., Suzuki, T., Suzuki, T., Siomi, H., and Siomi, M.C.
Morin, R.D., O’Connor, M.D., Griffith, M., Kuchenbauer, F., Delaney, A., (2007). Pimet, the Drosophila homolog of HEN1, mediates 20 -O-methylation
Prabhu, A.L., Zhao, Y., McDonald, H., Zeng, T., Hirst, M., et al. (2008). Appli- of Piwi- interacting RNAs at their 30 ends. Genes Dev. 21, 1603–1608.
cation of massively parallel sequencing to microRNA profiling and discovery Scadden, A.D. (2005). The RISC subunit Tudor-SN binds to hyper-edited
in human embryonic stem cells. Genome Res. 18, 610–621. double-stranded RNA and promotes its cleavage. Nat. Struct. Mol. Biol. 12,
489–496.
Neumüller, R.A., Betschinger, J., Fischer, A., Bushati, N., Poernbacher, I.,
Mechtler, K., Cohen, S.M., and Knoblich, J.A. (2008). Mei-P26 regulates micro- Schwamborn, J.C., Berezikov, E., and Knoblich, J.A. (2009). The TRIM-NHL
RNAs and cell growth in the Drosophila ovarian stem cell lineage. Nature 454, protein TRIM32 activates microRNAs and prevents self-renewal in mouse
241–245. neural progenitors. Cell 136, 913–925.
Newman, M.A., Thomson, J.M., and Hammond, S.M. (2008). Lin-28 interaction Shen, B., and Goodman, H.M. (2004). Uridine addition after microRNA-
with the Let-7 precursor loop mediates regulated microRNA processing. RNA directed cleavage. Science 306, 997.
14, 1539–1549. Siomi, M.C., Mannen, T., and Siomi, H. (2010). How does the royal family of
Ohara, T., Sakaguchi, Y., Suzuki, T., Ueda, H., Miyauchi, K., and Suzuki, T. Tudor rule the PIWI-interacting RNA pathway? Genes Dev. 24, 636–646.
(2007). The 30 termini of mouse Piwi-interacting RNAs are 20 -O-methylated. Tang, X., Zhang, Y., Tucker, L., and Ramratnam, B. (2010). Phosphorylation of
Nat. Struct. Mol. Biol. 14, 349–350. the RNase III enzyme Drosha at Serine300 or Serine302 is required for its
Paroo, Z., Ye, X., Chen, S., and Liu, Q. (2009). Phosphorylation of the human nuclear localization. Nucleic Acids Res. 38, 6610–6619.
microRNA-generating complex mediates MAPK/Erk signaling. Cell 139, van Wolfswinkel, J.C., Claycomb, J.M., Batista, P.J., Mello, C.C., Berezikov,
112–122. E., and Ketting, R.F. (2009). CDE-1 affects chromosome segregation through
Qi, H.H., Ongusaha, P.P., Myllyharju, J., Cheng, D., Pakkanen, O., Shi, Y., Lee, uridylation of CSR-1-bound siRNAs. Cell 139, 135–148.
S.W., Peng, J., and Shi, Y. (2008). Prolyl 4-hydroxylation regulates Argonaute 2 Viswanathan, S.R., Daley, G.Q., and Gregory, R.I. (2008). Selective blockade
stability. Nature 455, 421–424. of microRNA processing by Lin28. Science 320, 97–100.
Rajasethupathy, P., Fiumara, F., Sheridan, R., Betel, D., Puthanveettil, S.V., Yang, W., Chendrimada, T.P., Wang, Q., Higuchi, M., Seeburg, P.H., Shiekhat-
Russo, J.J., Sander, C., Tuschl, T., and Kandel, E. (2009). Characterization tar, R., and Nishikura, K. (2006). Modulation of microRNA processing and
of small RNAs in Aplysia reveals a role for miR-124 in constraining synaptic expression through RNA editing by ADAR deaminases. Nat. Struct. Mol.
plasticity through CREB. Neuron 63, 803–817. Biol. 13, 13–21.
Ramachandran, V., and Chen, X. (2008). Degradation of microRNAs by a family Yu, B., Yang, Z., Li, J., Minakhina, S., Yang, M., Padgett, R.W., Steward, R.,
of exoribonucleases in Arabidopsis. Science 321, 1490–1492. and Chen, X. (2005). Methylation as a crucial step in plant microRNA biogen-
Rybak, A., Fuchs, H., Smirnova, L., Brandt, C., Pohl, E.E., Nitsch, R., and esis. Science 307, 932–935.
Wulczyn, F.G. (2008). A feedback loop comprising lin-28 and let-7 controls Zeng, Y., Sankala, H., Zhang, X., and Graves, P.R. (2008). Phosphorylation of
pre-let-7 maturation during neural stem-cell commitment. Nat. Cell Biol. 10, Argonaute 2 at serine-387 facilitates its localization to processing bodies.
987–993. Biochem. J. 413, 429–436.

Cell 143, November 24, 2010 ª2010 Elsevier Inc. 709


The ER UDPase ENTPD5 Promotes Protein
N-Glycosylation, the Warburg Effect,
and Proliferation in the PTEN Pathway
Min Fang,1 Zhirong Shen,1 Song Huang,1 Liping Zhao,1 She Chen,2 Tak W. Mak,3 and Xiaodong Wang1,2,*
1Howard Hughes Medical Institute and Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas,

5323 Harry Hines Boulevard, Dallas, TX 75390, USA


2National Institute of Biological Sciences, Zhongguancun Life Science Park, Beijing 102206, China
3The Campbell Family Institute for Breast Cancer Research, Princess Margaret Hospital, Toronto, ON M5G 2M9, Canada

*Correspondence: xiaodong.wang@utsouthwestern.edu
DOI 10.1016/j.cell.2010.10.010

SUMMARY et al., 2002). PTEN, by dephosphorylating PIP3 back to PIP2,


antagonizes the signal generated by PI3K (Maehama and Dixon,
PI3K and PTEN lipid phosphatase control the level of 1998). The importance of the PI3K/PTEN pathway has been
cellular phosphatidylinositol (3,4,5)-trisphosphate, manifested by frequent PI3K gain of function, or PTEN loss of
an activator of AKT kinases that promotes cell function, in a variety of human cancers (reviewed by Yuan and
growth and survival. Mutations activating AKT are Cantley, 2008; Keniry and Parsons, 2008).
commonly observed in human cancers. We report AKT activation also contributes to the elevation of aerobic
glycolysis seen in tumor cells, known as the Warburg effect
here that ENTPD5, an endoplasmic reticulum (ER)
(Elstrom et al., 2004; Warburg, 1925). AKT promotes cell-surface
enzyme, is upregulated in cell lines and primary
expression of glucose transporters while sustaining activation of
human tumor samples with active AKT. ENTPD5 hexokinase and phosphofructose kinase-1 (PFK1), thus acceler-
hydrolyzes UDP to UMP to promote protein N-glyco- ating influx and capture of glucose for glycolysis (reviewed by
sylation and folding in ER. Knockdown of ENTPD5 in Vander Heiden et al., 2009). Of interest, in cancer cells, there is
PTEN null cells causes ER stress and loss of growth invariant expression of the embryonic M2 splice version of pyru-
factor receptors. ENTPD5, together with cytidine vate kinase, an enzyme working in the last step of glycolysis,
monophosphate kinase-1 and adenylate kinase-1, instead of a more active M1 splicing isoform expressed in
constitute an ATP hydrolysis cycle that converts most of the adult tissues (Christofk et al., 2008). The combined
ATP to AMP, resulting in a compensatory increase effects of more glucose entering into the glycolysis pathway
in aerobic glycolysis known as the Warburg effect. and slowing down pyruvate kinase activity build up intermediate
metabolites for synthesis of growth-enabling macromolecules.
The growth of PTEN null cells is inhibited both
One noticeable example is the entry of glucose-6-phosphate
in vitro and in mouse xenograft tumor models.
to the pentose shunt pathway to generate ribose for nucleotide
ENTPD5 is therefore an integral part of the PI3K/ synthesis (reviewed by Vander Heiden et al., 2009).
PTEN regulatory loop and a potential target for anti- Another outlet of glucose-6-phosphate is to form UDP-
cancer therapy. glucose, a substrate for protein glycosylation. In mammalian
cells, most secreted proteins and membrane proteins are glyco-
INTRODUCTION sylated at the asparagine (Asn) sites, i.e., N-glycosylated. Of
interest, receptor tyrosine kinases that promote cell growth
Class I phosphatidylinositol 3-kinases (PI3Ks) and lipid phospha- and proliferation, such as the epidermal growth factor receptor,
tase PTEN balance cellular response to growth and survival EGFR, are much more highly N-glycosylated than receptors
signals (reviewed by Engelman et al., 2006). In response to acti- whose functions do not (Lau et al., 2007). Most of the glycosyla-
vation of receptor tyrosine kinases, PI3K phosphorylates phos- tion reactions happen in the Golgi apparatus, with two known
phatidylinositol 4,5-bisphosphate (PIP2) at the 3-OH position of exceptions. One is the dolichol-linked 14 sugar core glycan
the inositol ring to generate phosphatidylinositol 3,4,5-trisphos- (Glc3Man9GlcNAc2) that is synthesized in cytoplasm and ER
phate (PIP3) that recruits and activates serine/threonine kinase membrane before being flipped into the lumen of ER, where it
AKT (Whitman et al., 1988; Franke et al., 1997; Stephens et al., is transferred to the Asn of the nascent polypeptide chain (re-
1998). AKT subsequently activates many downstream targets viewed by Helenius and Aebi, 2004). Another is reglucosylation
for cell growth and survival, including the rapamycin-sensitive in ER after the third and second glucose (Glc) on the core glycan
mTOR complex 1 (mTORC1), which then phosphorylates are trimmed by glycosidase I and II. Trimming and reglucosyla-
p70S6K and translation initiation factor 4E-BP1 to accelerate tion by UDP-glucose:glycoprotein glucosyltransferase (UGGT)
the translational rate, thus accommodating rapid growth (Fingar generate monoglucosylated structures that are recognized by

Cell 143, 711–724, November 24, 2010 ª2010 Elsevier Inc. 711
calnexin/calreticulin, an ER molecular chaperone system for ENTPD5 Is Responsible for the Elevated ATPase Activity
N-glycosylated proteins (reviewed by Ellgaard et al., 1999). in PTEN Knockout Cells
The removal and addition of glucose allow the binding and We decided to purify the ATPase from large-scale cultured PTEN
release of calnexin/calreticulin to and from nascent polypeptide knockout MEFs. We took 800 mg of S-100 from PTEN null MEFs
chains until the proteins are correctly folded and transferred to and put it through five chromatographic steps (Figure 2A). The
Golgi for further glycosylation. If proteins are misfolded beyond ATP hydrolysis activity was measured as in Figure 1D, and the
repair, they are subjected to degradation by the ER-associated active fractions from each column step were pooled, dialyzed,
protein degradation system (ERAD) (reviewed by Fewell et al., and loaded onto the next column. Finally, after a Superdex 200
2001). gel-filtration column, the active fractions were loaded onto
During a study using the embryonic fibroblasts (MEFs) from a 100 ml Mini Q column and the bound protein was eluted with
the PTEN null mice and PTEN heterozygous littermates (Stam- a linear salt gradient. Fractions of 100 ml were collected and as-
bolic et al., 1998), we made a surprising finding that an ER sayed. Shown in Figure 2B, a single ATP hydrolysis peak
UDP hydrolysis enzyme is upregulated by AKT activation. This centered at fraction 6 was observed. When these fractions
enzyme, ENTPD5, seems to mediate many of the observed were analyzed by SDS-PAGE followed by silver staining,
cancer-related phenotypes associated with AKT activation. a protein band just below the 50 kDa molecular weight marker
correlated perfectly with the activity.
RESULTS This protein was excised from the gel and subjected to mass
spectrometry analysis. The enzyme was identified as ectonu-
PTEN Knockout MEFs Have an Elevated Activity that cleoside triphosphate diphosphohydrolase 5, ENTPD5, a mem-
Hydrolyzes ATP to AMP ber of the ENTPD enzyme family known to hydrolyze tri- and/or
As reported previously, the PTEN null MEFs showed elevated diphospshonucleotide to monophosphonucleotide (reviewed
levels of phosphorylated AKT and p70S6 kinase, whereas the by Robson et al., 2006).
total protein level of these two kinases remained the same as To verify that ENTPD5 is indeed the enzyme that caused the
in PTEN heterozygous MEFs (Stambolic et al., 1998) (Figure 1A). higher rate of ATP-to-AMP conversion in PTEN null MEFs, we
We noticed that S-100 cell extracts (prepared after collecting the first did a western blotting analysis of ENTPD5 in these MEFs.
supernatants of 100,000 3 g spin of broken cells) from PTEN null As shown in Figure 2C (bottom), ENTPD5 was only prominently
MEFs had a lower ATP level compared to that from the heterozy- detected in PTEN null extracts, but not in PTEN heterozygous
gous MEFs (Figure 1B, columns 7 and 8). Given that cellular ATP extracts (Figure 2C, lanes 1 and 2). When mouse ENTPD5 was
levels are relatively stable, we reasoned that the difference in exogenously expressed in the PTEN heterozygous MEFs, the
their ATP contents occurred during S-100 preparation, which extracts from these cells showed the ability to hydrolyze ATP
took about 1 hr. Indeed, as shown in Figure 1B, the ATP levels to AMP just like that from PTEN null cells (Figure 2C, lanes
in PTEN null MEFs were only slightly lower than those in the 3–5). Moreover, when ENTPD5 was knocked down in PTEN
heterozygous MEFs if the measurement was carried out immedi- null MEFs with two different siRNA oligos, the ATP-to-AMP
ately after cells were harvested (Figure 1B, columns 1 and 2). conversion was diminished in each case, and a control siRNA
When the broken cell suspension, or supernatants after oligo had no effect (Figure 2C, lanes 6–10).
10,000 3 g spin (S-10), or S-100 were incubated on ice for 1 hr To confirm that the elevated level of ENTPD5 is due to deletion of
before the ATP levels were measured, ATP concentrations in PTEN, we transfected a wild-type PTEN cDNA, or the phospha-
the extracts from PTEN knockout MEFs were much lower than tase active site mutant (PTEN C124S), into PTEN null MEFs.
those from heterozygous MEFs (Figure 1B, columns 3–8). Such Indeed, restoring PTEN expression in these cells lowered phos-
an observation indicated that there was higher ATP hydrolysis phoAKT and diminished ENTPD5 expression, whereas the catalyt-
activity in the PTEN knockout cell extracts. To measure this ically dead mutant PTEN had no effect (Figure 2D, lanes 2 and 3).
activity directly, we incubated a-P32-ATP with the S-100 extracts Consistently, treatment of PTEN null MEFs with a PI3 kinase inhib-
and analyzed the radioactivity using thin layer chromatography. itor also lowered the level of ENTPD5 (Figure 2E, lanes 2 and 3).
As shown in Figure 1C, more radiolabeled ATP was hydrolyzed The upregulation of ENTPD5 in PTEN null cells is at transcrip-
when incubated with the S-100 from PTEN null MEFs, and the tional level. Its mRNA is 6-fold higher in PTEN null MEFs
nucleotide was hydrolyzed all the way to AMP. compared to that in PTEN heterozygous MEFs (Figure S1 avail-
To sort out whether the accelerated ATP hydrolysis was due to able online). The promoter region of ENTPD5 is negatively regu-
a specific activity or a combination of nonspecific ATPases, we lated by the FoxO family of transcription factors (Figure S2),
fractioned the same amount of S-100 extracts from PTEN null which upon phosphorylation by AKT, are displaced from the
and PTEN heterozygous MEFs side by side on a Q Sepharose nucleus into the cytoplasm (Brunet et al., 1999).
ion-exchange column. The fractions from each column run To directly demonstrate the nucleotide hydrolysis activity of
were dialyzed, and ATPase activity was measured by adding ENTPD5, we generated recombinant human ENTPD5 protein
each column fraction to the S-100 from PTEN heterozygous in insect cells using a baculovirus vector and purified the enzyme
MEF, which served as the baseline activity. A single peak of to homogeneity (Figure 3A, right). The purified enzyme was then
elevated ATP-to-AMP activity centered at fractions 11–13 was incubated with ATP, ADP, CTP, CDP, GTP, GDP, UTP, and UDP,
observed in fractionated S-100 from PTEN null MEFs, whereas and the released phosphate was measured. Unexpectedly, the
much less activity was seen in the corresponding fractions purified recombinant ENTPD5 could only hydrolyze UDP and
from PTEN heterozygous MEFs (Figure 1D). GDP (Figure 3A, left).

712 Cell 143, 711–724, November 24, 2010 ª2010 Elsevier Inc.
-

-/-
+/
B

EN
EN
A

PT
PT

Luminescence Intensity
PTEN 300000
*

pAKT 200000

AKT 100000
pP70S6K
0

PTEN+/-

PTEN-/-

PTEN+/-

PTEN-/-

PTEN+/-

PTEN-/-

PTEN+/-

PTEN-/-
P70S6K

Actin

Start

Broken cells

S-10

S-100
C D
AMP 1600
1000
UV Abs. (mAU)

1400 750

NaCl (mM)
1200 500
ADP 1000 250
+/-

800 0
ATP AMP
PTEN+/-

PTEN-/-

ADP

ATP
1600 1000
UV Abs. (mAU)

1400 750

NaCl (mM)
1200 500
1000 250
800 0
-/-

AMP

ADP

ATP

Fraction №
17+18
+/- Start

+/- Flow
-/- Start

-/- Flow

3 4 5 6 7 8 9 10 111213 14 1516

S-100 (+/-)

Figure 1. Identification of ATP Hydrolysis Activity in PTEN Knockout MEF Cells


(A) Total cell extracts from PTEN+/ and PTEN / cells were prepared as described in Experimental Procedures. Aliquots of 10 mg protein were subjected to 10%
SDS-PAGE followed by western analysis of PTEN (asterisk denotes a cross-reactive band), phosphorylated AKT (pAKT), AKT, phosphorylated P70S6 kinase
(pP70S6K), P70S6 kinase, and b-actin.
(B) Cell extracts were prepared from PTEN+/ and PTEN / cells, and at indicated steps of preparation, aliquots of 20 ml samples were incubated on ice for 1 hr
followed by immediate measurement of ATP using a Cell Titer-Glo kit. Error bar represents standard deviation of two independent experiments.
(C) Aliquots of 30 mg of S-100 fractions from PTEN+/ or PTEN / cells were incubated with a-P32-labeled ATP and analyzed by TLC as described in the
Experimental Procedures. Positions for ATP, ADP, or AMP were indicated.
(D) 6 ml each of S-100 from PTEN+/ or PTEN / cells (3.5 mg/ml) was separated by a 1 ml Q Sepharose HP column with a salt gradient elution as indicated.
Fractions of 1 ml were collected and dialyzed overnight at 4 C. 10.5 ml of each fraction was mixed with another 10.5 ml of undialyzed S-100 from PTEN+/ cells
and assayed for ATP hydrolysis activity as in (C). The positions of ATP, ADP, and AMP were indicated. FPLC histograms were presented in top panels.

UMP or GMP Is a Required Cofactor ATP-to-AMP converting activity after dialysis (Figure 3B,
for the ATP Hydrolysis Activity lane 4), and the activity was restored with addition of a small
During our ENTPD5 purification efforts, we noticed that a small molecule fraction prepared by a 10 kDa cutoff filter (Figure 3B,
molecule cofactor was required for the observed ATP-to-AMP lane 6). There was no difference in such a small molecule in
hydrolysis activity. S-100 extracts from PTEN null MEFs lost PTEN heterozygous and PTEN null MEFs (Figure 3B, lanes 6

Cell 143, 711–724, November 24, 2010 ª2010 Elsevier Inc. 713
A B
Start PTEN-/- S-100 127.5mM NaCl
120mM

Fraction № 3 4 5 6 7 8 9 10 11
250KD
SP HP 150KD
100KD
75KD
Q HP 50KD
ENTPD5
37KD

Phenyl HP 25KD
20KD
15KD
10KD
Super-dex 200 AMP

Mini Q

ADP
-/- siRNA ENTPD5 1#
-/- siRNA ENTPD5 2#

ATP
+/- ENTPD5 1#
+/- ENTPD5 2#

C
-/- siRNA GFP

D PTEN-/- MEF E
+/- Vector

PTEN-CD
Treatment DMSO LY
PTEN

Tranfection
Vec

Cell Line +/- -/-


+/-

+/-
-/-

-/-

Lane 1 2 3
Lane 1 2 3 4 5 6 7 8 9 10 Lane 1 2 3
ENTPD5
AMP PTEN

pAKT
pAKT
ADP
AKT
AKT
ATP
ß-Actin
ENTPD5
*
ß-Actin
ENTPD5

Figure 2. Purification and Characterization of ENTPD5


(A) Diagram of the purification scheme for ATP hydrolysis activity from S-100 of PTEN / MEF cells.
(B) Final step of purification. (Top) Aliquots of 30 ml of indicated fractions from the Mini Q column were subjected to 4%–10% gradient SDS-PAGE gels followed by
staining using a silver staining kit from Invitrogen. (Bottom) Aliquots of 3 ml of indicated fractions were incubated with 10.5 ml of undialyzed S-100 from PTEN+/
MEF cells and assayed for ATP hydrolysis activity.
(C) (Lanes 1–5) ATP hydrolysis activity in S-100 from PTEN+/ MEF cells expressing exogenous ENTPD5. PTEN+/ vector or PTEN+/ ENTPD5 1# and 2# (two
individual clones with different expression levels of ENTPD5) were established as described in the Experimental Procedures. Cell lysates (S-100) from indicated
cell lines were prepared, and aliquots of 30 mg were used for ATP hydrolysis assay. (Lanes 6–10) ENTPD5 expression in PTEN / MEFs was knocked down as
described in the Experimental Procedures. The cells were harvested, and S-100 were prepared and normalized for ATP hydrolysis assay. Positions of ATP, ADP,
and AMP are indicated. (Bottom) Aliquots of 10 mg protein of indicated samples were subjected to 10% SDS-PAGE followed by western analysis of ENTPD5.
Asterisk denotes cross-reactive proteins.
(D) PTEN / MEF cells were transfected with 4 mg plasmid DNA containing vector control or cDNA encoding PTEN or PTENcs as indicated. At 24 hr after trans-
fection, cells were harvested and total cell lysates were prepared. Aliquots of 10 mg of protein were loaded onto 10% SDS-PAGE followed by western analysis of
levels of PTEN, AKT, phosphorylated AKT(pAKT), ENTPD5, and b-actin as indicated.
(E) PTEN+/ and PTEN / MEF cells were treated with DMSO or LY294002 (50 mM) for 24 hr. Aliquots of 20 mg total cell extracts were subjected to 10% SDS-
PAGE followed by western analysis using indicated antibodies.
See also Figure S1 and Figure S2.

and 8), and the molecule was also present in S-100 from HeLa (Figure 3C, lanes 1–15). In contrast, thymidine nucleotides
cells (Figure 3B, lane 10), which have a wild-type PTEN. have no activity, whereas CMP only showed a slight activity.
Based on its biochemical properties, we deduced that the To see whether the conversion of UTP/UDP to UMP is neces-
cofactor is a nucleotide. Testing a variety of nucleotides revealed sary for the observed activity, we tested various forms of
that uracil and guanine, either in tri-, di-, or monophosphate nonhydrolyzable uracil, including UTPgS, UTPaS, and UMP-
form, substituted the small molecule fraction from cells PNP (Figure 3D). All of these nucleotides worked except UTPaS,

714 Cell 143, 711–724, November 24, 2010 ª2010 Elsevier Inc.
A 16

Normalized [Pi] Fold Increase


Reaction 0hr
12
Reaction 2hr

0
ATP ADP CTP CDP GTP GDP UTP UDP
Substrates

B C Tri-phosphate Di-phosphate Mono-phosphate

15.625

15.625

15.625
1000

1000

1000
Small Molecule from +/- -/- -/- +/- Hela

62.5

62.5

62.5
250

250

250
Dialysis Conc.(μM)

4
S-100 +/- -/- +/- -/- +/- -/- +/- -/- +/- -/- Lane 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15
Lane 1 2 3 4 5 6 7 8 9 10
Uracil AMP
AMP
Guanine AMP

ADP
Cytosine AMP
ATP
Thymidine AMP

D
1000

1000

1000
1000

62.5
62.5

62.5

62.5
250
250

250

250
16
16

16

16

Conc.(μM)
4
4

Small Molecule - UMP UTPγS UTPαS PNP-UMP


Lane 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21

AMP

ADP

ATP

Figure 3. Small Molecule Requirement for ENTPD5-Mediated ATP Hydrolysis


(A) (Right) the recombinant human ENTPD5 was generated and purified as described in the Experimental Procedures. An aliquot of 120 ng recombinant ENTPD5
was subjected to SDS-PAGE followed by Coomassie brilliant blue staining. Arrows indicate recombinant ENTPD5. (Left) The nucleotide hydrolysis reactions were
carried out in triplicate by mixing 0.1 mg/ml ENTPD5 with 50 mM indicated nucleotides. After 2 hr incubation at 30 C, released free phosphate was measured by
malachite green assay as described in the Experimental Procedures. Data shown are representative of three independent experiments. Error bars indicate SEM.
(B) Small molecule (<10 kDa) was extracted from either PTEN+/ or PTEN / MEF cells or HeLa S3 cell lysates (S-100 fractions) as described in the Experimental
Procedures. Aliquots of 10.5 ml undialyzed cell lysates (lane 1 and 2) or dialyzed cell lysates (lane 3–10) from PTEN+/ (lanes 1, 3, 5, 7, and 9) or PTEN / (lanes 2, 4,
6, 8, 10) MEFs (3.5 mg/ml) were mixed with another 10.5 ml buffer A (lane 1 to 4) or small molecule recovered from PTEN+/ (lanes 5 and 8), PTEN / cells (lanes 6
and 7), or from HeLa S3 cells (lanes 9 and 10) and were assayed for ATP hydrolysis activity. The positions of ATP, ADP, and AMP were indicated.
(C) Aliquots of 10.5 ml dialyzed S-100 from PTEN / MEF cells (3.5 mg/ml) were incubated in the presence of indicated final concentration of UTP, UDP, and UMP;
or GTP, GDP, and GMP; or CTP, CDP, and CMP; or TTP, TDP, and TMP as indicated at 30 C with a-P32-labeled ATP in a total volume of 30 ml at 30 C for 1 hr
followed by TLC to resolve radioactive adenosine nucleotides. Position of AMP on TLC plate is indicated.
(D) Aliquots of dialyzed S-100 prepared from PTEN / MEF cells were mixed with buffer A (lane 1) or indicated final concentration of indicated nucleotides and
assayed for ATP hydrolysis activity.
See also Figure S3.

which could not be hydrolyzed to UMP, indicating that the heterozygous MEFs, we realized that there must be more factors
conversion to UMP is critical for this cofactor to function. The in the S-100, which are also required to hydrolyze ATP to AMP.
same holds true for guanine nucleotides (Figure S3). These factors presented in cells regardless of their PTEN status.
For example, when we added purified, recombinant ENTPD5
UMP, ENTPD5, UMP/CMP Kinase-1, and Adenylate and UMP to the dialyzed S-100 from large-scale cultured HeLa
Kinase-1 Constitute an ATP-to-AMP Hydrolysis Cycle cells, the ATP-to-AMP hydrolysis was reconstituted (Figure 4A,
Based on the facts that purified ENTPD5 is unable to hydrolyze lanes 1–6). This observation made purification of these factors
ATP directly and the assay also contained S-100 from PTEN easier because HeLa cells can be grown in large quantity in

Cell 143, 711–724, November 24, 2010 ª2010 Elsevier Inc. 715
A C
Hela S-100 Q-FL Q-30 Q-FL + Q-30
UMP - + - - + + - - + + - - + + - - + + Fraction № ST 13 14 15 16 17 18 19 20 21 22 23 24 25
rENTPD5 + + - + - + - + - + - + - + - + - +
Lane 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 AMP

AMP
ADP

ADP ATP

ATP
AK1

B 120 mM NaCl D
40 mM
Fraction № 4 5 6 7 8 9 10 11 12
UMP + + + + + + + - ENTPD5

PNGase F
Start Hela S-100 CMPK1 - - + - + + + +

CMPK1
75KD ENTPD5 - + - + - + + +

AK1
N/T
50KD AK1 + - - + + - + +
Q HP
37KD Lane 1 2 3 4 5 6 7 8 9 10 11 12
25KD
SP HP 20KD Glycosylated
AMP ENTPD5
UnGlycosylated
Heparin HP AMP ENTPD5
PNGaseF
ADP
AK1
Super-dex 200 CMPK1
ADP ATP

Mini Q ATP

Figure 4. Reconstitution of ENTPD5-Mediated ATP Hydrolysis


(A) HeLa S3 cell S-100 was fractionated by Q Sepharose column to two fractions (Q-FL [flowthrough] and Q-30). Q-30 represents fraction eluted with 300 mM
NaCl. Aliquots of 15 ml buffer A (lane 1 and 2), or dialyzed HeLa cell S-100 (lanes 3–6), or Q-FL (lanes 7–10), or Q-30 (lanes 11–14), or Q-FL combined with Q-30
(7.5 ml each) (lanes 15–18) were mixed with (lanes 1, 2, 4, 6, 8, 10, 12, 14, 16, and 18) or without (lanes 3, 5, 7, 9, 11, 13, 15, and 17) ENTPD5 in the presence (lanes 2,
5, 6, 9, 10, 13, 14, 17, and 18) or absence (lanes 1, 3, 4, 7, 8, 11, 12, 15, and 16) of 100 mM UMP and assayed for ATP hydrolysis activity.
(B) (Left) Diagram of the purification scheme for the required factor in Q-30. (Right) Final step of purification of CMPK1. (Top) Aliquots of 60 ml indicated Mini Q
fractions that were subjected to 4%–10% gradient SDS-PAGE followed by silver staining. Arrow indicates the protein band correlated with ATP hydrolysis
activity. (Bottom) Aliquots of 5 ml indicated fractions that were mixed with 15 ml of dialyzed Q-FL fraction in the presence of 100 mM UMP and 18 ng recombinant
ENTPD5 and were assayed for ATP hydrolysis activity.
(C) 3 ml of the Q-FL fraction was concentrated to 600 ml with a spin column and analyzed on a Supdex-200 column (10/30). Fractions of 1 ml were collected, and
aliquots of 7.5 ml of indicated fractions were combined with 7.5 ml dialyzed Q-30 fraction, 100 mM UMP, and 18 ng recombinant ENTPD5 and were assayed for ATP
hydrolysis activity. Positions of radioactive ATP, ADP, and AMP are indicated. (Bottom) Aliquots of 10 ml of indicated fractions were subjected to 10% SDS-PAGE
followed by western blotting analysis using an antibody against human adenylate kinase 1 (AK1).
(D) (Left) Aliquots of recombinant AK1 (lane 1), ENTPD5 (lane 2), and CMPK1 (lane 3) (final concentration, 1 mg/ml) were incubated alone or were sequentially
combined as indicated (lane 4 to 8) in the presence (lane 1 to 7) or absence (lane 8) of UMP (100 mM) for ATP hydrolysis activity. Position of ATP, ADP, or
AMP was indicated. (Right) Aliquots of 10 mg recombinant AK1 (lane 9), ENTPD5 (lane 10) or ENTPD5 pretreat with PNGase F (NEB) (50 units/mg ENTPD5)
(lane 11), and CMPK1 (lane 12) were subjected to 10% SDS-PAGE followed by Coomassie brilliant blue staining.

suspension. To identify these factors, we fractionated HeLa cell HeLa S-100 onto four sequential column chromatographic steps
S-100, using a Q Sepharose column, and collected both the and finally onto a Mini Q column (Figure 4B, left). The activity
flowthrough (Q-FL) and column-bound fractions eluted with was eluted from this column with a linear salt gradient from 40
300 mM NaCl (Q-30). Neither fraction alone was able to hydro- to 120 mM NaCl, and fractions eluted from the column were as-
lyze ATP to AMP, although the Q-30 fraction, when ENTPD5 sayed in the presence of recombinant ENTPD5, UMP, and the
and UMP were present, hydrolyzed ATP to ADP (Figure 4A, lanes Q-FL fraction (Figure 4B, right-bottom). A peak of activity was
13 and 14). When both the Q-FL and Q-30 fractions were observed at fractions 8–10. The same fractions were subjected
included, the ATP-to-AMP activity was fully reconstituted (Fig- to SDS-PAGE followed by silver staining, and two protein bands
ure 4A, lane 18). close to 37 and 20 kDa markers correlated perfectly with activity
We purified the activity present in the Q-30 fraction. The (Figure 4B, right-top). Both bands were identified by mass
activity present in the Q-30 fraction was purified by subjecting spectrometry as human UMP/CMP kinase-1 (CMPK1).

716 Cell 143, 711–724, November 24, 2010 ª2010 Elsevier Inc.
The identification of UMP/CMP kinase in the Q-30 fraction Knockdown of ENTPD5 Causes ER Stress and Growth
shed light on why UMP is a cofactor for the ATPase activity Inhibition
and how ENTPD5 plus this enzyme generates ADP from ATP. Because cells with an activated PI3K/AKT pathway increase
In this reaction, UMP is phosphorylated into UDP by CMPK1 their cellular protein translation level, cells need to evolve a corre-
and ATP, generating ADP. UDP is subsequently hydrolyzed by sponding system in ER to accommodate the high demand for
ENTPD5 to UMP, completing the cycle with net conversion of protein folding process. It is possible that cells may do so by up-
ATP to ADP. regulating ENTPD5 to increase the conversion of UDP to UMP in
With this knowledge, we then made an educated guess that the ER, thereby promoting N-glycosylation and folding. Thus,
third protein factor present in the Q flowthrough fraction should reducing the level of ENTPD5 in cells with active AKT should
be an adenylate kinase, which converts two ADP into one ATP induce ER stress. In addition, because many growth-promoting
and AMP, causing the ATP-to-AMP conversion seen in PTEN cell membrane receptors are highly N-glycosylated, loss of
null cell extracts. To confirm this, we took the Q flowthrough frac- function of ENTPD5 could affect their folding process, resulting
tion and subjected it to a gel-filtration column and collected the in their reduction and, subsequently, cell growth arrest. To test
fractions eluted from the column to assay for ATP-to-AMP hydro- this hypothesis, we engineered several cell lines based on
lysis in the presence of UMP, purified recombinant ENTPD5, and the PTEN null MEFs in which the expression of ENTPD5 could
the Q-30 fraction that contains CMPK1. An ATP-to-AMP activity be knocked down with the addition of doxcycline (Dox),
peak centered at fractions 17 and 18 was observed (Figure 4C, which turned on a Tet-suppressor-controlled shRNA-targeting
top). When these factions were subjected to western blotting ENTPD5. The results from a representative cell line were shown
analysis using an antibody against adenylate kinase-1 (AK1), in Figure 5C. Comparing to PTEN null MEFs expressing GFP
the detected western blotting band correlated perfectly with the shRNA, addition of Dox to the culture media resulted in success-
activity peak (Figure 4C, bottom). The correlation was maintained ful knockdown of ENTPD5 expression in these cells. As a result,
with additional chromatographic steps (data not shown). an ER stress marker, GRP78/BiP, was induced, and cellular
We subsequently generated recombinant CMPK1 and AK1 in N-glycosylation level, as measured by PHA blotting, was down
bacteria and purified them to homogeneity (Figure 4D, lanes 9 (Figure 5C, lanes 5–8). Of interest, the levels of receptor tyrosine
and 12). Purified recombinant ENTPD5 expressed in insect cells kinases, including EGFR, Her-2/Erb-2, and type I insulin-like
runs as a triplet on an SDS-PAGE gel that could be shifted down growth factor receptor (IGF-IR) b, were significantly decreased
to a doublet after treatment by PNGase F, indicating that after ENTPD5 knockdown.
ENTPD-5 is glycosylated (Figure 4D, lanes 10 and 11). To confirm that the above-mentioned cellular effects after
These purified recombinant proteins allowed us to reconsti- ENTPD5-targeting shRNA expression were specific, we intro-
tute the ATP-to-AMP hydrolysis cycle. Only when all three duced into these cells a cDNA encoding ENTPD5 with silent
enzymes and UMP were present, efficient ATP-to-AMP conver- mutations in the shRNA target sequence. In these cells, although
sion was observed (Figure 4D, lanes 1–8). the endogenous ENTPD5 was still knocked down after addition
of Dox (Figure 5D, lanes 2, 4, and 6), the expression of an
ENTPD5 Is an ER Enzyme shRNA-resistant wild-type transgene (three flag tags were fused
After purification and identification of ENTPD5 from PTEN null to ENTPD5 coding sequence so it migrated higher) led to
cells, we realized that ENTPD5 is identical to a previously purified complete reversal of BiP induction, lowered glycosylation, and
ER UDPase (Trombetta and Helenius, 1999). Although we iden- downregulation of these growth factor receptors (Figure 5D,
tified and purified ENTPD5 from the S-100, the enzyme most lane 4). In contrast, introducing an E171A mutant that abolishes
likely fractionated there as a result of broken ER from physical UDP hydrolysis activity of ENTPD5 was not able to rescue these
shearing during the cell-breaking process. When we expressed phenotypes (Figure 5D, lane 6). In addition to BiP, another ER
an ENTPD5-GFP fusion protein in cells, the GFP signal was co- stress marker, CHOP, was also induced when ENTPD5 was
localized with the coexpressed ER-DsRed marker (Figure 5A). knocked down (Figure 5D).
The ER location of ENTPD5 and its preferred specificity for Consistent with the loss of growth factor receptors after
UDP suggested that ENTPD5 functions in the process of reglu- ENTPD5 knockdown, cell growth was also dramatically attenu-
cosylation catalyzed by UGGT for calnexin/calreticulin-mediated ated. As shown in Figure 5E, when ENTPD5 in PTEN null MEFs
protein folding (Trombetta and Parodi, 2003). In the process, was knocked down after addition of Dox, very few colonies
UDP is generated after the conjugated glucose gets transferred grew on the culture dish after 10 days, although the same
to the glycosidase I/II trimmed core glycan on N-glycosylated number of cells was plated initially, and they were cultured under
proteins. UDP-glucose is made in cytosol and transported into the same condition (Figure 5E, left row). The growth inhibition
ER through the UDP-sugar transporter, which is an antiporter was rescued when the shRNA-resistant ENTPD5 cDNA was ex-
that must exchange out one molecule of UMP for each UDP pressed (Figure 5E, middle row), whereas the inhibition was
sugar conjugate imported into the ER (Hirschberg et al., 1998). exacerbated if an enzymatic dead mutant of ENTPD5 was ex-
UDP therefore needs to be hydrolyzed to UMP to prevent end pressed instead (Figure 5E, right row).
product feedback inhibition of UGGT, as well as to serve as
a substrate for the antiporter (Trombetta and Helenius, 1999). ENTPD5 Promotes Aerobic Glycolysis
UMP is phosphorylated back to UDP by CMPK1 in the cytosol, One implication of elevated ENTPD5 expression is that a
and the generated ADP is converted to ATP and AMP by AK1 significant percentage of cellular ATP is consumed through
(diagramed in Figure 5B). the ENTPD5/CMPK1/AK1 enzyme cascade. To maintain the

Cell 143, 711–724, November 24, 2010 ª2010 Elsevier Inc. 717
A C Cell Line
PTEN-/- sh RNA
GFP ENTPD5
Time 2d 4d 2d 4d
Dox - + - + - + - +
Lane 1 2 3 4 5 6 7 8

ENTPD5
ENTPD5-GFP DsRed Mer ge
GRP78/BiP

PHA Blot
*

EGFR
ENTPD5-GFP ER-DsRed Merge
Her-2/ErbB-2

B IGFRß
1/2 AK1 1/2
ß-Actin
AMP
ATP Input 2xATP 2xADP AMP Output +
2x Pi

CMPK1
Cytosol D PTEN-/- MEF
Cell Line
ER Lumen sh RNA ENTPD5

sr ENTPD5

sr ENTPD5
2xUMP 2xUDP

Vec

E171A
ENTPD5 Rescue
2xPi

Dox - + - + - +
UMP
Lane 1 2 3 4 5 6
UDP- Glucose Phosphate
sr ENTPD5-3Flag
Antiporter ENTPD5
Cytosol
GRP78/BiP

ENTPD5 CHOP
ER Lumen Phosphate
PHA Blot

Pi Transporter *
P U
P P U
P P U
EGFR

Folding Her-2/ErbB-2
UGGT
Glycoprotein
IGFRß

ß-Actin

N-acetylglucosamine E
manose U uridine Rescue Vec sr ENTPD5 sr ENTPD5 E171A
Cell line PTEN-/- MEF shRNA ENTPD5
glucose P
P phosphate
- Dox
+ Dox

Figure 5. Biological Function of ENTPD5 in PTEN /


MEF Cells
(A) PTEN / MEF cells were cotransfected with mouse ENTPD5-GFP and free DsRed or with ENTPD5-GFP and ER-localized DsRed (ER-DsRed). ENTPD5-GFP
colocalized with ER-DsRed (bottom row), but no obvious codistribution with free DsRed was observed (top row). Scale bars, 10 mm.
(B) Working model for ENTPD5. See the text for details.
(C) PTEN / MEF cells with doxycycline (Dox)-inducible expression of shRNA-targeting ENTPD5 was generated as described in the Experimental Procedures.
After 2 or 4 days induction with Dox (0.125 mg/ml), cells were harvested and total cell lysates were prepared as described in the Extended Experimental Proce-
dures. Aliquots of 10 mg protein were subjected to SDS-PAGE followed by western blotting analysis using the indicated antibodies. Glycosylation was visualized
by PHA blot as indicated. Asterisk denotes decreased glycosylated proteins.

718 Cell 143, 711–724, November 24, 2010 ª2010 Elsevier Inc.
intracellular ATP level, the extra ATP consumed should come AKT activity (Figure 5C and Figure S5), which stimulates the
from either increased oxidative phosphorylation or glycolysis. glucose transporter activity on cell surface (Kohn et al., 1996;
We therefore tested both by measuring oxygen consumption Plas et al., 2001). In addition, knockdown of ENTPD5 may reduce
and lactate production, respectively. Consistent with previous the production of ADP/AMP, which allosterically activate glycol-
reports, there was not much difference in the respiration rate of ysis enzymes such as phosphofructose kinase (PFK) (Gevers
PTEN null and PTEN heterozygous MEFs (Figure S4), but and Krebs, 1966). To distinguish these possibilities, cellular
PTEN null cells showed 40% higher lactate production in their fructose-6-phosphate and fructose-1,6-bisphosphate were
cultured medium (Figure 6A, columns 4 and 5). When ENTPD5 measured using LC-Mass. As shown in Figure 6E, the former
was ectopically expressed in two PTEN heterozygous MEF lines, was lowered by 20% after ENTPD5 knockdown (Figure 6E,
lactate production was increased, and the level of increase left), whereas the latter dropped by 60% (Figure 6E, right).
correlated with that of ENTPD5 expression (Figure 6A, columns These results suggested that ENTPD5 indeed affects glucose
2 and 3). Consistently, when ENTPD5 was knocked down with influx to cells, but its major impact on glycolysis is to directly
addition of Dox as in Figure 5D, lactate production was signifi- activate glycolysis enzymes such as PFK by hydrolyzing ATP.
cantly decreased (Figure 6B, columns 1 and 2). In PTEN null
MEFs harboring Dox-inducible ENTPD5-targeting shRNA that ENTPD5 Expression Correlates with AKT Activation in
also expressed shRNA-resistant ENTPD5 transgene, addition Human Cancer Cell Lines and Primary Tumor Samples
of Dox did not result in a decrease in lactate production, and PTEN mutation and AKT activation are common features for
the basal lactate production also became higher, correlated human cancer. To check whether what was observed in PTEN
with the higher than endogenous expression of the transgene null MEFs is also true for human cancer cells, we screened
(Figure 6B, columns 3 and 4, and Figure 5D). In contrast, when a panel of human cancer cell lines for the expression of PTEN,
the catalytic site mutant ENTPD5 transgene was expressed to activated AKT, and ENTPD5. As shown in Figure 7A, AKT activa-
the similar level, lactate production still reduced with the addition tion was seen in human prostate cancer lines C42 and LNCaP
of Dox (Figure 6B, columns 5 and 6). cells, and in these two cell lines, elevated ENTPD5 expression
If the ATP hydrolysis cycle initiated by ENTPD5 discovered was also observed.
in vitro is operational in cells, and the extra ATP consumed by We also examined ENTPD5 expression and AKT activation in
a higher level of ENTPD5 is compensated by increased glycol- primary human tumor samples by staining two adjacent sections
ysis, glucose starvation of these cells should result in much from a formalin-fixed, paraffin-embedded human primary pros-
faster decrease of intracellular ATP level compared to cells tate cancer sample with rabbit monoclonal antibodies against
with lower ENTPD5 expression. Indeed, when intracellular ATP human ENTPD5 and phosphoAKT, respectively. The specificity
concentrations in PTEN heterozygous and PTEN null MEFs of this anti-ENTPD5 antibody was verified by western blotting
were measured after glucose withdrawal from the culture media, analysis using LNCaP cell lines with or without their ENTPD5
that in PTEN null MEFs decreased to about half of the original knocked down (Figure S6A). The staining intensity for ENTPD5
level within the first hour, while there was little change of ATP in tumor was significantly greater compared with adjacent normal
in PTEN-heterozygous MEF within 2 hr (Figure 6C). tissue and was correlated with pAKT staining (Figure 7B). Out of
To confirm that the faster ATP level dropping in PTEN null cells 10 samples from patients between age 57 and 76, only one tumor
was due to higher expression of ENTPD5, the same set of MEFs sample from a 57-year-old patient and another sample collected
as in Figure 6B was subjected to glucose starvation after from a patient who had just gone through therapy did not show
ENTPD5 was knocked down with the addition of Dox. Knock- strong ENTPD5 staining, and the same tumors were also negative
down of ENTPD5 for 2 days in PTEN null MEFS caused the total for pAKT (Figure S6B2 and Figure S6B10). The remaining eight
cellular ATP level to decrease (Figure 6D, columns 3 and 4). samples all showed greater tumor staining of pAKT and ENTPD5
However, the ATP level did not decrease further after glucose (Figure 7B and Figures S6B3–S6B9).
starvation for 1 hr, whereas cells without Dox treatment Because microarray data of many tumors are publicly avail-
consumed 50% of original ATP during this period (Figure 6D, able, we also analyzed a group of recently publicized microarray
columns 1 and 2). The MEFs expressing the shRNA-resistant data from human prostate cancer samples (Bermudo et al.,
ENTPD5 did not lower their ATP level with Dox treatment, but 2008). We found that ENTPD5 is highly expressed in all 20 tumor
their ATP levels were even more drastically lowered after glucose samples compared to normal prostate epithelium cells (Fig-
starvation, possibly due to ENTPD5 overexpression (Figure 6D, ure S7). In addition, after clustering all gene expression profiles
columns 5–8). In contrast, cells expressing a similar level of the from prostate tumor microarray data using SOM (self-organiza-
catalytic dead mutant ENTPD5 behaved the same as cells tion method), we identified dozens of genes associated with
without transgene expression (Figure 6D, columns 9–12). AKT activation, including Her-3, PI3KCB, Ras, S6 kinase,
The observed decrease of glycolysis after ENTPD5 knock- CD36, IL8, EGF, osteropontin, and FoxO1, which are signifi-
down could be due to lowered tyrosine kinases receptors and cantly coregulated with ENTPD5 (Figure S7).

(D) Rescue cell lines with expression of shRNA-resistant wild-type or catalytic dead mutant (E171A) ENTPD-5 were established as described in the Extended
Experiments Procedures. Same as in (C), after 2 days culture, cells were harvested, and total cell lysates (10 mg/lane) were subjected to SDS-PAGE followed
by western analysis as indicated. Glycosylation was visualized by PHA blot analysis.
(E) Rescue cell line was plated at density of 5 3 104/100 mm dish and treated with Dox as in (C). Cell medium was changed each 3 days. After 10 days culture, the
plates were stained by methylene blue.

Cell 143, 711–724, November 24, 2010 ª2010 Elsevier Inc. 719
A 800 B 1200

lactate (nmol/mg protein)

lactate (nmol/mg protein)


600
800
400

200
400

0
ENTPD5 Flag
ENTPD5
0

ß-Actin Dox - + - + - +

Cell Line
Rescue Vec sr ENTPD5 sr ENTPD5
PTEN+/- -/- E171A
Cell Line PTEN-/- MEF shRNA ENTPD5
Stable transfected with Vec ENTPD5-Flag

Clone № 1# 2# D 12

10
C
ATP (nmol/mg protein)

8
10

6
ATP (nmol/mg protein)

8
4
6
2
4
0

2 sr ENTPD5 3Flag
ENTPD5
0 ß-Actin

- Glucose 0 1h 2h 0 1h 2h
- Glucose - + - + - + - + - + - +
Cell Line PTEN+/- PTEN-/-
Dox - + - + - +

Rescue Vec sr ENTPD5 sr ENTPD5 E171A

Cell Line PTEN-/- MEF shRNA ENTPD5


E
Relative Fructose-1, 6-2P Content

1.2 1.2
Relative Fructose-6-P Content

F-6-P F-1,6-2P
1.0 1.0

0.8 0.8

0.6 0.6

0.4 0.4

0.2 0.2

0 0

Dox - + Dox - +

PTEN-/- MEF PTEN-/- MEF


shRNA ENTPD5 shRNA ENTPD5

Figure 6. ENTPD5 Promotes ATP Hydrolysis and Glycolysis In Vivo


(A) Lactate in the culture media of PTEN+/ and PTEN / MEF cells (columns 4 and 5) as well as PTEN+/ MEF clones stably transfected with vector control
(column 1) or Flag-tagged mouse ENTPD5 (column 2, clone 1 and column 3, clone 2, as used in Figure 2C) was measured as described in the Extended
Experimental Procedures, and the value was normalized to total protein amount.
(B) Lactate in the culture media of ENTPD5 knockdown and rescue cell lines was measured as in (A) except pretreatment with or without Dox for 2 days.
(C) PTEN+/ and PTEN / MEF were deprived of glucose for indicated time periods, and intracellular ATP was determined as described in Extended Experimental
procedures.
(D) The intracellular ATP was measured 1 hr after glucose starvation on ENTPD5 knockdown and rescue cell lines as in (B).
(E) ENTPD5 was knocked down as in (B), and the intracellular fructose-6-phosphate (left) and fructose-1,6-bisphosphate (right) were separated and quantified by
HPLC mass spectrometry (ABI 3200 Q TRAP LC/MS/MS Systems). The relative amount of metabolite is normalized to total ion count (TIC). All experiments were
repeated at least two times, and the error bar represents standard deviation.
See also Figure S4 and Figure S5.

ENTPD5 Is Important for Cancer Cell Growth be induced by Dox. In these cells, knockdown of ENTPD5 by
To verify the functional significance of ENTPD5 expression in adding Dox to the culture media also lowered N-glycosylation
human cancer cells, we generated a cell line from the original (Figure 7C, comparing lanes 7 and 8, 9 and 10, and 11 and 12)
LNCaP cells in which an shRNA against human ENTPD5 could and induced BiP expression (Figure 7C, lanes 8, 10, and 12).

720 Cell 143, 711–724, November 24, 2010 ª2010 Elsevier Inc.
These phenotypes were rescued by the expression of a wild- gulated in PTEN null cells (Figure S1). The mRNA of an extracel-
type shRNA-resistant ENTPD5 transgene, but not by the active luar ENTPDase, ENTPD2, although expressed at a much lower
site mutant ENTPD5 (Figure 7C, lanes 13–24). Several growth level than ENTPD5, was also elevated in PTEN null cells (Fig-
factor receptors were also checked in these cell lines after Dox ure S1). The significance of such is unknown.
treatment. As shown in Figure 7D, EGFR and Her2/ErbB-2
were significantly down, and IGFRb was slightly down (Figure 7D, ENTPD5 Contributes to Warburg Effect
lanes 1–4). They were restored to the normal level by the expres- One of the surprising findings reported here is how quickly ATP
sion of shRNA-resistant ENTPD5 transgene (Figure 7D, lanes 5 can be consumed as a result of ENTPD5 upregulation. One
and 6), but not the active site mutant (Figure 7D, lanes 7 and naturally raised question is where the extra consumed ATP
8). Consistently, when the cell number was measured after comes from. After measuring both oxygen consumption and
4 day knockdown of ENTPD5, only about half of LNCaP cells lactate generation, we found that the lactate production was
were there, compared to a control knockdown cell line, and elevated in these PTEN null cells, whereas oxygen consumption
the defect was rescued by expression of wild-type ENTPD5 did not change (Figure 6A and Figure S4). When ENTPD5 was
transgene, but not active site mutant (Figure 7E). knocked down, higher lactate production returned to normal
To test whether knocking down ENTPD5 in LNCaP cells also (Figure 6B). Moreover, simply ectopically expressing ENTPD5
has an effect on their growth in vivo, we implanted the LNCaP in PTEN heterozygous MEF elevated their lactate production
cells bearing a Dox-inducible shRNA targeting ENTPD5 in matri- (Figure 6A). These results indicate that ENTPD5 is a critical player
gel in nude mice. As a control, LNCaP cells with a Dox-inducible in causing the Warburg effect, i.e., elevated lactate production
shRNA targeting GFP were also implanted. After the xenograft under aerobic conditions, in these PTEN null cells.
tumors reached the size of 500 mm3, a cohort of seven mice In addition to being part of the activation loop for AKT that
were fed with Dox-containing water. The level of ENTPD5 in promotes glucose uptake into cells (Kohn et al., 1996; Plas
these tumors was measured after 6 weeks. Compared with et al., 2001), a major effect of ENTPD5 on glycolysis might be
mice fed with normal water, the ENTPD5 levels in ENTPD5-tar- its ability to generate ADP/AMP through the aid of CMPK1 and
geting shRNA containing tumors from mice fed with Dox-con- AK1. Elevated AMP levels (and to a lesser extent, ADP) activate
taining water were significantly lower except in one mouse phosphofructokinase and inhibit fructose diphosphatase to drive
(Figure 7F). Whereas ENTPD5-targeting shRNA containing glycolysis and prevent gluconeogenesis, resulting in higher
tumors in mice fed with normal water continued to grow, the lactate production (Gevers and Krebs, 1966). Consistently, the
tumors in mice fed with Dox-containing water shrank (Figure 7G). fructose-1,6-bisphosphate level dropped to a much lower level
Amazingly, when these tumor samples were analyzed under than that of fructose-6-phosphate when the ENTPD5 in PTEN
a microscope after fixing and staining with hematoxylin and null cells was knocked down (Figure 6E).
eosin, there were very few tumor cells left in the matrigel in In addition to UDP, ENTPD5 also use GDP as a substrate and
tumors grown in Dox-fed mice, whereas in mice fed with normal hydrolyze it to GMP (Figure 3A and Figure S3). It is interesting to
water, the matrigel was filled with tumor cells (Figure 7H). The note that GDP-conjugated sugars are another group of major
GFP shRNA-containing tumors did not respond to Dox treatment substrates for glycosylation. The significance of hydrolyzing
and continued to grow during the period of experiment. GDP by ENTPD5 is not clear because it is believed that GDP
sugars are transferred to proteins in the Golgi.
DISCUSSION
ENTPD5 Is Potentially an Anticancer Target
ENTPD5 Is an Important Link in the PI3K/PTEN The current study highlighted ENTPD5 as a critical link in the
Signaling Loop PI3K/PTEN pathway that promotes cell growth and survival,
The experimental data reported here identify ENTPD5, an ER a pathway that is often activated in cancer cells. We saw good
UDPase, as an important link in the PI3K/PTEN/AKT signaling correlation between ENTPD5 expression and AKT activation in
loop. We reason that ENTPD5 upregulation is important for both cultured prostate cancer cell lines and primary human pros-
AKT-activated cells to cope with elevated translational activity tate carcinoma samples (Figures 7A and 7B and Figures S6 and
that generates more nascent polypeptide chains destined for S7). Therefore, inhibition of this enzyme, similar to knockdown,
the ER. can potentially generate benefits for anticancer activity. It should
ENTPD5 is a member of the ectonucleoside triphosphate induce more severe ER stress in cancer cells with active AKT due
diphosphohydrolase family, which consists of seven other to higher protein traffic through the secretory pathway. It may
members (reviewed by Robson et al., 2006). ENTPD 1–3 cause synthetic lethality in these cells, which otherwise maintain
(CD39, CD39L1, and CD39L3) are typical ectoenzymes, whereas survival advantage and resistance to common anticancer drugs.
the other five members have a predominant intracellular localiza- It will also lower many growth factor receptors on the cell surface
tion including ER, Golgi apparatus, and lysosomal/auto- due to their high N-glycosylation nature, a phenomenon that may
phagic vacuoles. The functions of these organelle-associated reflect the evolutionary connection between fast growth and
ENTPDases are still largely unexplored, but judging by their loca- nutrient availability in mammalian cells (Lau et al., 2007). Among
tion and substrate preference, it would not be surprising if they all such receptors, EGFR, Her2/ErB2, and IGFR levels were down
turn out to regulate protein glycosylation. after ENTPD5 knockdown (Figure 5 and Figure 7), whereas
Among members of this group of enzymes, however, ENTPD5 a nongrowth-promoting TGFb receptor did not change (data
is the only intracellular ENTPDase that is transcriptionally upre- not shown).

Cell 143, 711–724, November 24, 2010 ª2010 Elsevier Inc. 721
B ENTPD5 pAKT
A
Prostate Breast

MBA231
SKBR-3
LAPC4

LNCaP

MCF-7
DU145

T47-D
PC3

C42
PTEN
a b
pAKT

ENTPD5
AKT

ß-Actin c d

sr ENTPD5
sr ENTPD5
C D

None

E172A
Rescue None sr ENTPD5 sr ENTPD5 E172A Rescue
Cell Line LNCaP shRNA GFP LNCaP shRNA ENTPD5
Time 2d 4d 6d 2d 4d 6d 2d 4d 6d 2d 4d 6d
Cell Line (LNCaP shRNA) GFP ENTPD5
Dox - + - + - + - + - + - + - + - + - + - + - + - +
Dox- + - + - + - +
Lane 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24
Lane 1 2 3 4 5 6 7 8
sr ENTPD5-3Flag
ENTPD5

GRP78/BiP sr ENTPD5-3Flag
ENTPD5
PHA Blot

*
EGFR

Her-2/ErbB-2
ß-Actin
IGFRß

E ß-Actin
120
100
F
inhibition (%)
Cell growth

80
LNCaP shRNA ENTPD5 tumors
60
-Dox +Dox
40

20 ENTPD5
0
Lane Ponceau S
1 2 3 4 5 6 7 8
Dox - -
+ - + - + +
LNCaP shRNA GFP ENTPD5
sr ENTPD5

sr ENTPD5
None

E172A

Rescue H

G
LNCaP sh RNA ENTPD5 tumors
Tunor size change after Dox (%)

190%
LNCaPshRNA GFP -Dox
170% LNCaPshRNA GFP +Dox
LNCaPshRNA ENTPD5 -Dox
LNCaPshRNA ENTPD5 +Dox - DOX - DOX
150%
130% 500um 100um

110% *
90%

70%
50%
1 2 3 4 5 6 + DOX + DOX
Weeks after Dox

Figure 7. Knockdown of ENTPD5 in LNCaP Cells Decreases Glycosylation, Expression of Cell Surface Receptors, and Tumor Progression
(A) Aliquots of 20 mg of total cell extracts from indicated cell lines were subjected to 10% SDS-PAGE followed by western blotting analysis using antibodies as
indicated.
(B) Immunohistochemical staining for ENTPD5 (a and c) and pAKT (b and d) in adjacent sections of a human prostatic carcinoma sample (a/b 103 and c/d 203
lenses). Scale bar, 200 mM (a and b); 100 mM (c and d). Arrows indicate tumor.
(C) Inducible ENTPD5 knockdown and rescue stable cell lines were treated with or without Dox (0.0625 mg/ml) for indicated time periods. Aliquots of 10 mg of total
cell extracts were subjected to 10% SDS-PAGE for western blotting analysis of ENTPD5, BiP, glycosylated proteins (PHA blot), and b-actin. Asterisk indicates
decreased glycosylated proteins.
(D) Indicated cell lines were plated and treated with or without Dox for 6 days, total cell lysates were prepared, and aliquots of 10 mg protein were subjected to
SDS-PAGE followed by western blotting analysis using indicated antibodies.
(E) Indicated cells (1 3 104) were seeded in 96-well plates and then treated with and without Dox (1 mg/ml) for 4 days. Cell contents were measured.
(F–H) 2 3 106 LNCaP cells with Dox-inducible shRNA target GFP or ENTPD5 were injected subcutaneously into the flank of nude mice as described in the
Extended Experimental Procedures. When the tumors reached a volume of 500 mm3, mice were fed with normal or Dox-containing water.

722 Cell 143, 711–724, November 24, 2010 ª2010 Elsevier Inc.
Chronic inhibition of ENTPD5 may cause liver and male fertility Measurement of Intracellular Fructose-6-P and Fructose-1,6-2P
defects because mice with ENTPD5 deficiency show hepatop- The preparation and measurement of these two phosphosugars by LC-Mass
were described in the Extended Experimental Procedures.
athy and aspermia (Read et al., 2009). These defects in mice,
however, only become obvious after 1 year of age. Given the
Cell Survival Assay
poor prognosis of PI3K/PTEN mutations in human cancers and Cell survival analysis was performed using the Cell Titer-Glo Luminescent Cell
potential synthetic lethal effect of AKT activation and ENTPD5 Viability Assay kit (Promega) following manufacturer’s instruction with minor
inhibition, developing ENTPD5 inhibitors for cancer therapy modification. In brief, 25 ml of Cell Titer-Glo reagent was added to the cell
may be a worthwhile pursuit. culture medium. Cells were placed on a shaker for 10 min and were then
incubated at room temperature for an additional 10 min. Luminescent reading
was carried on a Tecan SPECTRAFluor Plus reader (Tecan).
EXPERIMENTAL PROCEDURES

General Reagents and Methods SUPPLEMENTAL INFORMATION


General chemicals are from Sigma unless otherwise described. We obtained
Supplemental Information includes Extended Experimental Procedures, seven
a-P32-labeled ATP from GE Healthcare. All other nucleotides are from Sigma.
figures, and one table and can be found with this article online at doi:10.1016/j.
Nonhydrolyzable uracil and guanine nucleotide analogs are from Gena Biosci-
cell.2010.10.010.
ence (Germany). HRP-conjugated E-type PHA is from USBioLogical
(Ca#P3371-25). Puromycin, blasticidin, and hygromycin, which are used for
establishment and maintenance of stable cell lines, are purchased from Inviv- ACKNOWLEDGMENTS
ogen (Ca#ant-pr-1, ant-bl-1, and ant-hg-1, respectively). G418 is from Calbio-
chem (Ca#345810). The sources of antibodies used are listed in the Extended We would like to express our gratitude to Drs. Fenghe Du and Liping Liu for
Experimental Procedures. excellent technical assistance. We are grateful for Dr. Aijun Liu from the 301
Hospital in Beijing for providing the human prostate tumor samples and
Dr. Benjamin Tu from University of Texas Southwestern for help with the phos-
Cell Culture
phofructose sugar measurement. We thank Mr. Gregory Kunkel and Dr. Lai
PTEN+/ and PTEN / MEF cells are established previously (Stambolic et al.,
Wang for critical reading of the manuscript. This work is also supported by
1998). The sources of all other cell lines used and their culture conditions are
a grant from the National Cancer Institute (NCI) (PO1 CA 95471) and the
described in the Extended Experimental Procedures.
National High Technology Projects 863 from Chinese Ministry of Science
and Technology.
In Vitro ATP Hydrolysis Assay
The ATP hydrolysis assays were carried out by incubation-indicated cell Received: May 14, 2010
extracts or purified enzymes with a-P32-ATP and were analyzed by thin layer Revised: September 10, 2010
chromatography (TLC). The detailed method was described in the Extended Accepted: October 7, 2010
Experimental Procedures. Published online: November 11, 2010

Purification of ENTPD5 and CMPK REFERENCES


All purification steps were carried out at 4 C. All chromatography steps were
carried out using an automatic fast protein liquid chromatography (FPLC) Bermudo, R., Abia, D., Ferrer, B., Nayach, I., Benguria, A., Zaballos, A., del
station (Pharmacia). The details of purification methods were described in Rey, J., Miró, R., Campo, E., Martı́nez-A, C., et al. (2008). Co-regulation anal-
the Extended Experimental Procedures. ysis of closely linked genes identifies a highly recurrent gain on chromosome
17q25.3 in prostate cancer. BMC Cancer 8, 315.

ENTPD5 Expression and ENTPD5 shRNA Constructs Brunet, A., Bonni, A., Zigmond, M.J., Lin, M.Z., Juo, P., Hu, L.S., Anderson,
All ENTPD5 expression and shRNA constructs were made as described in the M.J., Arden, K.C., Blenis, J., and Greenberg, M.E. (1999). Akt promotes cell
Extended Experimental Procedures. survival by phosphorylating and inhibiting a Forkhead transcription factor.
Cell 96, 857–868.

Preparation of Recombinant ENTPD5, CMPK1, and Adenylate Christofk, H.R., Vander Heiden, M.G., Harris, M.H., Ramanathan, A., Gerszten,
Kinase R.E., Wei, R., Fleming, M.D., Schreiber, S.L., and Cantley, L.C. (2008). The M2
Human ENTPD5 recombinant protein was generated using Bac-to-Bac Bacu- splice isoform of pyruvate kinase is important for cancer metabolism and
lovirus Expression Systems (Invitrogen Cat# 10359-016). Human CMPK1 and tumour growth. Nature 452, 230–233.
AK1 were generated by bacterial expression. The details of methods were Ellgaard, L., Molinari, M., and Helenius, A. (1999). Setting the standards:
described in the Extended Experimental Procedures. quality control in the secretory pathway. Science 286, 1882–1888.
Elstrom, R.L., Bauer, D.E., Buzzai, M., Karnauskas, R., Harris, M.H., Plas, D.R.,
Measurement of Lactate Production in Cell Culture Medium Zhuang, H., Cinalli, R.M., Alavi, A., Rudin, C.M., and Thompson, C.B. (2004).
We purchased Lactate Assay kit from Biovision (Cat#K627-100). Measure- Akt stimulates aerobic glycolysis in cancer cells. Cancer Res. 64, 3892–3899.
ment of Lactate concentration in cell culture medium was performed accord- Engelman, J.A., Luo, J., and Cantley, L.C. (2006). The evolution of phosphati-
ing the manufacturer’s instruction and described in detail in the Extended dylinositol 3-kinases as regulators of growth and metabolism. Nat. Rev. Genet.
Experimental Procedures. 7, 606–619.

(F) Tumors lysates were extracted after 6 weeks, and aliquots of 20 mg of protein were subjected to 10% SDS-PAGE and transfer to nitrocellulose filter. The filter
was stained with Ponceau S staining (bottom) followed by western blotting analysis using an antibody against ENTPD5 (top).
(G) Time course of tumor shrinkage caused by ENTPD5 knocking down. Tumor size was measured, and statistic analysis was performed as described in the
Extended Experimental Procedures. Each group consisted of seven mice. The values are represented as mean ± SD. *p < 0.05.
(H) Hematoxylin and eosin staining of resected tumors. (Left) Scale bar, 500 um. (Right) Scale bar, 100 um. (Top) Tumor without Dox. (Bottom) Tumor with Dox.
See also Figure S6 and Figure S7.

Cell 143, 711–724, November 24, 2010 ª2010 Elsevier Inc. 723
Fewell, S.W., Travers, K.J., Weissman, J.S., and Brodsky, J.L. (2001). The Read, R., Hansen, G., Kramer, J., Finch, R., Li, L., and Vogel, P. (2009).
action of molecular chaperones in the early secretory pathway. Annu. Rev. Ectonucleoside triphosphate diphosphohydrolase type 5 (Entpd5)-deficient
Genet. 35, 149–191. mice develop progressive hepatopathy, hepatocellular tumors, and spermato-
Fingar, D.C., Salama, S., Tsou, C., Harlow, E., and Blenis, J. (2002). genic arrest. Vet. Pathol. 46, 491–504.
Mammalian cell size is controlled by mTOR and its downstream targets Robson, S.C., Sévigny, J., and Zimmermann, H. (2006). The E-NTPDase family
S6K1 and 4EBP1/eIF4E. Genes Dev. 16, 1472–1487. of ectonucleotidases: Structure function relationships and pathophysiological
Franke, T.F., Kaplan, D.R., Cantley, L.C., and Toker, A. (1997). Direct regula- significance. Purinergic Signal. 2, 409–430.
tion of the Akt proto-oncogene product by phosphatidylinositol-3,4-bisphos-
phate. Science 275, 665–668. Stambolic, V., Suzuki, A., de la Pompa, J.L., Brothers, G.M., Mirtsos, C.,
Sasaki, T., Ruland, J., Penninger, J.M., Siderovski, D.P., and Mak, T.W.
Gevers, W., and Krebs, H.A. (1966). The effects of adenine nucleotides on
(1998). Negative regulation of PKB/Akt-dependent cell survival by the tumor
carbohydrate metabolism in pigeon-liver homogenates. Biochem. J. 98,
suppressor PTEN. Cell 95, 29–39.
720–735.
Helenius, A., and Aebi, M. (2004). Roles of N-linked glycans in the endoplasmic Stephens, L., Anderson, K., Stokoe, D., Erdjument-Bromage, H., Painter, G.F.,
reticulum. Annu. Rev. Biochem. 73, 1019–1049. Holmes, A.B., Gaffney, P.R.J., Reese, C.B., McCormick, F., Tempst, P., et al.
Hirschberg, C.B., Robbins, P.W., and Abeijon, C. (1998). Transporters of (1998). Protein kinase B kinases that mediate phosphatidylinositol 3,4,5-tris-
nucleotide sugars, ATP, and nucleotide sulfate in the endoplasmic reticulum phosphate-dependent activation of protein kinase B. Science 279, 710–714.
and Golgi apparatus. Annu. Rev. Biochem. 67, 49–69. Trombetta, E.S., and Helenius, A. (1999). Glycoprotein reglucosylation and
Keniry, M., and Parsons, R. (2008). The role of PTEN signaling perturbations in nucleotide sugar utilization in the secretory pathway: identification of a nucle-
cancer and in targeted therapy. Oncogene 27, 5477–5485. oside diphosphatase in the endoplasmic reticulum. EMBO J. 18, 3282–3292.
Kohn, A.D., Summers, S.A., Birnbaum, M.J., and Roth, R.A. (1996). Expression Trombetta, E.S., and Parodi, A.J. (2003). Quality control and protein folding in
of a constitutively active Akt Ser/Thr kinase in 3T3-L1 adipocytes stimulates the secretory pathway. Annu. Rev. Cell Dev. Biol. 19, 649–676.
glucose uptake and glucose transporter 4 translocation. J. Biol. Chem. 271,
31372–31378. Vander Heiden, M.G., Cantley, L.C., and Thompson, C.B. (2009).
Lau, K.S., Partridge, E.A., Grigorian, A., Silvescu, C.I., Reinhold, V.N., Understanding the Warburg effect: the metabolic requirements of cell prolifer-
Demetriou, M., and Dennis, J.W. (2007). Complex N-glycan number and ation. Science 324, 1029–1033.
degree of branching cooperate to regulate cell proliferation and differentiation. Warburg, O. (1925). Uber den Stoffwechsel der Carcinomzelle. Klin.
Cell 129, 123–134. Wochenschr. 4, 534–536.
Maehama, T., and Dixon, J.E. (1998). The tumor suppressor, PTEN/MMAC1,
dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-tris- Whitman, M., Downes, C.P., Keeler, M., Keller, T., and Cantley, L.C. (1988).
phosphate. J. Biol. Chem. 273, 13375–13378. Type I phosphatidylinositol kinase makes a novel inositol phospholipid, phos-
phatidylinositol-3-phosphate. Nature 332, 644–646.
Plas, D.R., Talapatra, S., Edinger, A.L., Rathmell, J.C., and Thompson, C.B.
(2001). Akt and Bcl-xL promote growth factor-independent survival through Yuan, T.L., and Cantley, L.C. (2008). PI3K pathway alterations in cancer:
distinct effects on mitochondrial physiology. J. Biol. Chem. 276, 12041–12048. variations on a theme. Oncogene 27, 5497–5510.

724 Cell 143, 711–724, November 24, 2010 ª2010 Elsevier Inc.
Stepwise Histone Replacement by SWR1
Requires Dual Activation with Histone
H2A.Z and Canonical Nucleosome
Ed Luk,1,* Anand Ranjan,1 Peter C. FitzGerald,2 Gaku Mizuguchi,1 Yingzi Huang,1 Debbie Wei,1 and Carl Wu1,*
1Laboratory of Biochemistry and Molecular Biology
2Genome Analysis Unit
National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
*Correspondence: luked@mail.nih.gov (E.L.), carlwu@helix.nih.gov (C.W.)
DOI 10.1016/j.cell.2010.10.019

SUMMARY lelic histone variants, which have an important role in major chro-
mosome activities of the cell, including transcription, DNA repli-
Histone variant H2A.Z-containing nucleosomes are cation, and repair (Ausió, 2006).
incorporated at most eukaryotic promoters. This The widely conserved histone H2A.Z variant shares 60%
incorporation is mediated by the conserved SWR1 sequence identity with the canonical H2A histone and plays
complex, which replaces histone H2A in canonical essential, nonredundant roles in higher eukaryotes (Guillemette
nucleosomes with H2A.Z in an ATP-dependent and Gaudreau, 2006). In yeasts, H2A.Z is not essential, but cells
exhibit slow growth (Carr et al., 1994; Santisteban et al., 2000),
manner. Here, we show that promoter-proximal
chromosome instability (Carr et al., 1994; Krogan et al., 2004),
nucleosomes are highly heterogeneous for H2A.Z in
gene silencing defects (Meneghini et al., 2003), and sensitivity
Saccharomyces cerevisiae, with substantial repre- to genotoxic and environmental stress (Jackson and Gorovsky,
sentation of nucleosomes containing one, two, or 2000; Kobor et al., 2004; Mizuguchi et al., 2004). Crystallo-
zero H2A.Z molecules. SWR1-catalyzed H2A.Z graphic studies have shown that H2A.Z-containing nucleosomes
replacement in vitro occurs in a stepwise and unidi- are in general structurally similar to canonical nucleosomes but
rectional fashion, one H2A.Z-H2B dimer at a time, possess distinct internal and surface features (Suto et al.,
producing heterotypic nucleosomes as intermedi- 2000). Biophysical studies also reported differences in nucleo-
ates and homotypic H2A.Z nucleosomes as end some stability, positioning, and higher-order interactions
products. The ATPase activity of SWR1 is specifically (Zlatanova and Thakar, 2008). Of interest, it has been recently
stimulated by H2A-containing nucleosomes without demonstrated that purified nucleosomes containing both
histone H2A.Z and the histone H3.3 variant are the least stable
ensuing histone H2A eviction. Remarkably, further
among native nucleosomes to salt-induced dissociation
addition of free H2A.Z-H2B dimer leads to hyperstim-
(Jin and Felsenfeld, 2007; Zhang et al., 2005).
ulation of ATPase activity, eviction of nucleosomal Genome-wide mapping of nucleosome distribution indicates
H2A-H2B, and deposition of H2A.Z-H2B. These that the vast majority of budding yeast promoters have a stereo-
results suggest that the combination of H2A-contain- typical chromatin architecture, characterized by two well-posi-
ing nucleosome and free H2A.Z-H2B dimer acting as tioned nucleosomes (+1 and 1) flanking an 80–230 base pair
both effector and substrate for SWR1 governs the region that is relatively depleted for histones and is commonly
specificity and outcome of the replacement reaction. referred to as the ‘‘nucleosome-free region’’ (NFR) (Cairns,
2009; Jiang and Pugh, 2009b; Weiner et al., 2010). With its
NFR-proximal edge covering the transcription start site (TSS),
INTRODUCTION the +1 nucleosome acts as a barrier that occludes the TSS and
helps position downstream nucleosomes in the coding region
The eukaryotic genome is packaged into chromatin within the (Jiang and Pugh, 2009b). Formaldehyde crosslinking and chro-
cell nucleus. The fundamental packaging unit of chromatin is matin immunoprecipitation (ChIP) experiments conducted on
the nucleosome, which consists of an octameric histone core the budding yeast Saccharomyces cerevisiae first demonstrated
around which 147 base pairs (bp) of DNA are wrapped in 1.7 that histone H2A.Z (called Htz1) is enriched at intergenic regions
left-handed superhelical turns, plus linker DNA of variable length upstream of PHO5 and GAL1 even under repressed conditions
between adjacent nucleosome core particles (Kornberg and (Santisteban et al., 2000). It was subsequently shown in
Lorch, 1999). The canonical nucleosome, containing two each genome-wide studies that H2A.Z is dramatically enriched at
of the four main histones, H2A, H2B, H3, and H4, is representa- the promoter-proximal +1 and 1 nucleosomes (Albert et al.,
tive of the bulk of chromatin in the cell nucleus. However, a minor 2007; Raisner et al., 2005), with enrichment diminishing progres-
fraction of the nucleosome population is assembled from nonal- sively away from the promoter (Albert et al., 2007). The presence

Cell 143, 725–736, November 24, 2010 ª2010 Elsevier Inc. 725
of H2A.Z nucleosomes surrounding most yeast promoters in the In this study, we investigated whether the homotypic and
absence of transcription has led to the proposal that H2A.Z-con- heterotypic states of H2A.Z-containing nucleosomes are
taining nucleosomes help poise genes for transcription (Jin and present at the promoters of budding yeast in vivo. We found
Felsenfeld, 2007; Li et al., 2005; Santisteban et al., 2000; Zhang that promoter-proximal nucleosomes are highly heterogeneous
et al., 2005). In metazoans, H2A.Z is localized to nucleosomes in histone variant composition, with substantial representation
proximal to promoters of active genes (Rando and Chang, of nucleosomes containing one, two, or zero H2A.Z molecules.
2009). More recently, H2A.Z has also been implicated in DNA To further understand this phenomenon, we developed an
repair (Morrison and Shen, 2009) and in suppression of spurious in vitro assay to distinguish between compositional states and
noncoding transcription (Zofall et al., 2009). The molecular func- found that the histone replacement reaction is stepwise and
tion of H2A.Z in transcription and DNA repair remains obscure. unidirectional, i.e., progressing from AA (canonical) to AZ to ZZ
Previous studies have shown that the 14 subunit S. cerevisiae nucleosomes. Further investigation of the underlying mechanism
SWI/SNF-related SWR1 complex (SWR1) is required for the showed that ATP hydrolysis by the SWR1 complex is specifically
incorporation of H2A.Z (Kobor et al., 2004; Krogan et al., 2003; activated by H2A-containing nucleosome and additionally by
Mizuguchi et al., 2004). Human counterparts of SWR1, named H2A.Z-H2B dimer, leading to histone replacement. These results
SRCAP and p400, have also been identified (Gévry et al., 2007; lead to a model in which specific activation of SWR1 by the two
Ruhl et al., 2006). SWR1 is itself enriched at promoters, coinci- in vivo histone substrates drives the stepwise, unidirectional
dent with the maxima of H2A.Z distribution (Venters and Pugh, pathway of histone H2A.Z replacement.
2009). The recruitment of SWR1 to promoters is attributed in
part to the bromodomain-containing Bdf1 subunit of SWR1 RESULTS
and its interaction with acetylated histone H3 and H4 tails (Altaf
et al., 2010; Koerber et al., 2009; Raisner et al., 2005). Both AZ and ZZ Nucleosomes Are Present
How SWR1 carries out the ATP-dependent replacement of in Saccharomyces cerevisiae
nucleosomal H2A with H2A.Z is not well understood. Studies To investigate whether budding yeast nucleosomes contain one
from our laboratory have shown that the histone replacement or two copies of the H2A.Z variant, we performed coimmunopre-
reaction can be sufficiently reconstituted in vitro with purified cipitation (co-IP) analysis with the use of a diploid strain in which
components (Luk et al., 2007; Mizuguchi et al., 2004). This basic one allele of HTZ1, the gene encoding S. cerevisiae H2A.Z, is left
reaction has also been demonstrated with purified components untagged and the second HTZ1 allele bears a Flag epitope tag
from mammalian and insect cells and can be enhanced by (HTZ1FLAG) to facilitate purification. Cells in asynchronous
acetylation of the nucleosomal substrate (Altaf et al., 2010; culture were fixed with formaldehyde to preserve nucleosome
Kusch et al., 2004; Ruhl et al., 2006). In the replacement reaction, integrity, and mononucleosomes were generated by MNase
the H2A.Z-H2B dimer is delivered as a unit to SWR1 (Mizuguchi digestion (Figure S1A available online). We immunoprecipitated
et al., 2004; Ruhl et al., 2006) specifically to its Swc2 subunit. Htz1Flag-containing nucleosomes with anti-Flag antibodies and
Delivery is assisted by an H2A.Z-specific chaperone Chz1, analyzed their composition by reversal of crosslinking and
which is displaced upon H2A.Z-H2B binding (Luk et al., 2007). western blotting. Probing with anti-Htz1 antibodies showed
A second binding site for H2A.Z-H2B was recently localized to that untagged Htz1 copurifies with Htz1Flag, indicating the pres-
the N-terminal domain of the Swr1 ATPase subunit (Wu et al., ence of homotypic H2A.Z (ZZ) nucleosomes in yeast cells
2009). The binding of H2A.Z-H2B to SWR1 is independent of (Figure 1A). Moreover, reprobing the same blot with anti-H2A
ATP (Wu et al., 2005). antibodies shows copurification of H2A with Htz1Flag, demon-
Other important steps of the histone replacement reaction strating the existence of heterotypic H2A.Z (AZ) nucleosomes
involve the ATP-dependent eviction of nucleosomal H2A-H2B as well (Figure 1B). Based on the experimentally determined
and insertion of H2A.Z-H2B. However, the mechanisms by which ratios (Z:ZF = 0.29, Figure 1A; A:ZF = 0.49, Figures 1C and 1D),
these steps are carried out are obscure. It is also unclear whether we calculated the relative distribution of ZZ and AZ nucleosomes
SWR1 replaces one or both histone H2A-H2B dimers in a canon- to be 35% and 65%, respectively (Figure S1B).
ical nucleosome with H2A.Z-H2B, producing heterotypic (AZ) or In principle, AZ nucleosomes could be generated from AA
homotypic (ZZ) H2A.Z-containing nucleosomes. In vitro reconsti- nucleosomes by stepwise replacement with H2A.Z-H2B dimers.
tution by salt dialysis shows that the two species can be reconsti- This replacement could occur in a replication-independent
tuted from purified histones and DNA (Chakravarthy et al., 2004; manner in all phases of the cell cycle, including the G1 phase
Suto et al., 2000). Therefore, it is of particular interest to determine (S. Sen and C.W., unpublished data). In addition, AZ nucleo-
whether promoter-proximal H2A.Z nucleosomes are organized in somes could arise as a consequence of disruption of ZZ nucle-
the AZ or ZZ state because they are indistinguishable by standard osomes and reassembly with a mixed histone dimer pool during
ChIP procedures (Albert et al., 2007; Raisner et al., 2005). Muta- DNA replication in S phase. The latter contribution can be mini-
tion of the ATP-binding pocket of the Swr1 ATPase subunit and mized in our analysis by the use of yeast cells arrested in G1
studies with nonhydrolyzable ATP analogs documented that phase by a-mating factor (Figure 1F). Under these conditions,
ATP hydrolysis is an absolute requirement for the histone a haploid yeast strain carrying Htz1Flag as the sole copy still
replacement reaction (Mizuguchi et al., 2004). How the ATPase exhibits substantial copurification of H2A with Htz1Flag (75%
activity of the SWR1 complex is transduced to the eviction of compared to asynchronous cells) (Figure 1E).
H2A-H2B and insertion of H2A.Z-H2B and whether the ATPase We measured the relative proportion of H2A.Z to H2A bound to
activity is regulated in the course of the reaction are unknown. chromatin in G1-arrested cells by quantitative western blotting

726 Cell 143, 725–736, November 24, 2010 ª2010 Elsevier Inc.
Figure 1. Isolation of Homotypic ZZ and
Heterotypic AZ Nucleosomes
(A and B) Histone co-IP analysis of mononucleo-
somes prepared from fixed diploid HTZ1FLAG/
HTZ1 cells (yEL021). (A) The SDS-PAGE and
anti-Htz1 (a-Htz1) western analyses of MNase-
treated nuclear extract (Input), flowthrough of
anti-Flag IP (FT), and anti-Flag immunoprecipi-
tates eluted with Flag peptides (Flag eluate). 20,
10, 5, and 2 ml of the Flag eluate was loaded in
lanes 3, 4, 5, and 6, respectively. Lane 3 was
imaged from a separate western blot. The ratio
of untagged Htz1-to-Htz1Flag for the Flag eluate
is 0.29 ± 0.08 (average and range of two western
analyses). The membrane was stripped and
reprobed with anti-H2A (a-H2A) antibodies in (B).
(C and D) The Flag eluate of (A) was quantified by
a-Htz1 and a-H2A western analyses using
recombinant Htz1 and H2A standards. The esti-
mated molar ratio of H2A to Htz1Flag in the Flag
eluate is 0.49.
(E) Co-IP and western analyses of the Flag eluate
from G1-arrested, asynchronous haploid cells
(yJL036). Numbers indicate quantification of the
Htz1Flag western signal relative to H2A.
(F) FACS analysis of asynchronous and G1-arrested
cells.
(G and H) Quantification of total H2A and Htz1 poly-
peptides in the nuclear extract (Input) of G1-ar-
rested cells. Asterisk (*) indicates a cross-reactive
band. The two panels in (H) are imaged from the
same western blot.
See also Figure S1.

tively depleted for AZ and ZZ nucleo-


somes, was subjected to additional IP
with anti-H2A to give a bound fraction
highly enriched for AA nucleosomes.
using purified bacterially expressed Htz1 and H2A as protein We mapped the locations of each distinct nucleosomal pop-
standards (Figures 1G and 1H). Htz1 constitutes 9% of total ulation by hybridization of amplified, fluorescently labeled DNA
H2A-like histones in chromatin, comparable to previous results to oligonucleotide tiling microarrays covering two yeast chro-
obtained for mammalian cells (4%) (West and Bonner, 1980). mosomes (chromosomes 3 and 6 plus other selected regions),
at 10 bp resolution, for both DNA strands. The results are pre-
ZZ and AZ Nucleosomes Are Enriched at Promoters sented as normalized ratios of nucleosomal to genomic DNA
The presence of homotypic and heterotypic H2A.Z nucleosomes fluorescence (Figure 2A and Figure S3). Consistent with
in G1-arrested cells prompted us to map their genomic previous studies (Albert et al., 2007; Raisner et al., 2005), we
locations. Both AZ and ZZ nucleosomes could be enriched at confirmed that Htz1-containing nucleosomes (Z total) map
promoters genome-wide, or they could be differentially distrib- predominantly to the promoter-proximal +1 and 1 nucleo-
uted among distinct sets of genes. To distinguish between these somes, with enrichment tapering off away from the promoter
possibilities, we used sequential IP to fractionate the heteroge- (Figure 2). Of interest, we found that the subpopulations of
neous nucleosome population into three subpopulations AZ and ZZ nucleosomes are similarly enriched at most
representing ZZ, AZ, and AA nucleosomes. We first immunopuri- promoters (Figure 2A). This is especially evident in the normal-
fied Htz1Flag-containing nucleosomes from haploid yeast cells ized average profiles for 466 nucleosomes in the +1 position
(expressing Htz1Flag as sole source) with the use of anti-Flag (Figure 2B). The relative abundances of AZ and ZZ nucleo-
antibodies to isolate ZZ and AZ nucleosomes in the bound somes at the +1 location are highly correlated (R = 0.89),
fraction, followed by secondary IP of the eluate with anti-H2A arguing against differential enrichment of AZ or ZZ nucleo-
antibodies to separate ZZ from AZ nucleosomes (Figure S2A). somes for a specific subset of promoters (Figure S2C). The
Western blotting of bound and flowthrough fractions confirmed average AZ and ZZ nucleosome profiles surrounding the
that IP was highly efficient (Figure S2B). In addition, the promoter region also show differences. ZZ nucleosome enrich-
flowthrough fraction from the first anti-Flag IP, which is quantita- ment is more restricted to the 1 and +1 positions, whereas AZ

Cell 143, 725–736, November 24, 2010 ª2010 Elsevier Inc. 727
Figure 2. Genomic Distribution of the AA, AZ, and ZZ Nucleosomes
(A) Tiling microarray data of a representative region in chromosome 3 showing the genomic distribution of the Z total (orange), ZZ (green), AZ (purple), AA (red),
and total (blue) nucleosomes. The data are presented as the normalized ratio of nucleosomal and genomic DNA signals. Gray bars indicate coding regions.
(B) Normalized average nucleosome distribution in and around the +1 nucleosome center of 466 genes (Jiang and Pugh, 2009a). Circles illustrate the estimated
positions of the 1, +1, +2, +3, and +4 nucleosomes.
See also Figure S2 and Table S2.

enrichment is comparatively lower and declines more gradually H2A.Z is thought to be predominant but in fact exhibits a similar
away from the promoter (Figure 2B). abundance to canonical H2A (Figure 2B). We conclude that
steady-state histone variation at promoter-proximal nucleo-
Substantial Presence of AA Nucleosomes at Promoters somes is quite heterogeneous in a population of budding yeast,
Previous studies of H2A.Z enrichment at promoters genome- showing significant levels of both variant and canonical nucleo-
wide did not include canonical (AA) nucleosomes, which are somes. Clustering analysis of H2A.Z nucleosome distributions
commonly assumed to be depleted at promoters. To test this for the TATA-containing and TATA-less promoters shows that
assumption, we determined the genomic distribution of the puri- histone heterogeneity appears to be a common feature of most
fied AA nucleosome subpopulation on tiling microarrays yeast promoters, irrespective of gene category (Figure S2D).
(Figure S2A). As anticipated, the normalized AA nucleosome
distribution is similar to that observed for the total nucleosome SWR1 Generates Nucleosomal AZ Intermediate and ZZ
pool (total) (Figures 2A and 2B and Figure S3). However, the abun- End Product In Vitro
dance of AA nucleosomes at promoters is surprisingly substan- The steady-state level of H2A.Z at promoter-proximal nucleo-
tial, despite enrichment of the ZZ and AZ variants. This is espe- somes is a product of opposing H2A.Z assembly and disas-
cially evident at the 1 and +1 nucleosome positions, where sembly pathways in vivo. Incorporation of H2A.Z in nucleosomes

728 Cell 143, 725–736, November 24, 2010 ª2010 Elsevier Inc.
Figure 3. In Vitro Assay Showing the Step-
wise Assembly of AZ and ZZ Nucleosomes
(A and B) Overview and experiment for the in vitro
histone replacement assay. Bead-bound canon-
ical nucleosome arrays (depicted with three nucle-
osomes for simplicity) were incubated with
Htz1Flag-H2B dimer (chaperoned by Chz1, not de-
picted), SWR1, and ATP for 1 hr (step 1). After
washing, the chromatin was digested with MNase
to liberate mononucleosomes (step 2), which were
subsequently analyzed by nondenaturing PAGE
(step 3). AA (bottom), AZF (middle), and ZFZF
(top) nucleosomes were detected by SYBR green
staining.
(C) In vitro histone replacement time course.
SWR1-mediated histone replacement reactions
were stopped at various times by bead pull-
down and washing. Nucleosomal products were
analyzed as described in (A). (Middle) Densito-
metric measurement of the indicated gel region.
(Right) Peak height versus time.

species continues to accumulate past


30 min, reaching a plateau where ZZ
nucleosomes represent the bulk of the
nucleosome population, and AA nucleo-
somes are correspondingly diminished
to a minor fraction (Figure 3C and Fig-
is catalyzed by the SWR1 chromatin remodeling complex, which ure S4B). Thus, reaction kinetics suggests that SWR1 converts
could convert AA nucleosomes to the ZZ state by replacing both AA nucleosomes to the AZ and ZZ species in a stepwise manner.
nucleosomal H2A-H2B dimers with Htz1-H2B in a concerted Data of the above experiment do not inform whether a fully
reaction. Alternatively, SWR1 could replace the H2A-H2B dimers replaced ZZ nucleosome is the reaction end product or
in a stepwise manner involving AZ nucleosomes as a reaction a substrate for additional rounds of H2A.Z replacement (i.e.,
intermediate. To distinguish these models, we developed H2A.Z replacing H2A.Z). We addressed this question by first
a new histone replacement assay. generating a mixed population of immobilized AA, AZ, and ZZ
In this assay, immobilized arrays of canonical nucleosomes nucleosomes by a partial replacement reaction in which Htz1-
are incubated with SWR1 purified from an htz1D strain, H2B dimers provided to SWR1 bear a fluorescent Alexa633 label
Htz1Flag-H2B dimers, and ATP as previously described (Fig- on Htz1 and a Flag tag on H2B (Htz1Alexa-H2BFlag dimers) (Fig-
ure 3A). The chromatin product is then subjected to MNase ure 4A). (Analysis of an aliquot by MNase digestion confirms
digestion to liberate mononucleosomes. Because Htz1 bears that mononucleosome products exhibit retarded electrophoretic
a 33Flag epitope tag, replacement of one nucleosomal H2A- mobility and Alexa633 fluorescence depending on the extent of
H2B dimer with Htz1Flag-H2B retards the native electrophoretic replacement—the bottom band corresponding to unreplaced
mobility of the nucleosome, and replacement of two dimers nucleosomes, and the middle and top bands to nucleosomes
retards the mobility further. Thus, in an ATP-dependent, limited containing one and two Htz1Alexa-H2BFlag dimers, respectively
replacement reaction, three nucleosomal species with discrete [Figures 4A and 4B, lane I].) A second round of SWR1-mediated
mobility can be resolved by nondenaturing PAGE (Figures 3A histone replacement using untagged, unlabeled Htz1-H2B dimers
and 3B). We examined the identities of each nucleosome enabled us to evaluate whether the two Htz1Alexa-H2BFlag dimers
species by western blotting and confirmed that the top, middle, in the ZZ nucleosome were replaceable, as shown by a loss of
and bottom gel bands correspond to ZZ nucleosomes with two Alexa633 fluorescence, SYBR green staining, and Htz1 content
Flags (ZFZF), AZ nucleosomes with one Flag (AZF), and AA nucle- in the top nucleosome band (Figure 4A). However, all three indica-
osomes, respectively (Figure S4A). tors remained essentially unchanged after the second SWR1
The detection of AZ nucleosomes in a partial replacement reaction, indicating that SWR1 does not catalyze replacement
reaction suggests that the heterotypic H2A.Z nucleosome may of ZZ nucleosomes with new H2A.Z-H2B dimers (Figure 4B).
be a reaction intermediate. To investigate this possibility, we This experiment also permitted us to confirm directly that the
monitored the progression of the SWR1-catalyzed replacement heterotypic AZ nucleosome (middle band) is a substrate for
reaction in vitro. Consistent with the hypothesis, we found that SWR1-catalyzed histone replacement by virtue of a potential
the AZ species briefly accumulates upon the addition of ATP, increase in Htz1 content without a change in electrophoretic
reaching a maximum at 30 min, followed by a gradual decrease mobility (Figures 4A and 4B, lane II). We found that the middle
over time (Figure 3C and Figure S4B). By contrast, the ZZ band indeed shows a major increase in the Htz1 western blotting

Cell 143, 725–736, November 24, 2010 ª2010 Elsevier Inc. 729
Figure 4. AA or AZ Nucleosomes Together with Htz1-H2B Dimer Are the Specific Substrates for SWR1
(A and B) Overview and experiment for the in vitro histone replacement assay. Nucleosomal arrays bearing a mixed population of AA, AZ, and ZZ nucleosomes
were marked with Htz1Alexa-H2BFlag dimers. After incubating with unlabeled, untagged Htz1-H2B, SWR1, and ATP, two potential scenarios depicted in II and III
could occur. (B) is the experiment. (Red) Htz1Alexa. (Flag) H2BFLAG. (Scan) Densitometric analysis of the a-Htz1 western blot.
(C–E) Standard histone replacement assay (Mizuguchi et al., 2004). Immobilized AA or ZZ nucleosomal arrays were incubated with SWR1 (or INO80), native Flag-
epitope-tagged histone dimers, and ATP where indicated. 60 nM of dimers and 15 nM nucleosome equivalents were used. The arrays were washed with 0.4 M
KCl before SDS elution and western analysis. (Top) SDS-eluted fraction of the chromatin-bound histones. (Bottom) Free histones in the supernatant fraction. AA
and ZZ ovals indicate the type of nucleosomal arrays used. ZF/B, Htz1Flag-H2B dimer; AF/B, H2AFlag-H2B dimer.
See also Figure S3.

signal, demonstrating that the AZ nucleosome, like the AA nucle- also tested whether the related INO80 remodeling complex
osome (bottom band) is a substrate for SWR1 activity (Figure 4B, could mediate a reverse replacement reaction and found no
bottom, lane II). Taken together, these results provide compel- detectable ATP-driven exchange of H2AFlag into ZZ nucleosomal
ling evidence that the AZ and ZZ nucleosomes are a bona fide arrays under reaction conditions (Figure 4D). Thus, other mech-
intermediate and end product, respectively, of the SWR1-medi- anisms may be responsible for the displacement of H2A.Z and
ated histone replacement reaction. reassembly of the canonical nucleosome. By contrast, incuba-
tion of AA nucleosome arrays with saturating H2A-H2B dimers
(60 nM) gave a small but reproducible level of ATP-dependent
No Reverse Replacement of ZZ Nucleosomes with H2A-
incorporation of H2AFlag into chromatin (Figure 4E), consistent
H2B Dimers
with previous findings (Mizuguchi et al., 2004). We conclude
We confirmed that SWR1 does not replace ZZ nucleosomes with
that the histone replacement pathway mediated by SWR1 is
H2A.Z-H2B dimers using immobilized ZZ nucleosome arrays
unidirectional, with strong substrate specificity for H2A-contain-
reconstituted from bacterially expressed histones. Incubation
ing nucleosomes and the Htz1-H2B dimer.
of these arrays with Flag-tagged histone dimers, SWR1, and
ATP showed that SWR1 failed to replace ZZ nucleosomes with
Htz1Flag-H2B dimers even when dimers were in excess relative Canonical Nucleosomes Specifically Stimulate SWR1
to nucleosomes (Figure 4C). ATPase
Next, we examined whether AZ and AA nucleosomes could be Histone variant replacement by the multicomponent SWR1
produced from the ZZ species though a reverse reaction by incu- complex involves interaction with at least three essential
bation of immobilized ZZ nucleosome arrays with excess substrates: ATP, the H2A-containing nucleosome, and the
H2AFlag-H2B dimers, SWR1, and ATP. This reaction also failed Htz1-H2B dimer. The differential utilization of H2A-containing
to produce detectable incorporation of H2AFlag above back- nucleosomes suggests that SWR1 recognizes H2A- over
ground in the bead-bound chromatin fraction (Figure 4D). We H2A.Z-containing nucleosomes. Specific recognition could be

730 Cell 143, 725–736, November 24, 2010 ª2010 Elsevier Inc.
Figure 5. AA, but Not ZZ, Nucleosomes Stimulate SWR1 ATPase
(A–C) ATPase assay for chromatin remodelers. Inorganic phosphate (Pi) produced during ATP hydrolysis was monitored in real time by MDCC-PBP, which
increases in fluorescence upon phosphate binding (Brune et al., 1994). Reactions were performed at 23 C in the absence (orange) or presence of 15 nM AA nucle-
osomes (red), ZZ nucleosomes (green), or free DNA (blue). ATP was added 20 s before the first measurement (zero time) to final concentrations of 62.5 mM for
SWR1 and 500 mM for INO80 and SWI/SNF. Relative fluorescence was set as zero at zero time for all reactions.
(D) ATPase assay for SWR1 in the absence (orange) or presence of 15 nM recombinant Htz1-H2B dimers (Z/B, black), H2A-H2B dimers (A/B, gray), or AA nucle-
osomes (red).
(E) ATPase assay for SWR1 in the presence of AA nucleosomes and various ATP concentrations. Phosphate concentrations (calculated based on a linear phos-
phate standard curve) were plotted against time. Initial rate (vo) was determined by the slope of the linear part of each curve (0–300 s).
(F and G) Plots of initial rate versus substrate (ATP) concentrations for 1 nM SWR1 and 0.1 nM INO80 in the presence or absence of 15 nM AA nucleosomes, ZZ
nucleosomes, or DNA. The kinetic parameters Vmax and KM were determined by nonlinear fitting of the Michaelis-Menten curve over plotted values.
(H) Turnover number kcat (obtained from dividing Vmax by total enzyme concentration) and KM for the ATPase of SWR1, INO80, and SWI/SNF in the presence or
absence of 15 nM AA nucleosomes, ZZ nucleosomes, or DNA. Error bars represent the range of two measurements.
See also Figure S5.

a consequence of differential nucleosome binding and/or activa- DNA (Figure 5A and Figure S5A). Analysis of initial rates indicates
tion of the ATPase activity of SWR1. We examined whether AA an 2.5-fold increase of ATP hydrolysis at saturating nucleo-
and ZZ nucleosomes differentially stimulate the ATPase activity some and ATP levels. Strikingly, similar concentrations of ZZ
of SWR1 with the use of a real-time fluorescence assay that nucleosomes failed to stimulate the ATPase activity of the
monitors production of inorganic phosphate from ATP hydrolysis SWR1 complex (Figure 5A and Figure S5A). This demonstrates
(Brune et al., 1994). that SWR1 can functionally discriminate between conventional
Previously, we reported that the SWR1 complex exhibits and variant nucleosomes. By contrast, INO80 and SWI/SNF
nucleosome-stimulated ATPase activity as shown by hydrolysis exhibit no differential stimulation of ATPase activity by saturating
of P32-ATP (Mizuguchi et al., 2004). This was confirmed by the levels of AA and ZZ nucleosomes (Figures 5B and 5C and Fig-
fluorescence assay, which shows strong stimulation of ATP ure S5). Of interest, both free H2A-H2B and Htz1-H2B dimers
hydrolysis by conventional nucleosomes, and not by naked failed to stimulate the ATPase activity of SWR1 in the absence

Cell 143, 725–736, November 24, 2010 ª2010 Elsevier Inc. 731
Figure 6. Further Binding of H2A.Z-H2B Dimer Hy-
peractivates SWR1 ATPase and Evicts Nucleo-
somal H2A-H2B
(A) Standard histone replacement assay (Mizuguchi et al.,
2004). Immobilized AA nucleosomal arrays (reconstituted
with H2AHA histone) were incubated with SWR1,
Htz1Flag-H2B (Z/B), histone chaperones, and ATP where
indicated. (Top) Western analyses of chromatin-bound
histones eluted by SDS. (Bottom) Western analyses of
unincorporated histones. 22 nM of Chz1 or FACT was
added to facilitate possible histone eviction.
(B) ATPase assay for SWR1 in the presence of 15 nM AA
nucleosomes and 15 nM Htz1-H2B dimers (purple). Red
is the AA only control.
(C) Kinetic parameters of SWR1 ATPase in the presence of
15 nM AA nucleosomes and 15 nM Htz1-H2B dimers
(purple). For comparison, the curve and parameters for
AA alone (red) are reproduced from Figures 5H and 5F,
respectively. Errors represent the range of two measure-
ments.
(D) Specificity of SWR1 ATPase hyperstimulation. ATPase
assay was performed as described in Figure 5A except
with different combinations of nucleosomes (Nuc) and
histone dimers. Z/B, Htz1-H2B dimers; A/B, H2A-H2B
dimers; AA, AA nucleosomes; ZZ, ZZ nucleosomes; (–)
control, no dimer or nucleosome.
See also Figure S6.

The Michaelis constant (KM), which represents


the ATP concentration at half maximal velocity
(1/2 Vmax), shows little change for canonical
and variant nucleosomes (5 mM and 7 mM,
respectively). In comparison, the stimulated
SWI/SNF has a kcat of 5.5 s1 and KM of 80.5
mM, values consistent with previous determina-
tions (Smith and Peterson, 2005) (Figure 5H).

Nucleosome Stimulation of SWR1


ATPase Is Not Sufficient for H2A-H2B
Eviction
The stimulation of SWR1 ATPase by incubation
with conventional nucleosomes raised the
question of whether such ATP hydrolysis would
be sufficient for eviction of the nucleosomal
H2A-H2B dimer to facilitate Htz1-H2B deposi-
tion. To test this hypothesis, we incubated
SWR1 with immobilized arrays of conventional
nucleosomes carrying epitope-tagged histone
of nucleosomes, suggesting that H2A-specific recognition must H2AHA and monitored H2AHA-H2B eviction in the supernatant
be in the context of nucleosome architecture (Figure 5D). fraction by western blotting. Whereas the SWR1-catalyzed
We determined kinetic parameters for ATP hydrolysis by the Htz1 replacement reaction in the presence of Htz1-H2B dimer
SWR1 complex (Figures 5E, 5F, and 5H). SWR1 has an enzyme (and histone chaperone) occurs robustly with quantitative evic-
turnover rate (kcat) of 0.1 s1 in the presence or absence of DNA. tion of H2AHA, we did not detect any eviction of histone H2AHA
The kcat remains essentially the same when SWR1 is incubated in the absence of Htz1Flag-H2B dimer (Figure 6A). Thus, the stim-
with ZZ nucleosomes but increases to 0.25 s1 (2.5-fold) with ulation of ATP hydrolysis provided solely by canonical nucleo-
saturating AA nucleosomes (Figures 5F and 5H). Hence, binding some effector is inadequate for eviction of nucleosomal
of H2A-containing nucleosomes to SWR1 stimulates ATPase H2A-H2B. Moreover, eviction of H2A-H2B and insertion of
activity by increasing the catalytic efficiency of the enzyme. Htz1-H2B appear to be coupled processes.

732 Cell 143, 725–736, November 24, 2010 ª2010 Elsevier Inc.
Figure 7. A Model for SWR1-Mediated
Histone Replacement
(A) Promoter nucleosome cycle. An AA nucleo-
some at the +1 promoter-proximal position is con-
verted to the AZ and ZZ states by SWR1 via a step-
wise, unidirectional pathway. The ZZ nucleosome
is subsequently converted back to the AA state
through pathways most likely involving nucleo-
some eviction and reassembly with an AA nucleo-
some (dotted gray arrows).
(B) SWR1 catalytic cycle. SWR1 stochastically
binds to one face of an AA nucleosome and the
H2A.Z-H2B dimer, leading to hyperstimulation of
ATPase activity (deep red) and a conformational
change in SWR1 (shown) required for histone
replacement. The newly incorporated Z face of
the AZ nucleosome deactivates the ATPase and
stops further histone replacement activity. The
AZ nucleosome dissociates and rebinds stochas-
tically on the A face for a second replacement
reaction.

Htz1-H2B Dimer and Canonical Nucleosome Specifically nating. However, this balance can be shifted, for example, at
Activate SWR1 highly transcribed promoters (top 10% RNA Pol II occupancy)
The requirement for free Htz1-H2B dimers for SWR1-mediated in which the ZZ and AZ states are underrepresented relative
H2A.Z replacement prompted us to investigate whether addition to the AA state for the +1 nucleosome position (Figure S2E),
of Htz1-H2B to SWR1 further increases ATP hydrolysis. Indeed, suggesting that H2A.Z eviction is occurring at a faster rate
we observed a clear hyperstimulation of the ATPase activity of than incorporation. The greater restriction of the ZZ than AZ
SWR1 when saturating Htz1-H2B dimers (15 nM), and canonical state to +1 and 1 nucleosome positions is interesting and
nucleosomes are both provided to SWR1 in the reaction (Figures may be a consequence of the stepwise nature of the histone
6B and 6D). The hyperstimulated ATPase activity exhibits a kcat replacement reaction and the local concentration of SWR1
of 0.45 s1, which represents an additional 1.8-fold increase in recruited to gene promoters (Venters and Pugh, 2009; Yoshida
the kcat relative to the stimulation by nucleosomes only (a 4.1- et al., 2010).
fold increase in total), with little change of KM (Figure 6C). We Our in vitro studies show that SWR1 is capable of stepwise
observed less hyperstimulation when H2A-H2B dimers were deposition of H2A.Z-H2B into canonical nucleosomes, coupled
substituted for Htz1-H2B at the same molar concentration with H2A-H2B eviction, to give a fully replaced variant nucleo-
(Figure 6D, left). Importantly, a 4-fold increase of H2A-H2B some. However, once incorporated, H2A.Z cannot be evicted
dimers (60 nM) hyperstimulated ATPase activity to nearly by SWR1, even in excess of either H2A.Z-H2B or H2A-H2B
maximal level (Figure 6D, right), whereas hyperstimulation of dimers under otherwise identical reaction conditions. Therefore,
the ATPase activity upon addition of Htz1-H2B or H2A-H2B the SWR1-mediated pathway of H2A.Z replacement is unidirec-
dimers (at either concentration) to ZZ nucleosomes was much tional, terminating with ZZ nucleosomes. It is possible that
lower (Figure 6D). Given that incorporation of new H2A in canon- a reverse reaction from the ZZ to AA nucleosome state requires
ical nucleosomal arrays is low (Figure 4E) under conditions different conditions, cofactors, or modifications of the SWR1
wherein ATPase activity is high, these findings indicate that enzyme or histone substrates. Alternatively, a return to the AA
high ATPase activity per se is not sufficient for histone replace- state may occur through separate pathways. For example, other
ment. It is the presence of the correct in vivo substrates that SWI/SNF family members might possess the capability for
ensures efficient coupling of the high ATPase activity to histone specific replacement of nucleosomal H2A.Z-H2B with H2A-
replacement. H2B, but we have not observed such activity for INO80,
a chromatin remodeling complex paralogous to SWR1, under
DISCUSSION conditions in which INO80 displays robust nucleosome- or
DNA-stimulated ATPase and histone octamer sliding activities
The steady-state level of H2A.Z at promoter-proximal nucleo- (Figure 5, Figure S5, and unpublished data).
somes is a consequence of the opposing pathways of H2A.Z More likely, the well-documented high histone H3 turnover
incorporation and H2A.Z eviction. Our observation of three rate at promoters implies promoter-specific nucleosome
distinct variant states of promoter nucleosomes in a cell popu- disassembly, i.e., eviction of H2A.Z-H2B and H3-H4, and subse-
lation is complementary to previous mapping studies of H2A.Z quent nucleosome reassembly with new histones (Dion et al.,
in budding yeast (Albert et al., 2007; Raisner et al., 2005; San- 2007; Rufiange et al., 2007), thereby completing the dynamic
tisteban et al., 2000). The comparable representation of AA cycling of H2A and H2A.Z at gene promoters (Figure 7A).
and ZZ states suggests that the AA-to-ZZ and ZZ-to-AA path- These processes are likely to be mediated by a combination of
ways are balanced for many genes, without one pathway domi- SWI/SNF family enzymes (Barbaric et al., 2007; Gutiérrez

Cell 143, 725–736, November 24, 2010 ª2010 Elsevier Inc. 733
et al., 2007; Lorch et al., 2006), RNA polymerase (Weiner et al., lated by AA nucleosomes nor hyperstimulated by further addition
2010), and core histone chaperones (Corpet and Almouzni, of Htz1-H2B dimer (Figure S6). These findings indicate that the
2009; Das et al., 2010). In addition, the in vivo lability of H2A.Z- Swr1 ATPase is the key subunit whose activity is governed,
containing nucleosomes as reflected in salt sensitivity should directly and/or indirectly, by the histone effectors.
also contribute (Henikoff et al., 2009; Jin and Felsenfeld, 2007; It will be interesting to define the molecular determinants
Zhang et al., 2005). Indeed, histone modifications at promoters within the canonical nucleosome and the H2A.Z-H2B dimer
correlate with the signatures of newly deposited histones, such that are specifically recognized by the SWR1 complex, to identify
as H3K56Ac and H4K16 deAc (Rando and Chang, 2009). the SWR1 components interacting with the nucleosome, and to
The directional nature of the H2A.Z replacement pathway follow the fate of the evicted H2A-H2B dimer. Other questions
implies that SWR1 must functionally differentiate between ZZ are the importance of the two Htz1-binding modules in SWR1
and AA (or AZ) nucleosomes. We have traced this differentiation, (Swc2 and the N terminus of the Swr1 subunit itself); the relation-
at least in part, to a specific, 2.5-fold increase of the ATPase ship between ATPase activity, DNA translocase activity, and un-
activity (kcat) of SWR1 induced by AA, but not ZZ, nucleosomes. wrapping of nucleosomal DNA; the timing and coupling of H2A
However, this level of stimulation is insufficient for the eviction of eviction and H2A.Z insertion; and the structural transformations
H2A-H2B from nucleosomes. Only after further addition of free of SWR1 that accompany these events. Our present findings and
H2A.Z-H2B dimers is the ATPase activity of SWR1 hyperstimu- the biochemical assays that we have developed should facilitate
lated (4-fold increase of kcat), concurrent with H2A-H2B eviction future investigations on the mechanism of histone H2A.Z
and H2A.Z-H2B deposition. However, a hyperstimulated SWR1 replacement.
ATPase is only necessary, but not sufficient, to mediate robust
histone replacement, as saturating free H2A-H2B dimers can EXPERIMENTAL PROCEDURES
hyperstimulate SWR1 ATPase to nearly maximal level but with
Immunopurification of AA, AZ, and ZZ Nucleosomes
substantially reduced histone replacement (Figure 4E and Fig-
Crude chromatin was isolated from formaldehyde-fixed yeast cells as
ure 6D). This finding implies that unique features of H2A.Z-H2B described in Liang and Stillman (1997) and digested with MNase to mononu-
dimer, in addition to stimulating ATP hydrolysis, enhance histone cleosomal level. Sequential IP was performed with the use of anti-Flag M2
replacement by allosterically coupling the ATPase motor to agarose (Sigma) and anti-H2A antibodies (Active Motif) bound to nProtein A
histone transactions. This additional molecular specificity seems Sepharose (GE Healthcare).
biologically necessary, given that H2A-H2B dimers should be in
excess over H2A.Z-H2B dimers in vivo. Amplification and Labeling of Nucleosomal DNA for Microarray
Analysis
Overall, our data suggest a model in which SWR1 binding to
Nucleosomal DNA and MNase-treated genomic DNA control were treated with
and recognition of its two in vivo histone substrates (one face alkaline phosphatase (CIP, NEB) and end-repair enzyme mix (End-It kit, Epi-
of the AA nucleosome and the H2A.Z-H2B dimer) lead to hyper- centre) before being amplified by ligation-mediated PCR (Johnson et al.,
stimulation of ATPase activity as well as a conformational 2008). Labeling was performed using the BioPrime Plus labeling kit (Invitrogen)
change in SWR1 required for displacement of H2A-H2B and according to the manufacturer’s protocol.
insertion of H2A.Z-H2B (Figure 7B). The order of SWR1 binding
Microarrays
to nucleosomes and H2A.Z-H2B dimers should be stochastic.
Custom tiling microarrays were designed based on the Agilent 4 3 180K plat-
The newly incorporated Z face of the AZ nucleosome deactivates form. Each microarray contained 150,000 biological probes spanning
the ATPase and stops further histone replacement. The AZ selected genomic regions. The tiling probes were spaced, on average, 10 bp
nucleosome subsequently dissociates from and reassociates apart and covered both the sense and antisense DNA strands.
with SWR1 in a stochastic fashion (Figures 7B and 7C). In the
second round, recognition by SWR1 of the A face of the AZ Normalization of Microarray Data for Different Nucleosomal Species
nucleosome and new H2A.Z-H2B dimer binding restimulates Given that Htz1 is the only H2A variant in budding yeast, normalization of mi-
croarray data was performed based on the assumptions that, to a first approx-
SWR1 activity to catalyze replacement of the remaining nucleo-
imation, the sum of Z total and AA nucleosomes is equal to the total nucleo-
somal H2A-H2B with H2A.Z-H2B. Functional recognition of the A some signal and that the sum of AZ and ZZ nucleosomes is equal to the Z
face of an AA or AZ nucleosome and the requirement for free total nucleosome signal. Details are provided in Figure S3 and legend.
H2A.Z-H2B dimer ensures that only these effectors, which are
also substrates for SWR1, are productively utilized. This In Vitro Histone Replacement Assay
provides a way of controlling the specificity and outcome of The SWR1 histone replacement assay was performed according to Mizuguchi
et al. (2004) except the immobilized nucleosomal arrays (80 ng DNA equiva-
the replacement reaction, which terminates with the ZZ
lents) were digested with 0.16 U/ml MNase (+ 2 mM CaCl2) to liberate the nucle-
nucleosome. osomes. The reactions were stopped with 10 mM EDTA before analysis by
The SWR1 complex contains multiple ATP-binding subunits, nondenaturing PAGE.
including Swr1, actin, actin-related proteins Arp4 and Arp6,
and the Rvb1-Rvb2 dodecamer, members of the AAA+ family In Vitro Histone Replacement Assay Using Fluorescently Labeled
of ATPases (Jha and Dutta, 2009; Mizuguchi et al., 2004). We Htz1-H2B Substrate
have previously found that a mutation (K727G substitution) in To generate the mixed AA, AZ, and ZZ nucleosomal substrate for the experi-
ment in Figure 4B, AA nucleosomal arrays were incubated with SWR1 pre-
the ATP-binding motif of the Swr1 subunit is sufficient to
charged with Htz1Alexa-H2BFlag and with Htz1-H2BFlag. The resulting chromatin
abrogate Htz1 replacement in vivo and in vitro without affecting had comparable levels of AA, AZ, and ZZ nucleosomes, which also exhibited
assembly of the SWR1 complex (Mizuguchi et al., 2004). The comparable Alexa633 fluorescence for the AZ and ZZ nucleosomal species. In
ATPase activity of the purified mutant enzyme is neither stimu- the chase step, the labeled nucleosomes were incubated with SWR1

734 Cell 143, 725–736, November 24, 2010 ª2010 Elsevier Inc.
precharged with the unlabeled, untagged Htz1-H2B. After washing, the nucle- Corpet, A., and Almouzni, G. (2009). Making copies of chromatin: the challenge
osomal products were released by MNase digestion and analyzed by nonde- of nucleosomal organization and epigenetic information. Trends Cell Biol. 19,
naturing PAGE as described above. 29–41.
Das, C., Tyler, J.K., and Churchill, M.E. (2010). The histone shuffle: histone
ATPase Assay chaperones in an energetic dance. Trends Biochem. Sci. 35, 476–489.
ATPase assay was performed based on the procedure described in Brune Dion, M.F., Kaplan, T., Kim, M., Buratowski, S., Friedman, N., and Rando, O.J.
et al. (1994). In this assay, inorganic phosphate (Pi) produced during ATP (2007). Dynamics of replication-independent histone turnover in budding
hydrolysis is monitored by the fluorophore-modified phosphate-binding yeast. Science 315, 1405–1408.
protein MDCC-PBP (Phosphate Sensor, Invitrogen), which increases in fluo-
rescence upon Pi binding. Measurements were performed at 23 C on a Wallac Gévry, N., Chan, H.M., Laflamme, L., Livingston, D.M., and Gaudreau, L.
Victor plate reader using a 405 nm excitation, 460 nm emission filter set. (2007). p21 transcription is regulated by differential localization of histone
H2A.Z. Genes Dev. 21, 1869–1881.
Guillemette, B., and Gaudreau, L. (2006). Reuniting the contrasting functions of
ACCESSION NUMBERS
H2A.Z. Biochem. Cell Biol. 84, 528–535.
The GEO accession number for the microarray data sets is GSE24618. Gutiérrez, J.L., Chandy, M., Carrozza, M.J., and Workman, J.L. (2007). Activa-
tion domains drive nucleosome eviction by SWI/SNF. EMBO J. 26, 730–740.

SUPPLEMENTAL INFORMATION Henikoff, S., Henikoff, J.G., Sakai, A., Loeb, G.B., and Ahmad, K. (2009).
Genome-wide profiling of salt fractions maps physical properties of chromatin.
Supplemental Information includes Extended Experimental Procedures, six Genome Res. 19, 460–469.
figures, and three tables and can be found with this article online at doi:10. Jackson, J.D., and Gorovsky, M.A. (2000). Histone H2A.Z has a conserved
1016/j.cell.2010.10.019. function that is distinct from that of the major H2A sequence variants. Nucleic
Acids Res. 28, 3811–3816.
ACKNOWLEDGMENTS Jha, S., and Dutta, A. (2009). RVB1/RVB2: running rings around molecular
biology. Mol. Cell 34, 521–533.
We thank W.H. Wu for the yeast strain SWR1-FL htz1D and J. Landry for the
Jiang, C., and Pugh, B.F. (2009a). A compiled and systematic reference map of
yeast strain yJL036. We also thank H. Cam and C. Rubin for advice on micro-
nucleosome positions across the Saccharomyces cerevisiae genome.
array techniques, F. Pugh and members of the Wu lab for critical reading of the
Genome Biol. 10, R109.
manuscript, and anonymous reviewers for helpful suggestions. This work was
supported by the intramural research program of the National Cancer Institute Jiang, C., and Pugh, B.F. (2009b). Nucleosome positioning and gene regula-
(C.W.) and by the Leukemia and Lymphoma Society (E.L. and A.R.). tion: advances through genomics. Nat. Rev. Genet. 10, 161–172.
Jin, C., and Felsenfeld, G. (2007). Nucleosome stability mediated by histone
Received: May 27, 2010 variants H3.3 and H2A.Z. Genes Dev. 21, 1519–1529.
Revised: August 25, 2010
Johnson, D.S., Li, W., Gordon, D.B., Bhattacharjee, A., Curry, B., Ghosh, J.,
Accepted: October 12, 2010
Brizuela, L., Carroll, J.S., Brown, M., Flicek, P., et al. (2008). Systematic eval-
Published: November 24, 2010
uation of variability in ChIP-chip experiments using predefined DNA targets.
Genome Res. 18, 393–403.
REFERENCES
Kobor, M.S., Venkatasubrahmanyam, S., Meneghini, M.D., Gin, J.W., Jen-
Albert, I., Mavrich, T.N., Tomsho, L.P., Qi, J., Zanton, S.J., Schuster, S.C., and nings, J.L., Link, A.J., Madhani, H.D., and Rine, J. (2004). A protein complex
Pugh, B.F. (2007). Translational and rotational settings of H2A.Z nucleosomes containing the conserved Swi2/Snf2-related ATPase Swr1p deposits histone
across the Saccharomyces cerevisiae genome. Nature 446, 572–576. variant H2A.Z into euchromatin. PLoS Biol. 2, E131.

Altaf, M., Auger, A., Monnet-Saksouk, J., Brodeur, J., Piquet, S., Cramet, M., Koerber, R.T., Rhee, H.S., Jiang, C., and Pugh, B.F. (2009). Interaction of tran-
Bouchard, N., Lacoste, N., Utley, R.T., Gaudreau, L., and Côté, J. (2010). scriptional regulators with specific nucleosomes across the Saccharomyces
NuA4-dependent acetylation of nucleosomal histones H4 and H2A directly genome. Mol. Cell 35, 889–902.
stimulates incorporation of H2A.Z by the SWR1 complex. J. Biol. Chem. Kornberg, R.D., and Lorch, Y. (1999). Twenty-five years of the nucleosome,
285, 15966–15977. fundamental particle of the eukaryote chromosome. Cell 98, 285–294.
Ausió, J. (2006). Histone variants—the structure behind the function. Brief. Krogan, N.J., Keogh, M.C., Datta, N., Sawa, C., Ryan, O.W., Ding, H., Haw,
Funct. Genomics Proteomics 5, 228–243. R.A., Pootoolal, J., Tong, A., Canadien, V., et al. (2003). A Snf2 family
Barbaric, S., Luckenbach, T., Schmid, A., Blaschke, D., Hörz, W., and Korber, ATPase complex required for recruitment of the histone H2A variant Htz1.
P. (2007). Redundancy of chromatin remodeling pathways for the induction of Mol. Cell 12, 1565–1576.
the yeast PHO5 promoter in vivo. J. Biol. Chem. 282, 27610–27621. Krogan, N.J., Baetz, K., Keogh, M.C., Datta, N., Sawa, C., Kwok, T.C., Thomp-
Brune, M., Hunter, J.L., Corrie, J.E., and Webb, M.R. (1994). Direct, real-time son, N.J., Davey, M.G., Pootoolal, J., Hughes, T.R., et al. (2004). Regulation of
measurement of rapid inorganic phosphate release using a novel fluorescent chromosome stability by the histone H2A variant Htz1, the Swr1 chromatin re-
probe and its application to actomyosin subfragment 1 ATPase. Biochemistry modeling complex, and the histone acetyltransferase NuA4. Proc. Natl. Acad.
33, 8262–8271. Sci. USA 101, 13513–13518.
Cairns, B.R. (2009). The logic of chromatin architecture and remodelling at Kusch, T., Florens, L., Macdonald, W.H., Swanson, S.K., Glaser, R.L., Yates,
promoters. Nature 461, 193–198. J.R., III, Abmayr, S.M., Washburn, M.P., and Workman, J.L. (2004). Acetylation
Carr, A.M., Dorrington, S.M., Hindley, J., Phear, G.A., Aves, S.J., and Nurse, P. by Tip60 is required for selective histone variant exchange at DNA lesions.
(1994). Analysis of a histone H2A variant from fission yeast: evidence for a role Science 306, 2084–2087.
in chromosome stability. Mol. Gen. Genet. 245, 628–635. Li, B., Pattenden, S.G., Lee, D., Gutiérrez, J., Chen, J., Seidel, C., Gerton, J.,
Chakravarthy, S., Bao, Y., Roberts, V.A., Tremethick, D., and Luger, K. (2004). and Workman, J.L. (2005). Preferential occupancy of histone variant H2AZ at
Structural characterization of histone H2A variants. Cold Spring Harb. Symp. inactive promoters influences local histone modifications and chromatin re-
Quant. Biol. 69, 227–234. modeling. Proc. Natl. Acad. Sci. USA 102, 18385–18390.

Cell 143, 725–736, November 24, 2010 ª2010 Elsevier Inc. 735
Liang, C., and Stillman, B. (1997). Persistent initiation of DNA replication and Smith, C.L., and Peterson, C.L. (2005). A conserved Swi2/Snf2 ATPase motif
chromatin-bound MCM proteins during the cell cycle in cdc6 mutants. Genes couples ATP hydrolysis to chromatin remodeling. Mol. Cell. Biol. 25, 5880–
Dev. 11, 3375–3386. 5892.
Lorch, Y., Maier-Davis, B., and Kornberg, R.D. (2006). Chromatin remodeling Suto, R.K., Clarkson, M.J., Tremethick, D.J., and Luger, K. (2000). Crystal
by nucleosome disassembly in vitro. Proc. Natl. Acad. Sci. USA 103, 3090– structure of a nucleosome core particle containing the variant histone
3093. H2A.Z. Nat. Struct. Biol. 7, 1121–1124.
Luk, E., Vu, N.D., Patteson, K., Mizuguchi, G., Wu, W.H., Ranjan, A., Backus, Venters, B.J., and Pugh, B.F. (2009). A canonical promoter organization of the
J., Sen, S., Lewis, M., Bai, Y., and Wu, C. (2007). Chz1, a nuclear chaperone for transcription machinery and its regulators in the Saccharomyces genome.
histone H2AZ. Mol. Cell 25, 357–368. Genome Res. 19, 360–371.
Meneghini, M.D., Wu, M., and Madhani, H.D. (2003). Conserved histone Weiner, A., Hughes, A., Yassour, M., Rando, O.J., and Friedman, N. (2010).
variant H2A.Z protects euchromatin from the ectopic spread of silent hetero- High-resolution nucleosome mapping reveals transcription-dependent
chromatin. Cell 112, 725–736. promoter packaging. Genome Res. 20, 90–100.
Mizuguchi, G., Shen, X., Landry, J., Wu, W.H., Sen, S., and Wu, C. (2004). ATP- West, M.H., and Bonner, W.M. (1980). Histone 2A, a heteromorphous family of
driven exchange of histone H2AZ variant catalyzed by SWR1 chromatin re- eight protein species. Biochemistry 19, 3238–3245.
modeling complex. Science 303, 343–348. Wu, W.H., Alami, S., Luk, E., Wu, C.H., Sen, S., Mizuguchi, G., Wei, D., and Wu,
Morrison, A.J., and Shen, X. (2009). Chromatin remodelling beyond transcrip- C. (2005). Swc2 is a widely conserved H2AZ-binding module essential for ATP-
tion: the INO80 and SWR1 complexes. Nat. Rev. Mol. Cell Biol. 10, 373–384. dependent histone exchange. Nat. Struct. Mol. Biol. 12, 1064–1071.
Raisner, R.M., Hartley, P.D., Meneghini, M.D., Bao, M.Z., Liu, C.L., Schreiber, Wu, W.H., Wu, C.H., Ladurner, A., Mizuguchi, G., Wei, D., Xiao, H., Luk, E.,
S.L., Rando, O.J., and Madhani, H.D. (2005). Histone variant H2A.Z marks the Ranjan, A., and Wu, C. (2009). N terminus of Swr1 binds to histone H2AZ
50 ends of both active and inactive genes in euchromatin. Cell 123, 233–248. and provides a platform for subunit assembly in the chromatin remodeling
Rando, O.J., and Chang, H.Y. (2009). Genome-wide views of chromatin struc- complex. J. Biol. Chem. 284, 6200–6207.
ture. Annu. Rev. Biochem. 78, 245–271. Yoshida, T., Shimada, K., Oma, Y., Kalck, V., Akimura, K., Taddei, A., Iwahashi,
Rufiange, A., Jacques, P.E., Bhat, W., Robert, F., and Nourani, A. (2007). H., Kugou, K., Ohta, K., Gasser, S.M., and Harata, M. (2010). Actin-related
Genome-wide replication-independent histone H3 exchange occurs predom- protein Arp6 influences H2A.Z-dependent and -independent gene expression
inantly at promoters and implicates H3 K56 acetylation and Asf1. Mol. Cell 27, and links ribosomal protein genes to nuclear pores. PLoS Genet. 6, e1000910.
393–405. Zhang, H., Roberts, D.N., and Cairns, B.R. (2005). Genome-wide dynamics of
Ruhl, D.D., Jin, J., Cai, Y., Swanson, S., Florens, L., Washburn, M.P., Con- Htz1, a histone H2A variant that poises repressed/basal promoters for activa-
away, R.C., Conaway, J.W., and Chrivia, J.C. (2006). Purification of a human tion through histone loss. Cell 123, 219–231.
SRCAP complex that remodels chromatin by incorporating the histone variant Zlatanova, J., and Thakar, A. (2008). H2A.Z: view from the top. Structure 16,
H2A.Z into nucleosomes. Biochemistry 45, 5671–5677. 166–179.
Santisteban, M.S., Kalashnikova, T., and Smith, M.M. (2000). Histone H2A.Z Zofall, M., Fischer, T., Zhang, K., Zhou, M., Cui, B., Veenstra, T.D., and Grewal,
regulats transcription and is partially redundant with nucleosome remodeling S.I. (2009). Histone H2A.Z cooperates with RNAi and heterochromatin factors
complexes. Cell 103, 411–422. to suppress antisense RNAs. Nature 461, 419–422.

736 Cell 143, 725–736, November 24, 2010 ª2010 Elsevier Inc.
Sororin Mediates Sister Chromatid
Cohesion by Antagonizing Wapl
Tomoko Nishiyama,1 Rene Ladurner,1 Julia Schmitz,1,4 Emanuel Kreidl,1 Alexander Schleiffer,1 Venugopal Bhaskara,1
Masashige Bando,2 Katsuhiko Shirahige,2 Anthony A. Hyman,3 Karl Mechtler,1 and Jan-Michael Peters1,*
1Research Institute of Molecular Pathology, Dr. Bohr-Gasse 7, A-1030 Vienna, Austria
2Institute
of Molecular and Cellular Biosciences, University of Tokyo, Yayoi, Tokyo 113-0032, Japan
3Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, D-01307 Dresden, Germany
4Present address: World Health Organization, Avenue Appia 20, CH-1211 Geneva 27, Switzerland

*Correspondence: peters@imp.univie.ac.at
DOI 10.1016/j.cell.2010.10.031

SUMMARY are further associated with a heterodimer of two proteins, called


Wapl and Pds5, the latter of which also exists in two isoforms in
Sister chromatid cohesion is essential for chromo- vertebrates (Pds5A and Pds5B; Gandhi et al., 2006; Kueng et al.,
some segregation and is mediated by cohesin bound 2006).
to DNA. Cohesin-DNA interactions can be reversed Cohesin complexes are loaded onto DNA before replication (in
by the cohesion-associated protein Wapl, whereas animal cells already in telophase) and establish cohesion during
a stably DNA-bound form of cohesin is thought to replication. In the subsequent mitosis, cohesion is dissolved by
removal of cohesin from chromosomes. In vertebrate cells, this
mediate cohesion. In vertebrates, Sororin is essential
process occurs in two steps (Waizenegger et al., 2000): the
for cohesion and stable cohesin-DNA interactions,
bulk of cohesin is removed from chromosomes in prophase by
but how Sororin performs these functions is a mechanism that depends on Polo-like kinase 1 (Plk1/Plx1)
unknown. We show that DNA replication and cohesin and Wapl (Gandhi et al., 2006; Kueng et al., 2006). At centro-
acetylation promote binding of Sororin to cohesin, meres, small amounts of cohesin are protected from the
and that Sororin displaces Wapl from its binding prophase pathway by Shugoshin, and these complexes can
partner Pds5. In the absence of Wapl, Sororin only be removed from chromosomes by the protease separase
becomes dispensable for cohesion. We propose (reviewed in Sakuno and Watanabe, 2009). This process occurs
that Sororin maintains cohesion by inhibiting Wapl’s only in metaphase because a surveillance mechanism called the
ability to dissociate cohesin from DNA. Sororin has spindle assembly checkpoint (SAC) prevents separase activa-
only been identified in vertebrates, but we show tion until all chromosomes have been bioriented. The SAC
inhibits APC/CCdc20 (anaphase-promoting complex/cyclosome
that many invertebrate species contain Sororin-
associated with Cdc20), a complex whose ubiquitin ligase
related proteins, and that one of these, Dalmatian, activity is required for separase activation (reviewed in Peters,
is essential for cohesion in Drosophila. The mecha- 2006).
nism we describe here may therefore be widely How cohesion is established and maintained is poorly under-
conserved among different species. stood. Fluorescence recovery after photobleaching (FRAP)
experiments in mammalian cells revealed that cohesin binds to
INTRODUCTION DNA much more stably after than before DNA replication, sug-
gesting that cohesion depends on an unidentified event during
In eukaryotic cells, sister chromatids remain physically con- DNA replication that stabilizes cohesin on DNA (Gerlich et al.,
nected from the time of their synthesis during DNA replication 2006). The dynamic mode of cohesin binding to DNA might
until their separation during mitosis or meiosis. This sister chro- depend on Wapl because depletion of this protein from mamma-
matid cohesion is essential for biorientation of chromosomes on lian cells does not only interfere with the prophase pathway but
the spindle and for DNA-damage repair (reviewed in Nasmyth also increases the residence time of cohesin on chromatin during
and Haering, 2009; Onn et al., 2008; Peters et al., 2008). Cohe- interphase (Kueng et al., 2006).
sion is mediated by cohesin complexes. Three cohesin subunits, The only molecular event during DNA replication that is known
the ATPases Smc1 and Smc3 and the kleisin Scc1/Rad21/ to be essential for cohesion establishment is acetylation of cohe-
Mcd1, form triangular structures that have been proposed to sin (Ben-Shahar et al., 2008; Unal et al., 2008; Zhang et al., 2008).
mediate cohesion by embracing sister chromatids (Gruber This modification occurs on two lysine residues in the ATPase
et al., 2003; for an illustration of this ‘‘ring model,’’ see Figure 6C domain of Smc3 (K112/113 in budding yeast) and is catalyzed
below). Scc1 binds to a fourth core subunit, called Scc3 in yeast by the acetyltransferase Eco1. The lethality of yeast that is
and stromal antigen (SA) in vertebrates, where somatic cells caused by deletion of the ECO1 gene can be suppressed by
contain two SA paralogs (SA1 and SA2). Scc1 and SA proteins changing K112/113 to residues that might functionally mimic

Cell 143, 737–749, November 24, 2010 ª2010 Elsevier Inc. 737
Figure 1. Sororin Is Required for Cohesion in S Phase
(A) FISH of Sororin-depleted S phase cells. HeLa cells were synchronized in S phase by double thymidine arrest and transfected with control or Sororin siRNA.
Four hours after release from the second thymidine arrest, cells were labeled with BrdU for 15 min, pre-extracted, and subjected to FISH with a probe specific for

738 Cell 143, 737–749, November 24, 2010 ª2010 Elsevier Inc.
acetylated lysine but also by deletion of the WPL1/RAD61 gene, depleted cells only lose cohesion during metaphase and that So-
which encodes a Wapl ortholog, and by mutations in Pds5 (Ben- rorin is therefore not required for cohesion in early mitosis (Diaz-
Shahar et al., 2008; Rowland et al., 2009; Sutani et al., 2009; Unal Martinez et al., 2007). However, in time-lapse microscopy exper-
et al., 2008). Cohesin is also acetylated in mammalian cells on iments we observed that most Sororin-depleted cells failed to
Smc3 residues K105/106 (Zhang et al., 2008), where two Eco1 congress chromosomes, consistent with the existence of cohe-
orthologs exist, called Esco1 and Esco2 (Hou and Zou, 2005). sion defects before metaphase (Figures S1A–S1D available on-
In vertebrate cells, cohesin-DNA interactions are also regu- line). The function of Sororin is therefore not restricted to mitosis
lated by Sororin. This protein was identified as a substrate of and is instead already needed during or shortly after DNA
APC/CCdh1, a form of the APC/C that is active during mitotic replication.
exit and G1 phase, and Soronin was found to be essential for
cohesion in mammalian cells (Rankin et al., 2005). Interestingly, Sororin Associates with Chromatin during the Period
Sororin depletion also reduces the number of cohesin com- of the Cell Cycle Where Cohesion Exists
plexes that bind stably to DNA during G2 phase, indicating that We next analyzed the intracellular distribution of Sororin.
Sororin is required for the formation of stable cohesin-DNA inter- Previous IFM and fractionation experiments had shown that So-
actions (Schmitz et al., 2007). However, it is unknown how rorin associates with chromatin in interphase, but Sororin could
Sororin performs this function, and whether the role of Sororin not be detected on mitotic chromosomes (Rankin et al., 2005).
is related to the function of cohesin acetylation. Furthermore, it Because our antibodies could not detect Sororin in IFM experi-
is unknown how widespread the role of Sororin is because ments, we tagged Sororin at its carboxy-terminus with a localiza-
Sororin has only been identified in vertebrates. tion-affinity purification (LAP) tag that contains green fluorescent
Here we provide evidence that Sororin is recruited to chro- protein (GFP; Figure S1E). We modified the Sororin gene on
matin-bound cohesin complexes in a manner that depends on a bacterial artificial chromosome (BAC), enabling gene expres-
DNA replication and Smc3 acetylation, that Sororin causes sion from the endogenous promoter (Poser et al., 2008). We
a conformational rearrangement within cohesin by displacing used a mouse BAC for these experiments to enable RNAi
Wapl from Pds5, and that these molecular events stabilize cohe- ‘‘rescue’’ experiments and generated clonal HeLa cell lines
sin on DNA by antagonizing Wapl’s ability to dissociate cohesin that had stably integrated this BAC. The LAP-tagged version of
from DNA. Furthermore, we show that distant orthologs of So- mouse Sororin could substitute for the cohesion function of
rorin exist in many metazoan species, and that one of these endogenous human Sororin when this was depleted by RNAi
proteins, Dalmatian, is required for cohesion in Drosophila. We (Figures S1F and S1G), and in tandem affinity purification exper-
therefore propose that sister chromatid cohesion depends on iments mouse Sororin-LAP was found associated with human
stabilization of cohesin on DNA by Sororin-related proteins. cohesin (Figures S1H and S1I), indicating that this tagged
version of Sororin behaves similarly to endogenous Sororin.
RESULTS We therefore analyzed by IFM the intracellular distribution of So-
rorin-LAP, using antibodies to GFP. We stained proliferating cell
Sororin Is Required for Cohesion during S Phase nuclear antigen (PCNA) and Aurora B in the same cells as
We had previously shown that Sororin is required for cohesion in markers for S and G2 phases, respectively. Cellular Sororin-
G2 phase (Schmitz et al., 2007). To address whether Sororin’s LAP levels were low in G1, accumulated between early S and
function is already needed during S phase, we used RNA inter- G2 phases in the nucleus, and became dispersed in the cyto-
ference (RNAi) to deplete Sororin from HeLa cells that had plasm following nuclear envelope breakdown (Figures S1J–
been synchronized in the cell cycle and pulse-labeled these cells S1L). When we analyzed cells from which soluble proteins had
with bromodeoxyuridine (BrdU). Cells in S phase were identified been extracted before fixation, we observed that Sororin-LAP
by immunofluorescence microscopy (IFM) using BrdU anti- accumulated on chromatin between early S phase and G2
bodies, and the distance between sister chromatids was phase, whereas most Sororin-LAP disappeared from chromo-
measured by DNA fluorescence in situ hybridization (FISH) using somes in prophase (Figures 1C–1E). At this stage, the cellular
a probe for an arm region on chromosome 21. On average, FISH levels of Sororin were still high (Figure S1L), indicating that the
signals were twice as far separated in BrdU-positive, Sororin- removal of Sororin from prophase chromosomes is caused by
depleted cells than in control cells (Figures 1A and 1B), indicating dissociation, not degradation. Biochemical fractionation experi-
that Sororin is already required for cohesion during S phase. At ments confirmed this notion (Figure S1M). Importantly, however,
variance with these results, it has been reported that Sororin- small amounts of Sororin-LAP could still be detected by IFM on

the trisomic tff1 locus on chromosome 21. BrdU-labeled nuclei (blue) with three pairs of FISH signals (red) are shown. Higher-magnification images are shown in
the insets. Bar: 5 mm.
(B) Quantification of the distance between paired FISH signals in (A) (mean ± standard deviation [SD]; n R 30 per condition, *p < 0.01).
(C) Sororin-LAP cells were pre-extracted prior to fixation and stained for Sor-LAP (GFP), PCNA, and Aurora B. DNA was counterstained with Hoechst. Bar: 10 mm.
(D) Quantification of chromatin-bound Sororin-LAP levels in (C) (mean ± SD; n R 50 per class).
(E) Sororin-LAP cells were synchronized in mitosis, pre-extracted prior to fixation, and stained for Sor-LAP (GFP), Scc1, and DNA (Hoechst). Bar: 10 mm.
(F) Sororin-LAP localizes to centromeres in mitosis. Sororin-LAP cells were pre-extracted prior to fixation and stained for Sor-LAP (GFP), kinetochores (CREST),
and DNA (DAPI). Insets show magnified views. Bar: 10 mm.
See also Figure S1.

Cell 143, 737–749, November 24, 2010 ª2010 Elsevier Inc. 739
Figure 2. Association of Sororin with
Chromatin Depends on Cohesin and DNA
Replication
(A–D) Sororin-LAP cells were transfected with
siRNAs and synchronized in G2 phase. Cells
were fixed (C and D) or pre-extracted prior to
fixation (A and B) and stained for Sor-LAP (GFP),
Scc1, and DNA (Hoechst). Bar: 10 mm. Quantifica-
tion of Sororin-LAP levels in (A) and (C) is shown in
(B) and (D), respectively (mean ± SD; n R 110 (B)
and n R 130 (D) per condition).
(E) Sororin is stably present throughout the cell
cycle but associates with chromatin during S
phase in Xenopus egg extracts. CaCl2 and cyclo-
heximide were added to meiotic metaphase II
(MII) arrested CSF extract to induce meiotic exit.
At 90 min after CaCl2 addition, D90 Cyclin B was
added to induce mitosis. Samples were taken at
indicated time points after CaCl2 addition (release
from MII) or D90 Cyclin B addition (D90 Cyc B
addition). DNA replication (DNA repl.) was moni-
tored by incorporation of [a-32P]dCTP into sperm
chromatin. Chromatin-bound proteins in the
same extracts are also shown. Chromatin was
preincubated for 30 min in CSF extracts.
(F) Sororin association with chromatin depends
on cohesin. Xenopus interphase extracts were
subjected to mock or SA1/2 immunodepletion.
Two hours after sperm chromatin addition, chro-
matin fractions were analyzed by immunoblotting.
(G) Sororin association with chromatin depends
on DNA replication. Interphase extracts were
incubated for indicated times with sperm chro-
matin. DMSO, aphidicolin (Aph.), or actinomycin
D (ActD) was added to the extracts 25 min
after sperm addition. Chromatin fractions were
analyzed by immunoblotting.
See also Figure S2.

chromosomes in prophase, prometaphase, and metaphase, but Although the intracellular distribution of Sororin and cohesin is
not in anaphase or telophase (Figure 1E). Like cohesin (Waize- similar from prophase to anaphase, the two proteins behave
negger et al., 2000), Sororin-LAP was enriched at centromeres differently in telophase. Whereas cohesin reassociates with
in prometa/metaphase (Figure 1F). Sororin therefore associates chromatin at this stage, little if any Sororin-LAP could be de-
with chromatin from S phase until metaphase, i.e., as long as tected on chromatin in telophase (Figure 1E). This difference
cohesion exists. was not due to lower sensitivity in the detection of Sororin than
cohesin because Sororin-LAP could easily be observed on early
The Association of Sororin with Chromatin Depends mitotic chromosomes, where endogenous cohesin cannot be
on Cohesin detected (due to its low abundance; Waizenegger et al., 2000).
Because Sororin binds to cohesin and, like cohesin, is removed The absence of Sororin on telophase chromatin was also not
from mitotic chromosomes in two steps, during prophase and at caused by APC/CCdh1-mediated degradation of all cellular So-
the metaphase-anaphase transition, we tested whether the rorin because Sororin-LAP could be observed in fixed telophase
association of Sororin with chromatin depends on cohesin. cells (Figure S1L). Time-lapse microscopy of living cells showed
Scc1 depletion reduced the intensity of Sororin-LAP staining that Sororin levels begin to decrease in anaphase when APC/
on chromatin without affecting the cellular levels of Sororin- CCdh1 becomes active but revealed that most Sororin degrada-
LAP (Figures 2A–2D), indicating that Sororin can only efficiently tion occurs after telophase, i.e., during G1, as is typical for
associate with chromatin in the presence of cohesin. Biochem- APC/CCdh1 substrates (Figures S2B–S2E). The absence of So-
ical experiments in Xenopus egg extracts confirmed this notion rorin on chromatin in telophase is therefore not simply due to
(see Figure 2F below). The presence of Sororin on mitotic the complete degradation of Sororin.
chromosomes also depends on cohesin, as depletion of either
Scc1 or Shugoshin-like 1 (Sgo1) reduced chromosomal So- Efficient Association of Sororin with Chromatin
rorin-LAP staining, whereas depletion of Wapl or inhibition of Depends on DNA Replication
Plk1 increased the amounts of Sororin on chromosome arms The absence of Sororin on telophase chromatin could be caused
(Figure S2A). by local APC/CCdh1-mediated degradation on chromatin, or the

740 Cell 143, 737–749, November 24, 2010 ª2010 Elsevier Inc.
association of cohesin with chromatin could be required but not replication forks from which the replicative MCM helicase is un-
sufficient for Sororin binding to chromatin. To distinguish coupled, whereas actinomycin D inhibits progression of both
between these possibilities, we analyzed the chromatin associa- DNA polymerase and helicase (Pacek and Walter, 2004). In our
tion of Sororin in Xenopus eggs, which do not contain Cdh1 and assays, aphidicolin reduced association of Sororin with chro-
where Sororin is therefore predicted to be stable during mitotic matin partially, and actinomycin D inhibited this process largely,
exit. If cohesin was sufficient for recruiting Sororin to chromatin, even though Smc3 levels on chromatin were not reduced (Fig-
both proteins would be expected to associate with chromatin ure 2G). DNA replication is therefore required for efficient recruit-
simultaneously in Xenopus egg extracts. To test this possibility, ment of Sororin to chromatin. However, because aphidicolin and
we isolated two Xenopus Sororin cDNAs (Sororin-A and -B), actinomycin D inhibited DNA replication more efficiently than So-
which encode closely related 35 kDa proteins. Xenopus Sororin rorin binding, it is possible that some Sororin can associate with
antibodies recognized both Sororin isoforms in immunoblots chromatin in the absence of DNA replication. Similar observa-
(visible as a doublet of bands; see for example Figure 2E) and tions were made in HeLa cells where inhibition of DNA replication
could deplete both proteins from egg extracts (see Figure 4A by thymidine also reduced the Sororin-LAP levels on chromatin
below). Immunodepletion experiments also revealed that the (Figures S2G and S2H).
chromatin association of Xenopus Sororin proteins depends on
cohesin (Figure 2F) and that these proteins are required for cohe- Cohesin Acetylation Facilitates but Is Not Sufficient for
sion (see Figure 4B below), even though their amino acid the Association of Sororin with Chromatin
sequences are only 38% identical to the sequence of human So- Because Sororin associates with chromatin during DNA replica-
rorin. The two Xenopus proteins characterized here (hereafter tion, i.e., when cohesin is known to be acetylated, we analyzed
collectively called Xenopus Sororin) are therefore functionally whether Smc3 acetylation and Sororin binding depend on each
related to mammalian Sororin. other. To detect Smc3 acetylation, we used a monoclonal anti-
To address when Sororin and cohesin associate with chro- body that specifically recognizes Smc3 singly acetylated on
matin, we released Xenopus egg extracts from a cytostatic K106 or doubly acetylated on K105 and K106 (Figure S3A). We
factor (CSF) arrest in metaphase of meiosis II into interphase observed that Sororin binding to chromatin and Smc3 acetyla-
by addition of Ca2+, which leads to activation of APC/CCdc20, tion occurred at the same time (Figure 2E) and that inhibition of
degradation of mitotic Cyclins, and mitotic exit (Figure 2E). As DNA replication had similar effects on both events, supporting
a source of chromatin, demembranated sperm nuclei were the notion that the two events are linked (Figure 2G). However,
added. DNA replication was monitored by incorporation of depletion of Sororin from Xenopus extracts or from HeLa cells
[a-32P]dCTP into DNA and occurred within 60 min after Ca2+ affected neither the kinetics nor the degree of Smc3 acetylation,
addition. After 90 min, we added a recombinant form of nonde- suggesting that Sororin is not required for cohesin acetylation
gradable Cyclin B (D90 Cyc B) to induce entry of the extract into (Figures S3B and S3C).
a mitotic state. At different time points, proteins in the chromatin To test whether Smc3 acetylation is required for the chromatin
fraction or the total extract were analyzed by immunoblotting association of Sororin, we depleted Esco1 and Esco2 from HeLa
(Figure 2E). As expected, Ca2+ addition led to rapid degradation cells. Only depletion of both enzymes reduced Smc3 acetylation,
of Cyclin B2 (a substrate of APC/CCdc20), but the levels of the indicating that Esco1 and Esco2 can both acetylate cohesin
APC/CCdh1 substrates Sororin and Plx1 remained largely (Figure 3A). To analyze whether depletion of Esco1 and Esco2
unchanged (only the electrophoretic mobility of Sororin was affects the association of Sororin with chromatin, we synchro-
reduced by phosphorylation in CSF and mitotic extracts). Impor- nized cells in S phase by double thymidine arrest-release and
tantly, even though Sororin was present throughout all stages of measured the amount of Sororin-LAP on chromatin by immuno-
the cell cycle, it began to associate with chromatin only 60 min blotting and IFM. We also depleted endogenous Sororin in these
after addition of Ca2+, i.e., when DNA replication was initiated. experiments to ensure that Sororin-LAP was analyzed under
In contrast, the cohesin subunits Scc1 and Smc3 could be de- conditions where it is functional. To rule out that reduced chro-
tected on chromatin at least 30 min earlier. The association of matin binding of Sororin was caused indirectly by a delay in
Sororin with chromatin was abolished by Geminin (Figure S2F), DNA replication, we labeled cells with BrdU and quantified
a protein that inhibits cohesin loading onto DNA (Gillespie and Sororin-LAP IFM signals only in cells that had similar amounts
Hirano, 2004; Takahashi et al., 2004), indicating that our assay of BrdU incorporated. Both by immunoblotting and IFM we
reflected physiological binding of Sororin to chromatin. These observed a reduction in Sororin on chromatin (Figures 3B–3D).
observations suggest that local APC/CCdh1-mediated degrada- Depletion of Esco1 and Esco2 also reduced the amount of
tion of Sororin on chromatin cannot explain why Sororin associ- endogenous Sororin that was associated with chromatin-bound
ates with chromatin later than cohesin. Instead, our results indi- cohesin (Figures S3D and S3E). Esco1 and Esco2 are therefore
cate that the presence of cohesin on chromatin is not sufficient required for efficient binding of Sororin to cohesin on chromatin.
for recruitment of Sororin. It is possible that the residual amounts of Sororin on chromatin
Because Sororin associates with chromatin during S phase in that were seen in our assays were due to incomplete depletion
Xenopus extracts and in somatic cells (Figure 1C and Figure 2E), of Esco1 and Esco2.
we tested whether DNA replication is required for recruitment of To address whether Esco1 and Esco2 regulate Sororin by
Sororin to chromatin. We prevented replication in Xenopus acetylating Smc3, we mutated K105 and K106 in Smc3 to either
extracts by addition of aphidicolin or actinomycin D. Aphidicolin glutamine (Smc3QQ), arginine (Smc3RR), or alanine (Smc3AA)
allows initiation of DNA replication but leads to the stalling of residues. Smc3QQ has been proposed to mimic acetylated and

Cell 143, 737–749, November 24, 2010 ª2010 Elsevier Inc. 741
Figure 3. Acetylation of Smc3 Facilitates
but Is Not Sufficient for the Association of
Sororin with Chromatin
(A) RNAi against both Esco1 and Esco2 causes
a decrease in Smc3 acetylation. HeLa cells were
transfected with siRNAs and harvested at S
phase. Chromatin-bound proteins were analyzed
by immunoblotting. Asterisks indicate nonspecific
signals.
(B) Reduction of Smc3 acetylation causes a
decrease of Sororin on chromatin. Sororin-LAP
HeLa cells were synchronized at S phase and
chromatin fractions were analyzed by immuno-
blotting.
(C) Cells in (B) were treated with BrdU after the
second thymidine release, pre-extracted, and
costained for BrdU, Sor-LAP (GFP), and DNA
(DAPI). Bar: 10 mm.
(D) Quantification of chromatin-associated
Sororin-LAP signal in cells with similar levels of
BrdU incorporation. Cells described in (C) with
similar BrdU intensities (left) were analyzed for
Sor-LAP intensity (right) (mean ± confidence
interval; *p < 0.01).
(E) Soluble Smc3QQ and Smc3RR proteins stably
bind to Sororin in HeLa cells. HeLa cells express-
ing Smc3WT-, Smc3QQ-, or Smc3RR-LAP were
synchronized in G2 phase, Smc3-LAP was immu-
noprecipitated from the soluble fraction of cells,
and the coprecipitated proteins were analyzed
by immunoblotting using a 2-fold serial dilution.
(F) Acetylation of Smc3 is not sufficient for Sororin
binding to chromatin. Interphase Xenopus egg
extracts were incubated with sperm chromatin
in the presence (Esco1) or absence (buffer) of
Esco1 for indicated times. Chromatin fractions
were analyzed by immunoblotting (on chromatin).
Extracts without sperm chromatin were incubated
for 120 min in the presence or absence of Esco1
(extracts).
See also Figure S3.

Smc3RR and Smc3AA to mimic nonacetylated Smc3. We mutations that alter K105 and K106 (for possible interpretations
mutated a LAP-tagged version of the Smc3 gene on a BAC, of these results, see Discussion).
stably integrated the modified BACs into HeLa cells, purified We also attempted to generate acetylated cohesin in vitro by
the wild-type and mutant forms of Smc3 either from soluble using recombinant purified Esco1 (Figure S3I). We observed
extracts or from chromatin, and analyzed their interaction part- that Esco1 could acetylate Smc3 when cohesin was associated
ners by immunoblotting and mass spectrometry. For wild-type with chromatin in a Xenopus extract, but not in extract lacking
Smc3-LAP, these experiments confirmed that Sororin only asso- chromatin or when Esco1 was incubated with purified soluble
ciates with cohesin on chromatin but not, or only to a small cohesin (Figure 3F and data not shown). Esco1 may therefore
degree, with soluble cohesin (Figure S3G). However, when only be able to modify cohesin on chromatin. Consistent with
Smc3QQ-LAP was purified, Sororin could also reproducibly be this possibility, endogenous acetylated forms of Smc3 could
found in association with soluble cohesin, consistent with the only be detected by immunoblotting in chromatin fractions (Fig-
possibility that Smc3 acetylation promotes binding of Sororin ure S3J), and quantitative mass spectrometry indicated that the
to cohesin (Figure 3E and Figure S3G). This interaction was abol- fraction of acetylated Smc3 relative to total Smc3 is 97-fold
ished by depletion of Scc1, indicating that Smc3QQ does not higher for chromatin-bound than for soluble cohesin (data not
simply represent a misfolded protein to which Sororin binds shown).
nonspecifically (Figure S3H). Unexpectedly, similar results When we added Esco1 to Xenopus extract containing chro-
were also obtained when Smc3RR and Smc3AA were analyzed matin, we observed that Smc3 acetylation was advanced by at
(Figures 3E, Figure S3F, and Figure S3G). This suggests that So- least 30 min, but Esco1 had no effect on the chromatin associa-
rorin-cohesin interactions can be stabilized not only by muta- tion of Sororin (Figure 3F), indicating that Smc3 acetylation is not
tions that might chemically mimic acetylation but also by other sufficient for recruitment of Sororin to chromatin. In support of

742 Cell 143, 737–749, November 24, 2010 ª2010 Elsevier Inc.
Figure 4. Sororin Is Dispensable for Cohe-
sion in the Absence of Wapl
(A) Chromatin fractions from mock-, Sororin-,
Wapl-, and Wapl- and Sororin-depleted inter-
phase extracts were analyzed by immunoblotting.
(B) D90 Cyclin B was added to the extracts shown
in (A) and mitotic chromosomes were assembled.
Chromosomes were isolated 120 min after D90
Cyclin B addition and stained for XCAP-E
(magenta) and Bub1 (green). Higher-magnification
images are shown in lower panels. Distance
between two chromosome arms stained by
XCAP-E in each extract is shown in a histogram
as a comparison to the mock-depleted extract.
Depletion of SA1/2 is shown as an example of
cohesin depletion. Bar: 5 mm.
(C) Codepletion of Sororin and Wapl in HeLa cells.
Cells were transfected with the indicated siRNAs
and treated with nocodazole. After mitotic shake-
off for chromosome spreads (D and E), residual
cells were harvested for immunoblotting. See
also Figure S4A.
(D) Analysis of chromosome spreads after Sororin
and Wapl depletion. Mitotic cells harvested as in
(C) were examined by chromosome spreading
and Giemsa staining. Five hundred cells per RNAi
experiment were classified into three categories.
(E) Representative pictures of the most prominent
phenotype class upon RNAi depletion in the
Giemsa spread analysis. Color code is shown in
(D). Bar: 10 mm.

2006; Rankin et al., 2005; Schmitz et al.,


2007; Shintomi and Hirano, 2009).
To understand the functional relation-
ship between Sororin and Wapl we
depleted both proteins either individually
or simultaneously from Xenopus extracts
and analyzed cohesion in mitotic chro-
mosomes. We analyzed chromosome
morphology by staining the condensin
subunit XCAP-E and Bub1 as markers
this hypothesis, we found that the association of Sororin with for sister chromatid arms and kinetochores, respectively. Deple-
Smc3QQ was still partially dependent on DNA replication (Fig- tion of Sororin alone increased the distance between sister chro-
ure S3K). Taken together, these results indicate that Smc3 acet- matids, indicating a partial cohesion defect (Figures 4A and 4B).
ylation is required but not sufficient for efficient recruitment of This defect was similar in magnitude to the defect that was
Sororin to chromatin-bound cohesin. observed after simultaneous depletion of the cohesin subunits
SA1 and SA2, suggesting that also in Xenopus extracts Sororin
Sororin Is Dispensable for Cohesion in the Absence is similarly important for cohesion as cohesin itself (Figure 4B).
of Wapl As expected, depletion of Wapl had the opposite effect, i.e., re-
Several previous observations are consistent with the possibility sulted in tightly connected chromatids. Remarkably, depletion of
that Sororin and Wapl have antagonistic functions: depletion of both proteins caused a phenotype that was very similar to the
Sororin and Wapl has opposite effects on cohesion (increased phenotype caused by depletion of Wapl alone. Similar results
and decreased proximity between sister chromatids, respec- were obtained when Sororin and Wapl were depleted singly or
tively) and on the stability of cohesin-DNA interactions simultaneously by RNAi from HeLa cells and mitotic chromo-
(decreased and increased residence times of cohesin on chro- somes were analyzed by Giemsa staining (Figures 4C–4E).
matin, respectively). Likewise, addition of excess Sororin to Xen- Also in this case, the phenotype obtained after codepletion of
opus extracts mimics the ‘‘overcohesion’’ phenotype caused by Sororin and Wapl was nearly identical to the phenotype obtained
depletion of Wapl, and overexpression of Wapl causes cohesion after depletion of Wapl alone, i.e., in the majority of mitotic cells
defects, as does loss of Sororin (Gandhi et al., 2006; Kueng et al., sister chromatids were more tightly associated with each other

Cell 143, 737–749, November 24, 2010 ª2010 Elsevier Inc. 743
Figure 5. The FGF Motif of Sororin Is
Required for Cohesion
(A and B) Pds5 is required for Sororin association
with chromatin. Interphase Xenopus egg extracts
were subjected to mock or Pds5A and B immuno-
depletion. Two hours after sperm chromatin addi-
tion, chromatin fractions were analyzed by immu-
noblotting (A). DNA replication in the extracts
shown in (A) was monitored for 30 or 60 min by
incorporation of [a-32P]dCTP into sperm chro-
matin (B).
(C) Sequence comparison in the region including
FGF motifs of vertebrate Sororin and fly Dalmatian.
Identical and similar residues are shaded in black
and gray, respectively. In Xenopus, Sororin-A is
shown. In fruit fly, the latter two of three FGF motifs
are shown (see also Figure 7A).
(D) Anti-Pds5A antibody beads were incubated
with Sororin-WT or -AA mutant in the presence
or absence of Pds5A protein. Beads-bound pro-
teins were analyzed by immunoblotting.
(E) Anti-Pds5A antibody beads were incubated
with Sororin-WT or -AA mutant in the presence or
absence of the Pds5A-Wapl heterodimer. Beads-
bound proteins were separated from the superna-
tant and were analyzed by immunoblotting.
(F) Wapl removal activity of Sororin is increased in
a dose-dependent manner. Increasing amounts
(10–40 ng/ml) of Sororin-WT or -AA mutant were
used in the experiment shown in (E), supernatant
fractions were analyzed by immunoblotting (left),
and signal intensity of Wapl was quantified (right).
(G) Sororin-depleted interphase extracts were
combined with buffer, Sororin-WT, or -AA mutant.
Two hours after sperm chromatin addition, chro-
matin fractions were analyzed by immunoblotting.
(H) D90 Cyclin B was added to the extracts shown
in (G) and mitotic chromosomes were assembled.
Chromosomes were isolated 120 min after
D90 Cyclin B addition and stained for XCAP-E.
Magnified images are shown in lower panels.
Distance between two chromosome arms stained
by XCAP-E is shown in lower histogram as a
comparison to mock-depleted extract. Bar: 5 mm.
See also Figure S4.

than normally. These observations indicate that Sororin is only and data not shown). Sororin antibodies also immunoprecipi-
required for cohesion in the presence of Wapl, and they therefore tated Pds5A and Pds5B from solubilized chromatin of HeLa cells
suggest that Sororin’s key function is to antagonize Wapl. (Figure S4B), and when we immunodepleted Pds5A and Pds5B
We also observed in these experiments that Wapl depletion from Xenopus extracts the binding of Sororin to chromatin was
greatly increased the degree of Smc3 acetylation (Figure 4C greatly reduced (Figure 5A). The latter effect was not caused
and Figure S4A). Wapl depletion could cause this effect by by a delay in DNA replication because [a-32P]dCTP incorporation
increasing the residence time of cohesin on DNA, but it is also into sperm DNA was unaffected by depletion of Pds5 proteins
possible that Wapl inhibits cohesin acetylation and that this func- (Figure 5B). These observations are consistent with the possi-
tion is required for Wapl’s ability to allow cohesin dissociation bility that the association of Sororin with cohesin depends on
from DNA. Pds5 proteins.
To address directly whether Sororin interacts with Pds5
Sororin Interacts with Pds5 via a Conserved FGF Motif proteins or Pds5-Wapl heterodimers, we purified recombinant
and Can Displace Wapl from Pds5 forms of human Sororin, Pds5A and Wapl. As predicted, Wapl
When we isolated Sororin-LAP via tandem affinity purification, bound efficiently to Pds5A, either when expressed simulta-
we identified cohesin core subunits and Pds5A and Pds5B, indi- neously in Baculovirus-infected insect cells or when incubated
cating that Sororin can directly bind to these proteins (Figure S1I with each other as individually purified proteins (Figure S4C

744 Cell 143, 737–749, November 24, 2010 ª2010 Elsevier Inc.
A B Figure 6. Phosphorylated Sororin Is Unable
supernatant beads bound
to Dissociate Wapl from Pds5

M-Sor λPP

M-Sor λPP
(A) Sororin is dissociated from Pds5 in mitosis.
− + : human Sororin Sororin-WT was incubated in either interphase (I)

M-Sor

M-Sor
buffer

buffer
I-Sor

I-Sor
I M I M or mitotic (M) Xenopus egg extracts and immuno-
precipitated, and the precipitates were analyzed
Pds5B
* Wapl
by immunoblotting. Asterisk indicates nonspecific
Sororin signal.
Sororin (B) Wapl removal activity is abolished by phosphor-
ylation of Sororin. Wapl-Pds5A heterodimer on anti-
Pds5A Pds5A antibody beads was incubated with either
C
buffer, Sororin preincubated in interphase egg
Telo/G1-phase S/G2-phase M-phase extract (I-Sor) or mitotic egg extract (M-Sor), or
l-protein phosphatase-treated M-Sor (M-Sor
l-PP). Beads-bound proteins were separated from
the supernatant and analyzed by immunoblotting.
(C) Model for the role of Sororin in sister chromatid
DNA replication
Smc3 acetylation cohesion. The cohesin complex is loaded onto
chromatin during telo/G1 phase, where Wapl-
mitotic entry orin
l

P
Sor
ap

Pds5 destabilizes cohesin binding to chromatin


W

Wapl Sororin Sororin Wapl in the absence of Sororin. During DNA replication
Pds5 Pds5 Pds5 in S phase, Sororin associates with chromatin
depending on cohesin and this association is facil-
FGF
itated by acetylation of Smc3. Sororin binds to
dynamic stable dynamic Pds5 through its FGF motif and thereby can antag-
onize the function of Wapl by modulating the
topology of Wapl and Pds5 so that stable cohesion
is maintained. Upon entry into mitosis, phosphory-
lation of Sororin abolishes the ability to inhibit
Wapl, allowing cohesin removal in prophase.

and data not shown). The interaction between Pds5 and Wapl when excess Sororin was added to Xenopus extracts from which
depends on two sequence elements composed of phenylala- the endogenous protein had not been depleted: in this assay
nine-glycine-phenylalanine (FGF) residues in Wapl (Shintomi wild-type Sororin causes an ‘‘overcohesion’’ phenotype (Rankin
and Hirano, 2009), and we noticed that a similar FGF motif is et al., 2005), but the AA mutant had no effect (Figure S4E). These
also present at a conserved position in all known Sororin results show that the FGF motif of Sororin is required for its func-
sequences (Figure 5C and see Figure S5B). We therefore also tion in cohesion, and they suggest that Sororin might execute
generated a Sororin mutant in which the two phenylalanine resi- this function by displacing Wapl from Pds5.
dues in this motif were changed to alanines (hereafter called However, we could not obtain evidence that the Sororin-
‘‘Sororin-AA’’). Wild-type Sororin associated with Pds5A, dependent displacement of Wapl from Pds5 results in the disso-
whereas the AA mutant bound less well (Figure 5D). Also, ciation of Wapl from chromatin. Addition of recombinant Sororin
when added to Xenopus extracts, wild-type Sororin associated to Xenopus extracts increased, and did not decrease, the
with cohesin and Pds5B more efficiently than the AA mutant (Fig- amount of Wapl and Pds5A on chromatin, as if Sororin stabilized
ure S4D). When we performed Sororin-binding experiments with the interactions between Pds5A-Wapl and cohesin, rather than
Pds5A-Wapl, we observed, remarkably, that Sororin displaced dissociating Wapl from cohesin (Figure S4F). It is therefore
some Wapl from the Pds5A-Wapl heterodimers. Also, this effect possible that the Sororin-mediated displacement of Wapl from
was reduced when the AA mutant was used (Figures 5E and 5F). Pds5A causes a rearrangement in the topology of cohesin-asso-
These observations raised the possibility that Sororin regulates ciated proteins and does not lead to dissociation of Wapl from
cohesin by interacting with the Pds5-Wapl heterodimer. cohesin.

The FGF Motif of Sororin Is Essential for Its Cohesion Sororin Is Inactivated by Phosphorylation in Mitosis
Function The prophase pathway of cohesin dissociation depends on Wapl
To address whether Sororin’s ability to displace Wapl from Pds5 (Gandhi et al., 2006; Kueng et al., 2006). It is therefore conceiv-
is of functional relevance, we replaced Sororin in Xenopus able that Sororin has to be inactivated at the onset of mitosis
extracts by the Sororin-AA mutant and analyzed its effect on to relieve Wapl from its inhibition by Sororin. We therefore
cohesion. We immunodepleted Sororin from interphase egg analyzed whether Sororin’s ability to dissociate Wapl from
extracts, added either buffer, recombinant wild-type Sororin, Pds5 proteins is cell cycle regulated. Consistent with this possi-
or the AA mutant, and analyzed mitotic chromosomes as above. bility, recombinant Sororin could associate with Pds5B in Xeno-
Importantly, the cohesion defect observed after Sororin deple- pus interphase extracts but not in mitotic extracts where Sororin
tion could be restored by wild-type Sororin but not by the AA is phosphorylated (Figure 6A). Furthermore, we observed that
mutant (Figures 4G and 4H). Similar results were obtained Sororin could bind to recombinant purified Wapl-Pds5A

Cell 143, 737–749, November 24, 2010 ª2010 Elsevier Inc. 745
Figure 7. Dalmatian Is an Ortholog of So-
rorin in Drosophila
(A) Schematic sequence comparison of human
and Xenopus Sororin and Drosophila Dalmatian.
The conserved regions are shaded in gray and
KEN-box and FGF motifs are depicted with white
and black boxes, respectively.
(B) Dalmatian (Dmt) RNAi causes premature sister
chromatid separation in S2 cells. Cells were trans-
fected with dsRNA against Dmt or BubR1 or were
left untransfected (control). Chromosome spreads
were stained with DAPI. Representative images
are shown. Bar: 5 mm.
(C) Cells described in (B) were quantified for loss of
cohesion. Error bars denote standard deviations
between three independent experiments.
(D) Mitotic defects in Dalmatian knockdown cells.
Cells were transfected with dsRNA against Dmt
or BubR1 or were untransfected (control) and
costained for a-tubulin and Cyclin B to define
mitotic stages, CID (Cenp-A in Drosophila) to
assess centromere pairing, and DAPI (upper
panel). The lower table summarizes the observed
phenotype over all mitotic cells (n > 59 per condi-
tion). Bar: 5 mm.
See also Figure S5 and Table S1.

sequences in vertebrates and one


distantly related protein in the mollusc
Lottia gigantea. We used the C-terminal
portion of these sequences, where the
highest degree of similarity is found, to
perform iterative rounds of similarity
searches in invertebrate proteome data-
bases. We identified a single sequence
with significant similarity to Sororin in 18
different metazoan species belonging to
different taxa, including cephalochor-
dates, echinoderms, insecta, cnidaria,
and placozoa. All of these proteins
contain sequences related to the C
terminus of Sororin, which we therefore
heterodimers and dissociate Wapl from Pds5A when Sororin call the ‘‘Sororin domain’’ (Figure S5A). Furthermore, 17 of these
was preincubated in Xenopus interphase extracts but not when proteins also contain an FGF sequence motif (Figure S5B), or
Sororin had been incubated in a mitotic extract (Figure 6B). sometimes several of these motifs (Figure 7A).
The Wapl dissociation activity of mitotic Sororin was fully Of the 18 hypothetical proteins, only one has previously been
restored when Sororin was first dephosphorylated by l-protein characterized. This is a 95 kDa protein called Dalmatian, which is
phosphatase. These results suggest that Sororin phosphoryla- required for development of the Drosophila embryonic peripheral
tion in mitosis relieves Wapl from inhibition by Sororin (Figure 6C; nervous system (Prokopenko et al., 2000). Recent RNAi screens
for further discussion of this model see below). have shown that depletion of Dalmatian causes defects in mitotic
spindle assembly, chromosome alignment, and cell division
Dalmatian Is a Drosophila Ortholog of Sororin (Goshima et al., 2007; Somma et al., 2008). Dalmatian inactiva-
Wapl orthologs exist in species from yeast to human (Kueng tion also causes precocious sister chromatid separation in the
et al., 2006), but Sororin has only been identified in vertebrates presence of colchicine, a compound that activates the SAC. It
(Rankin et al., 2005). To address whether inhibition of Wapl by has therefore been proposed that Dalmatian is required for the
Sororin could also be required for cohesion in nonvertebrate SAC (Somma et al., 2008).
species, we searched for Sororin-related sequences in inverte- Because Dalmatian shares sequence similarity with Sororin,
brate genomes (Table S1). BLAST searches identified Sororin we tested whether Dalmatian is required for cohesion. If this

746 Cell 143, 737–749, November 24, 2010 ª2010 Elsevier Inc.
were the case, Dalmatian depletion would be predicted to cause Sororin recruitment are blocked less efficiently by aphidicolin
precocious sister chromatid separation, to activate the SAC, and and thymidine, in whose presence helicase progression can still
thus to cause an increase in mitotic index, whereas inactivation occur, it is possible that Smc3 acetylation and Sororin binding
of a SAC protein would shorten mitosis and cause a decrease are coupled to helicase progression. Within the cohesin
in mitotic index. We observed a modest increase in mitotic index complex, Sororin binds to Pds5 via an FGF sequence motif
from 3.2% in control Drosophila S2 cells to 5.3% in Dalmatian that is shared between Sororin and Wapl. Sororin displaces
RNAi cells, whereas depletion of BubR1, a protein required for Wapl from Pds5, but not from cohesin, suggesting that Sororin
the SAC (Perez-Mongiovi et al., 2005), decreased the mitotic induces a rearrangement in the topology of these cohesin-
index to 1.4% (data not shown). Chromosome spreading re- associated proteins. We propose that these changes inhibit
vealed that cohesion had been lost in 82% of all mitotic Dalma- Wapl’s ability to dissociate cohesin from DNA, and that the re-
tian RNAi cells, but only in less than 6% of mitotic control or sulting stable interaction of cohesin with DNA enables cohesin
BubR1 RNAi cells (Figures 6B and 6C). In IFM experiments, we to mediate cohesion. Our data further indicate that in prophase,
observed that Dalmatian depletion caused chromosome con- Sororin is inactivated by phosphorylation, enabling Wapl to
gression defects (‘‘scattered chromosomes’’) in 57.6% of prom- dissociate cohesin from mitotic chromosomes. Later in telo-
eta/metaphase cells (Figure 6D). Many of the scattered chromo- phase and G1, APC/CCdh1 targets Sororin for degradation. The
somes were single sister chromatids, as judged by staining of function of this process remains to be understood, but it might
the centromere protein centromere identifier (CID), and Cyclin ensure that Sororin associates with cohesin only after the initia-
B levels were similarly high in cells with scattered chromatids tion of DNA replication once APC/CCdh1 has been inactivated.
as in control prometaphase cells. Because SAC defects would This model makes a number of important predictions: (1) If So-
lead to precocious APC/CCdc20 activation and Cyclin B degrada- rorin is an antagonist of Wapl, one would expect that Sororin or-
tion, these results indicate that Dalmatian depletion does not thologs can be identified in species where Wapl exists. We show
inactivate the SAC. Instead, our results suggest that Dalmatian that this is indeed the case for many metazoans, including
is a distant ortholog of Sororin that is required for cohesion. species from evolutionarily old taxa such as cnidaria (jellyfish)
and placozoa, the simplest known metazoa. Our observation
DISCUSSION that depletion of the Drosophila member of this protein family
(Dalmatian) causes cohesion defects suggests that these
Although establishment and maintenance of sister chromatid proteins are also functionally related to Sororin. We have so far
cohesion are essential for chromosome segregation, it is poorly not been able to identify Sororin-related proteins in worms or
understood how cohesin generates cohesive structures during yeast. It therefore remains to be seen whether Sororin is required
DNA replication and how these are maintained for hours, or in for cohesion in all eukaryotes, or whether some species have
the case of mammalian oocytes even for years. Recent studies evolved cohesion mechanisms that are independent of Sororin.
have revealed that both the stability of cohesin-DNA interactions (2) If the key function of Sororin is to inhibit Wapl, then Sororin
(Gerlich et al., 2006) and the acetylation state of cohesin change is expected to be dispensable in the absence of Wapl. Our
during DNA replication (Ben-Shahar et al., 2008; Rowland et al., results indicate that this is indeed the case. An interesting impli-
2009; Unal et al., 2008; Zhang et al., 2008), suggesting that cohe- cation of this result is that Sororin might not be essential for the
sion is not simply established by doubling the number of sister initial entrapment of sister chromatids by cohesin rings, i.e., for
chromatids inside otherwise unchanged cohesin rings. Our cohesion establishment, at least in the absence of Wapl. It is
results further extend this view by showing that also the compo- therefore possible that Sororin’s main function is to prevent
sition of cohesin complexes changes during DNA replication dissociation of cohesin from DNA, rather than enabling opening
through the recruitment of Sororin, and importantly our data and closure of the ring around DNA. However, the situation could
suggest that only Sororin-associated cohesin complexes are be different in yeast where deletion of WAPL/RAD61 does not
able to mediate cohesion. Consistent with this view, we find result in accumulation of cohesin on DNA but has the opposite
that Sororin is the only known protein whose presence on chro- effect, a reduction of cohesin on chromatin (Rowland et al.,
matin coincides precisely with the periods of the cell cycle during 2009; Sutani et al., 2009). If a Sororin-related Wapl/Rad61 antag-
which cohesion exists (from initiation of DNA replication to meta- onist exists in yeast, such a protein (or protein domain) might
phase), whereas cohesin binds to DNA long before cohesion is therefore instead be needed for cohesion establishment by
established. having to overcome the proposed ‘‘anti-establishment’’ activity
Based on our results, we propose the following model for how of Wapl/Rad61 (Rowland et al., 2009; Sutani et al., 2009).
Sororin enables cohesin to become ‘‘cohesive’’ (Figure 6C): (3) If the stable postreplicative association of cohesin with
Smc3 acetylation and possibly other unidentified events during DNA was due to inhibition of Wapl by Sororin, depletion of
DNA replication promote the recruitment of Sororin to chro- Wapl should enable cohesin to bind to DNA also stably before
matin-bound cohesin. These events might occur directly at repli- Sororin has been recruited to cohesin, i.e., in G1 phase. At vari-
cation forks because Eco1 has been detected at these sites ance with this prediction, we observed previously that depletion
(Lengronne et al., 2006), Smc3 can only be acetylated on chro- of Wapl from HeLa cells increased the residence time of dynam-
matin (Unal et al., 2008; this study), and actinomycin D, ically bound cohesin complexes only modestly, from 8 min in
a compound that inhibits DNA polymerase and MCM helicase control cells to 18 min (Kueng et al., 2006), and not to many
progression (Pacek and Walter, 2004), prevents both Smc3 acet- hours, as is normally seen for cohesin complexes in G2 phase
ylation and Sororin recruitment. Because Smc3 acetylation and (Gerlich et al., 2006). However, we have in the meantime

Cell 143, 737–749, November 24, 2010 ª2010 Elsevier Inc. 747
measured the residence time of cohesin on chromatin in mouse 5 mM MgCl2, 100 mM KCl, HEPES-KOH pH 7.5) containing 0.25% Triton X-
embryonic fibroblasts from which the Wapl gene has been 100, overlaid onto a 30% sucrose/EB cushion, and spun at 15,000 g for 10
min. The pellets were washed with EB containing 0.25% Triton X-100 and re-
deleted, and in which therefore a more complete depletion of
suspended in SDS sample buffer.
Wapl can be achieved than by RNAi. In these cells the residence
time of cohesin on chromatin is increased from minutes to
SUPPLEMENTAL INFORMATION
several hours even before S phase (A. Tedeschi, personal
communication), indicating that it is indeed the presence of Supplemental Information includes Extended Experimental Procedures, five
Wapl that enables cohesin to interact with DNA dynamically figures, and one table and can be found with this article online at doi:10.
before replication. This result supports the hypothesis that inhi- 1016/j.cell.2010.10.031.
bition of Wapl by Sororin enables stable binding of cohesin to
DNA in postreplicative cells. ACKNOWLEDGMENTS
Our model also raises several important new questions. One of
We are grateful to O. Hudecz, P. Huis in’t Veld, I. Poser, and M. Sykora for
them is whether the essential function of Smc3 acetylation is to assistance and reagents; to G. Karpen, J. Knoblich, C. Lehner, and C. Sunkel
recruit Sororin, or whether this modification has other important for reagents and advice on Drosophila experiments; and to N. Kraut for BI
effects, for example on the ATPase activity of Smc3. The 2536. T.N. is supported by the European Molecular Biology Organization
absence of Sororin in yeast would suggest that cohesin acetyla- (EMBO) and the Japanese Society for the Promotion of Science (JSPS). K.S.
tion must have other essential functions, but given the low is supported by Grant-in-Aid for Scientific Research (S). Research in the
sequence similarity among Sororin orthologs it cannot be groups of J.-M.P. and K.M. is supported by Boehringer Ingelheim and the
Austrian Science Fund via the special research program ‘‘Chromosome
excluded that Sororin-related proteins also exist in yeast.
Dynamics’’ (F34-B03). Work in the groups of J.-M.P., K.M., and A.A.H. was
A related important question is how Smc3 acetylation might also supported by the EC via the Integrated Project ‘‘MitoCheck.’’
promote recruitment of Sororin. As Pds5 proteins are required
for the recruitment of Sororin to cohesin, and Sororin binds to Received: May 20, 2010
Pds5 proteins, we suspect that Smc3 acetylation promotes So- Revised: August 30, 2010
rorin binding indirectly, possibly by causing changes in how Accepted: October 21, 2010
Published: November 24, 2010
Pds5 or Wapl interact with cohesin or each other. Likewise, it
is unclear why replacement of K105/106 to not only glutamine
REFERENCES
(which is believed to mimic acetylated lysine) but also to arginine
or alanine residues can stabilize cohesin-Sororin interactions. It Ben-Shahar, T., Heeger, S., Lehane, C., East, P., Flynn, H., Skehel, M., and
is possible that it is not the presence of acetyl residues on Uhlmann, F. (2008). Eco1-dependent cohesin acetylation during establish-
K105/106 that creates a binding site, for example for a cohesin ment of sister chromatid cohesion. Science 321, 563–566.
subunit, but that any mutation that removes lysines at these posi- Diaz-Martinez, L.A., Gimenez-Abian, J.F., and Clarke, D.J. (2007). Regulation
tions will destroy a binding site or pocket, which would lead to of centromeric cohesion by sororin independently of the APC/C. Cell Cycle 6,
714–724.
subunit rearrangements that would facilitate Sororin recruitment.
A more detailed characterization of how cohesin interacts with Gandhi, R., Gillespie, P.J., and Hirano, T. (2006). Human Wapl is a cohesin-
binding protein that promotes sister-chromatid resolution in mitotic prophase.
Wapl, Pds5, and Sororin will be required to address these
Curr. Biol. 16, 2406–2417.
questions.
Gerlich, D., Koch, B., Dupeux, F., Peters, J.M., and Ellenberg, J. (2006). Live-
cell imaging reveals a stable cohesin-chromatin interaction after but not before
EXPERIMENTAL PROCEDURES DNA replication. Curr. Biol. 16, 1571–1578.
Gillespie, P.J., and Hirano, T. (2004). Scc2 couples replication licensing to
Immunodepletion and Monitoring of DNA Replication in Xenopus
sister chromatid cohesion in Xenopus egg extracts. Curr. Biol. 14, 1598–1603.
Egg Extracts
For immunodepletion of Xenopus egg extracts, affinity-purified antibody (70 Goshima, G., Wollman, R., Goodwin, S.S., Zhang, N., Scholey, J.M., Vale,
mg anti-Sororin, mixture of 40 mg anti-Pds5A and 25 mg anti-Pds5B, 200 mg R.D., and Stuurman, N. (2007). Genes required for mitotic spindle assembly
anti-Wapl, or 250 mg anti-SA1/2) was conjugated to 30 ml Affi-Prep Protein A in Drosophila S2 cells. Science 316, 417–421.
Matrix (Bio-Rad), mixed with 100 ml interphase extracts, incubated for 30 Gruber, S., Haering, C.H., and Nasmyth, K. (2003). Chromosomal cohesin
min for Sororin depletion or 1 hr for Pds5A/B, Wapl, and SA1/2 depletions forms a ring. Cell 112, 765–777.
on ice, and beads were removed by centrifugation. For add-back experiments, Hou, F., and Zou, H. (2005). Two human orthologues of Eco1/Ctf7 acetyltrans-
Sororin wild-type or AA mutant (F166A, F168A) was added to Sororin-depleted ferases are both required for proper sister-chromatid cohesion. Mol. Biol. Cell
extracts at 6.5 nM. 16, 3908–3918.
DNA replication was monitored by the incorporation of [a-32P]dCTP into
Kueng, S., Hegemann, B., Peters, B.H., Lipp, J.J., Schleiffer, A., Mechtler, K.,
DNA. Demembranated sperm nuclei (2000 nuclei/ml) were added to egg
and Peters, J.M. (2006). Wapl controls the dynamic association of cohesin with
extract containing [a-32P]dCTP (3.7 kBq/ml), incubated at 22 C, and the reac-
chromatin. Cell 127, 955–967.
tion stopped by addition of 2 volumes of stop solution (8 mM EDTA, 0.13%
phosphoric acid, 10% Ficoll, 5% SDS, 0.2% bromophenol blue, 80 mM Tris- Lengronne, A., McIntyre, J., Katou, Y., Kanoh, Y., Hopfner, K.P., Shirahige, K.,
HCl pH 8.0). The mixture was incubated with 2 mg/ml Proteinase K for 30 and Uhlmann, F. (2006). Establishment of sister chromatid cohesion at the
min at 37 C and analyzed by agarose gel electrophoresis followed by S. cerevisiae replication fork. Mol. Cell 23, 787–799.
autoradiography. Nasmyth, K., and Haering, C.H. (2009). Cohesin: its roles and mechanisms.
Annu. Rev. Genet. 43, 525–558.
Preparation of Xenopus Chromatin Fractions Onn, I., Heidinger-Pauli, J.M., Guacci, V., Unal, E., and Koshland, D.E. (2008).
Sperm nuclei were incubated in extracts at a concentration of 2000 nuclei/ml. Sister chromatid cohesion: a simple concept with a complex reality. Annu.
Thirty microliters of extract was diluted 10-fold with ice-cold extract buffer (EB; Rev. Cell Dev. Biol. 24, 105–129.

748 Cell 143, 737–749, November 24, 2010 ª2010 Elsevier Inc.
Pacek, M., and Walter, J.C. (2004). A requirement for MCM7 and Cdc45 in Schmitz, J., Watrin, E., Lenart, P., Mechtler, K., and Peters, J.M. (2007).
chromosome unwinding during eukaryotic DNA replication. EMBO J. 23, Sororin is required for stable binding of cohesin to chromatin and for sister
3667–3676. chromatid cohesion in interphase. Curr. Biol. 17, 630–636.
Perez-Mongiovi, D., Malmanche, N., Bousbaa, H., and Sunkel, C. (2005). Shintomi, K., and Hirano, T. (2009). Releasing cohesin from chromosome arms
Maternal expression of the checkpoint protein BubR1 is required for in early mitosis: opposing actions of Wapl-Pds5 and Sgo1. Genes Dev. 23,
synchrony of syncytial nuclear divisions and polar body arrest in Drosophila 2224–2236.
melanogaster. Development 132, 4509–4520.
Somma, M.P., Ceprani, F., Bucciarelli, E., Naim, V., De Arcangelis, V.,
Peters, J.M. (2006). The anaphase promoting complex/cyclosome: a machine
Piergentili, R., Palena, A., Ciapponi, L., Giansanti, M.G., Pellacani, C., et al.
designed to destroy. Nat. Rev. Mol. Cell Biol. 7, 644–656.
(2008). Identification of Drosophila mitotic genes by combining co-expression
Peters, J.M., Tedeschi, A., and Schmitz, J. (2008). The cohesin complex and its
analysis and RNA interference. PLoS Genet. 4, e1000126.
roles in chromosome biology. Genes Dev. 22, 3089–3114.
Poser, I., Sarov, M., Hutchins, J.R., Heriche, J.K., Toyoda, Y., Pozniakovsky, Sutani, T., Kawaguchi, T., Kanno, R., Itoh, T., and Shirahige, K. (2009). Budding
A., Weigl, D., Nitzsche, A., Hegemann, B., Bird, A.W., et al. (2008). BAC Trans- yeast Wpl1(Rad61)-Pds5 complex counteracts sister chromatid cohesion-
geneOmics: a high-throughput method for exploration of protein function in establishing reaction. Curr. Biol. 19, 492–497.
mammals. Nat. Methods 5, 409–415. Takahashi, T.S., Yiu, P., Chou, M.F., Gygi, S., and Walter, J.C. (2004). Recruit-
Prokopenko, S.N., He, Y., Lu, Y., and Bellen, H.J. (2000). Mutations affecting ment of Xenopus Scc2 and cohesin to chromatin requires the pre-replication
the development of the peripheral nervous system in Drosophila: a molecular complex. Nat. Cell Biol. 6, 991–996.
screen for novel proteins. Genetics 156, 1691–1715.
Unal, E., Heidinger-Pauli, J.M., Kim, W., Guacci, V., Onn, I., Gygi, S.P., and
Rankin, S., Ayad, N.G., and Kirschner, M.W. (2005). Sororin, a substrate of the Koshland, D.E. (2008). A molecular determinant for the establishment of sister
anaphase-promoting complex, is required for sister chromatid cohesion in chromatid cohesion. Science 321, 566–569.
vertebrates. Mol. Cell 18, 185–200.
Rowland, B.D., Roig, M.B., Nishino, T., Kurze, A., Uluocak, P., Mishra, A., Waizenegger, I.C., Hauf, S., Meinke, A., and Peters, J.M. (2000). Two distinct
Beckouet, F., Underwood, P., Metson, J., Imre, R., et al. (2009). Building sister pathways remove mammalian cohesin from chromosome arms in prophase
chromatid cohesion: Smc3 acetylation counteracts an antiestablishment and from centromeres in anaphase. Cell 103, 399–410.
activity. Mol. Cell 33, 763–774. Zhang, J., Shi, X., Li, Y., Kim, B.J., Jia, J., Huang, Z., Yang, T., Fu, X., Jung,
Sakuno, T., and Watanabe, Y. (2009). Studies of meiosis disclose distinct roles S.Y., Wang, Y., et al. (2008). Acetylation of Smc3 by Eco1 is required for
of cohesion in the core centromere and pericentromeric regions. Chromosome S phase sister chromatid cohesion in both human and yeast. Mol. Cell 31,
Res. 17, 239–249. 143–151.

Cell 143, 737–749, November 24, 2010 ª2010 Elsevier Inc. 749
Nonenzymatic Rapid Control
of GIRK Channel Function
by a G Protein-Coupled Receptor Kinase
Adi Raveh,1 Ayelet Cooper,1 Liora Guy-David,1 and Eitan Reuveny1,*
1Department Biological Chemistry Weizmann Institute of Science, Rehovot 76100, Israel

*Correspondence: e.reuveny@weizmann.ac.il
DOI 10.1016/j.cell.2010.10.018

SUMMARY cascade, where activated receptors are initially phosphorylated


by GPCR kinases (GRKs), to initiate intracellular events leading
G protein-coupled receptors (GPCRs) respond to to a clathrin-mediated endocytosis of the GPCRs. This process
agonists to activate downstream enzymatic path- occurs over a time scale of many minutes to hours.
ways or to gate ion channel function. Turning off In the context of GPCR-mediated regulation of ion channel
GPCR signaling is known to involve phosphorylation activity, short-term desensitization to an activating signal has
of the GPCR by GPCR kinases (GRKs) to initiate their been observed. For instance, regulation of GPCR-controlled
excitability through the activation of the G protein-coupled
internalization. The process, however, is relatively
potassium channels (GIRK/Kir3.x), displays short-term desensi-
slow and cannot account for the faster desensitiza- tization characterized by a reduction in channel currents in the
tion responses required to regulate channel gating. presence of the receptor agonist in a time scale of few seconds
Here, we show that GRKs enable rapid desensitiza- (Sickmann and Alzheimer, 2003). This short-term reduction in
tion of the G protein-coupled potassium channel postsynaptic GIRK channel activity is independent of elements
(GIRK/Kir3.x) through a mechanism independent of that are known to affect the G protein cycle and PtdIns(4,5)P2
their kinase activity. On GPCR activation, GRKs hydrolysis. It is, therefore, of great interest to identify the molec-
translocate to the membrane and quench channel ular mechanism that mediates this process.
activation by competitively binding and titrating G We set out to identify the mechanism responsible for short-
protein bg subunits away from the channel. Of term desensitization of GIRK channels. We found that for some
interest, the ability of GRKs to effect this rapid desen- GPCRs, continued activation of their receptors leads to GIRK
current desensitization (GCD). This current desensitization is
sitization depends on the receptor type. The findings
enhanced in the presence of GRK2 and, surprisingly, does not
thus reveal a stimulus-specific, phosphorylation-
involve its kinase activity, but rather depends on its ability to
independent mechanism for rapidly downregulating bind the Gbg subunits of the G protein. This binding appears
GPCR activity at the effector level. to compete for the available pool of the G protein subunits that
activate the channel and hence to effectively quench channel
activity. These findings assign a new role for the GRK proteins
INTRODUCTION in providing negative feedback control of GPCR function at the
effector level.
G protein-coupled receptors (GPCR) modulate the activity of
enzymes and ion channels to fine tune cellular activity (Pierce
et al., 2002). To avoid abnormal cellular activity, GPCR-mediated RESULTS
G protein cycles should be temporally precise. Several mecha-
nisms guarantee the precise length of GPCR activation by GRK2 Accelerates Desensitization of GIRK Currents
controlling the levels of agonist. For example, the level of free Induced by A1R and mOR, but Not by mGluR2 and M4R
neurotransmitters present in the synapse are limited by fast We set out to test the involvement of GRK2 in mediating short-
neurotransmitter reuptake at the presynaptic site (Torres et al., term desensitization of GIRK channels. GRK2 is involved in the
2003), or degradation at the synaptic cleft (Massoulie et al., desensitization of GPCRs after exposure to their agonists. For
1993). These processes are specific for specific types of ligands. this purpose we expressed GIRK1, GIRK4 (for now on referred
For regulation at a longer time scale, additional mechanisms as GIRK channels) and adenosine type 1 receptor (A1R) with or
control GPCR signaling efficacy. These mechanisms control without (control) GRK2 in HEK293 cells, and used whole cell
the robustness of the activation signals by regulating receptor patch-clamp recordings to measure various channel current
number at the plasma membrane, in a process termed downre- parameters after receptor activation by adenosine (Figure 1A).
gulation (Bunemann et al., 1999; Tsao and von Zastrow, 2000). After A1R activation by adenosine (100 mM), GIRK channel
This mechanism involves a receptor-mediated signaling currents desensitize (GCD) as evident from the monoexponential

750 Cell 143, 750–760, November 24, 2010 ª2010 Elsevier Inc.
60 Figure 1. GRK2 Accelerates the Desensiti-
A C
zation of GIRK Currents Induced by A1R,
but Not by mGluR2
40 (A) GIRK channel currents induced by the activa-
tion of A1R rapidly desensitize in the presence of
A1R

GRK2.
+GRK2

20 (B) GIRK channel currents induced by mGluR2


activation are insensitive to GRK2.
(C) Bar plot that depicts GCD rates of cells acti-
0 vated with A1R or mGluR2 without or with GRK2,
B GRK2 shRNA, or nontarget (NT) shRNA.
(D) Bar plot compares the normalized expression
levels of GRK2 in silenced and NT cells as de-
picted from western blot for GRK2 (inset).
mGluR2

(E) GIRK current traces induced by adenosine in


A1R mGluR2 control HL-1 cell (black) and of siRNA#1 silenced
D cell (gray).
100 shRNA NT
(F) Bar plot depicting GCD in HL-1 cells trans-
GRK2
+GRK2

fected with two independent siRNAs, NT, and


GK2 (%)

GFP siRNA#1 transfected cells rescued by the expres-


50 sion of silently mutated GRK2GFP (smGRK2GFP).
1000pA (G) GRK2 mRNA quantification in HL-1 cells trans-
10s fected with two independent siRNAs or NT control.
See also Figure S1.
0
E F
shGRK2 NT
100
% Current (@ 2 m)

80
NT respectively for mGluR2, and 37.7 ±
60
siGRK2 #1 10.7 s, n = 7 and 33.4 ± 11.7 s, n = 6,
40 respectively, for M4R. Like in the case
shown above for GRK2, GRK3, but not
20
GRK6, also accelerated GCD in a similar
0 receptor-specific manner (data not
NT siGRK2 #1 siGRK2 #2 siGRK2 #1 + shown).
100 pA G smGRK2GFP
10s 100 Rescue Because GRK2 is endogenously ex-
(%, normalized)

80 pressed in HEK293 cells (Violin et al.,


GRK2 RNA

2006), we were interested to know


60
whether there is a contribution of the
40
endogenous protein to current desensiti-
20 zation in cells not transfected with GRK2.
0 To address this question we silenced
NT siGRK2 #1 siGRK2 #2 endogenous GRK2 levels using shRNA
specific for the human GRK2 (shGRK2).
GRK2 expression levels were reduced
decay curve of the current traces with a time constant of 24.9 ± by 58%, as determined using western blot (Figure 1D). A1R-in-
11.1 s, n = 8 (Figures 1A, upper trace, and 1C). Interestingly, in duced GIRK currents were significantly slower in GRK2-silenced
cells cotransfected with GRK2, GCD rates were accelerated cells (42.9 ± 6.8 s, n = 12) in comparison with cells cotransfected
10-fold, to 2.6 ± 0.0 s, n = 9 (p < 0.05). To assess whether with nontarget (NT) shRNA (26.0 ± 4.5 s, n = 12) (Figure 1C), con-
the enhancement of current desensitization was a general firming that endogenous levels of GRK2 are sufficient to enhance
phenomena to all PTX-sensitive GPCRs, we also tested GCD GCD rate after A1R simulation. The above results suggest that
rates induced by m-opioid receptor (mOR). Similar to the effect GRK2 has a role in modulating current desensitization rates of
of GRK2 on A1R-mediated GCD, mOR activation (methionine GIRK currents in a receptor-selective manner.
enkephalin, ME, 100 nM) accelerated GCD in the presence of To study whether GRK is also involved in GCD in cells that
GRK2 compared to control cells, with a time constant of 38.9 ± natively express GIRK, A1R and the kinase, we measured
5.9 s, n = 10 and 64.4 ± 6.18 s, n = 7, respectively (see Figures GIRK currents in HL-1 cells. HL-1 is a mouse cardiac muscle
S1A and S1C available online). In contrast, activation of GIRK cell-line that maintains the characteristics of adult cardiac myo-
channels in the absence or presence of GRK2 by metabotropic cytes, including contraction (Claycomb et al., 1998). These cells
glutamate type 2 receptor (mGluR2) (Figures 1B and 1C) or express both GIRK channels and the necessary components for
muscarinic acetylcholine type 4 receptor (M4R) activation their activation (Nobles et al., 2010). GIRK currents of HL-1 cells,
(Figures S1B and S1C) did not show any acceleration in GCD, where GRK2 was silenced using two independent siRNAs,
with time constants 41.7 ± 8.6 s, n = 9 and 41.7 ± 9.5 s, n = 9, (siGRK2#1 and siGRK2#2) displayed significantly smaller

Cell 143, 750–760, November 24, 2010 ª2010 Elsevier Inc. 751
desensitizations compared to cells transfected with NT (Figures GPCR Phosphorylation and Receptor Downregulation
1E and 1F). After continuous application of adenosine, the Are Not Required for GRK2-Mediated GIRK Current
induced currents were reduced to 79.2 ± 11.0% (n = 6), 86.3 ± Desensitization
7.3% (n = 5) and 24.7 ± 7.4% (n = 6) at 2 min, for both silenced In the traditional view, after translocation to the membrane,
and NT cells, respectively. Expression of silently mutated GRKs are responsible for the phosphorylation of activated
GRK2-GFP (smGRK2-GFP) in cells silenced with siGRK2#1 GPCRs. This event initiates the process of receptor downregula-
rescued the reduction in current desensitization (31.5 ± 12.5%, tion by clathrin-mediated endocytosis (Tsao and von Zastrow,
n = 4) to levels comparable to NT cells (Figure 1F). Similarly, 2000). To examine the relationship between this process and
GRK2 mRNA levels were reduced in cells transfected with either the apparent GRK2-mediated acceleration in GCD as shown
siGRK2#1 or siGRK2#2 compared to NT control cells with 54.0 ± above, we tested the ability of GRK2/K220R (dnGRK2), a domi-
2.4% and 57.1 ± 0.6%, respectively (Figure 1G). Qualitatively nant negative mutant that lacks kinase catalytic activity (Kong
similar results were obtained using primary mouse hippocampal et al., 1994), in accelerating GCD rates (Figure 3A). The GCD
neurons (Figure S1). These experiments suggest that, qualita- rates of cell cotransfected with GIRK, A1R, and dnGRK2 (5.5 ±
tively, the effect of GRK in HEK cells is relevant at physiological 1.1 s, n = 9) were not different from cells expressing GRK2, the
expression levels, and is not due to overexpression of GRK, the receptor and channel components, with t of 2.6 ± 0.0 s (n = 9),
receptors or the channels. and significantly faster than in cells that were not cotransfected
with the kinase (24.9 ± 11.1 s; n = 8). These results suggest that
A1R Activation Recruits GRK2-GFP to the Membrane the enhancement of GCD rates is not mediated via the kinase
Simultaneously with GIRK Current Desensitization, activity of GRK2.
but Not mGluR2 Another possible mechanism for enhancing GCD might be
GRK2 is mainly cytosolic and translocates to the membrane to a change in receptor number, independent of GRK2-mediated
phosphorylate active receptors (Pitcher et al., 1998). We wanted phosphorylation, or channel number, at the plasma membrane.
to detect these translocations and to test whether there is a To test for these two possibilities, we C-terminally tagged the
correlation between the acceleration of GIRK desensitization A1R with GFP (A1R-GFP) or C-terminally tagged GIRK4 with
rates and GRK translocations. For this purpose, we C-terminally GFP (GIRK4-GFP) and measured plasma membrane-associated
tagged GRK2 with EGFP (GRK2-GFP) and used total internal fluorescence under TIRF. A1R-GFP and GIRK4-GFP plasma
reflection fluorescence (TIRF) microscopy to detect exclusively membrane levels remained constant in the first minute after
the membrane-associated fluorescence (Riven et al., 2003). agonist application both in control cells and in cells cotrans-
Cells transfected with GRK2-GFP and A1R showed a significant fected with GRK2, with DF/F of 96.3 ± 1.0%; n = 6 and 97.7 ±
GRK2-GFP basal membrane associated fluorescence (Fig- 0.3%; n = 12, for A1R-GFP and 96.4 ± 0.6%; n = 5 and
ure 2A), as previously reported (Garcia-Higuera et al., 1994). 106.5 ± 1.4%; n = 9, for GIRK4-GFP, respectively (Figure 3B).
On A1R activation (Figures 2B and 2C) the membrane-associ- These results suggest that GRK2-mediated acceleration of the
ated fluorescent signal increased by 22.2 ± 6.2% with a t of GCD is neither due to a loss of receptors nor due to a loss of
1.5 ± 0.4 s (Figures 2D and 2F). mOR also increased membrane GIRK channels from the plasma membrane.
associated fluorescence on activation by 10.8 ± 2.8% with a t
of 23.4 ± 3.9 s (n = 11), temporally correlated with GCD for this Pertussis Toxin-Insensitive Pathways Are Sufficient
receptor (Figure S1D). Similar to the inability of mGluR2 to to Induce GRK2 Translocations and Acceleration
accelerate GCD, membrane associated fluorescence also did of GIRK Current Desensitization
not significantly increase after mGluR2 activation (Figure 2D). The sensitivity of A1R and mOR to GRK2-mediated desensitiza-
Similarly, M4R activation by carbachol did not induce GRK2 tion was distinct in comparison to mGluR2 and M4R, GPCRs that
translocation to the membrane (data not shown). The transloca- display pure Gi/o activation. However, whereas A1R and mOR
tions of GRK2-GFP to the membrane were reversible, as primarily activate the Gi/o pathway, they may have also a
membrane fluorescence returned to its basal level after washing secondary transduction mechanism through different G protein
out the agonist (Figure S2). These results may indicate a strong subsets (Cordeaux et al., 2004). We therefore tested whether
correlation between GRK2 translocation to the plasma other minor secondary G protein activation mechanisms might
membrane and the acceleration in GCD rates. To further explain the selectivity of only a subset of receptors to induce
strengthen this idea, we recorded A1R induced GIRK currents GRK2-mediated GCD. To inactivate the Gai/o pathway, we
and measured GRK-GFP translocation simultaneously, using coexpressed the catalytic subunit of pertussis toxin, PTX-S1,
whole cell recording of the patch clamp technique, and quantita- that been shown to effectively abolish GPCR-mediated GIRK
tive fluorescence under TIRF, respectively (Figure 2E). In cells activation (Sadja and Reuveny, 2009). In cells cotransfected
measured this way, GIRK desensitization and GRK2 recruit- with PTX-S1, A1R, and GIRK channels, A1R activation did not
ments to the membrane occurred simultaneously, with change induce GIRK currents, in agreement with Gai/o sensitivity to
of currents and membrane-associated fluorescence displaying t PTX (Figure 3C, middle). In contrast, when cells cotransfected
of 2.4 ± 0.5 s and 4.6 ± 0.9 s, n = 5, respectively. Additional with both GRK2 and PTX-S1 were activated, the basal activity
independent observations of GCD rates and membrane-associ- of the GIRK channels, assessed by barium sensitivity of the
ated fluorescence increase of GRK2-GFP were also temporally inward K+ currents at 80 mV, was rapidly reduced, in agree-
correlated with t of 1.3 ± 0.3 s, n = 20 and 1.5 ± 0.4 s, n = 11, ment with the observation that a major part of GIRK basal activity
respectively (Figure 2F). is Gbg-dependent (Rishal et al., 2005). Along the same line,

752 Cell 143, 750–760, November 24, 2010 ª2010 Elsevier Inc.
A t=0 B t=60s +Ade A B

C D
180 Ade 30
Fluorescence

140 20
(%)(%)

C D
dF/F
dF/F

100 10

60 0
0 20 Time (s) 40 6 A1R mGluR2
E F
Ade
2.0

E
(s) t 0.67 (s)

1.0

Fluorescence
Current 0.0
Current GRK2
desensitization translocation
20pA
1s

Figure 2. A1R Activation Recruits GFP-Tagged GRK2 to the


Membrane Simultaneously with GCD as Revealed under TIRF
Figure 3. Kinase Catalytic Activity Is Not Required for GRK2 Effect
(A) A TIRF image of HEK293 cell transfected with GRK2-GFP. Basal membra-
on GCD
nous fluorescence can be detected before stimulation by A1R.
A1R-GFP or GIRK1/GIRK4-GFP plasma membrane levels are not affected by
(B) Image of the same cell in the presence of adenosine.
A1R stimulation. PTX treatment is not affecting basal GCD and membrane
(C) Time course of fluorescence increase seen on receptor activation.
recruitment.
(D) A bar plot comparing the relative membrane-associated fluorescent
(A) A bar graph summarizing measurements of GCD rates (t, s) from cells
change (DF/F, in %) of GRK2-GFP after activation of A1R or mGluR2.
cotransfected with GRK2/K220R (dnGRK2), GIRK, and A1R.
(E) Typical trace of whole-cell GIRK currents (black) and TIRF signal (green)
(B) The relative change of membrane fluorescence under TIRF (DF/F, %) asso-
recorded simultaneously from the same cell.
ciated with either A1R-GFP or GIRK1/GIRK4-GFP before and during A1R
(F) Bar graph depicting the similarity between GRK2-GFP translocation and
activation (1 min after adenosine application).
GCD rates.
(C) Typical current traces of cells expressing GIRK and A1R (control); GIRK,
See also Figure S2.
A1R and PTX (+PTX); and GIRK, A1R, PTX and GRK2 (+PTX +GRK2).
(D) A bar plot summarizing DF/F of GRK2-GFP signal after A1R activation,
measured under TIRF in cells expressing GIRK, PTX, and GRK2.
GRK2 translocation to the plasma membrane remained intact, (E) A typical TIRF data of the membrane fluorescence change of GRK2-GFP
demonstrating that GRK2 membrane recruitment is not depen- overtime of a cell expressing PTX after A1R activation.
dent on the Gai/o pathway (Figures 3D and 3E). DF/F values See also Figure S3.
without or with PTX were 7.8 ± 0.6%, n = 13 and 8.0 ± 1.0%,
n = 7, respectively. As shown above, PTX-insensitive pathways
were sufficient to induce GRK2 translocations. The involvement ability of GRK2 to accelerate GCD rates. GRK2 mutations that
of other G protein signaling pathways, Gaq and Gas, were also disrupt GRK2-Gaq interaction, GRK2/R106A;D110A (Day et al.,
tested and were found not to be involved in GRK2 action on 2004; Sterne-Marr et al., 2003) were tested. These mutations
GCD (Figures S3). are located in the RGS homology domain that is known to bind
Gaq but not Gai/o (Carman et al., 1999). GRK2/R106A;D110A
The Effects of Mutations in GRK2 that Impair Its also accelerated GCD, similar to wt GRK2 (Figure S4A), with t
Interaction with Various Auxiliary Molecules of 1.3 ± 0.4 s, n = 6 and 1.3 ± 0.3 s, n = 20, respectively.
GRK2 is known to form a quaternary complex with Gaq and Gbg GRK2D97-140, a GRK2 mutant that lacks the two helices that
(Tesmer et al., 2005). We set out to test whether impairing its are involved in GRK2-Gaq interaction, was also able enhance
ability to interact with these auxiliary proteins may affect the GCD with t of 3.2 ± 0.8 s; n = 8. These results indicate that

Cell 143, 750–760, November 24, 2010 ª2010 Elsevier Inc. 753
A K567E;R578E mutant that disrupts GRK2-PtdIns(4,5)P2 interac-
tions. Disrupting GRK2 interactions with Gbg abolished the
GRK2-mediated enhancement of GCD with t of 15.6 ± 1.9 s,
n = 32 and 12.6 ± 1.8 s, n = 15 for the GRK2/R587Q and
GRK2/K663E;K665E;K667E, respectively (Figure 4B). These
rates are comparable with cells that do not coexpress GRK2,
(t of 19.3 ± 2.1 s, n = 37). Furthermore, mutations that interrupt
GRK2 interactions with PtdIns(4,5)P2, GRK2/K567E;R578E
partially reduced the enhancement of GCD with t of 5.8 ±
B 0.6 s, n = 13. When the ability of membrane translocation after
receptor activation was tested for both PtdIns(4,5)P2 and Gbg
interaction mutants, using GRK2/K567E;R578E-GFP, GRK2/
K663E;K665E;K667E-GFP or GRK2/R587Q-GFP, respectively,
translocations to the membrane could be seen, but were
reduced in comparison to the wt GRK2 (Figure S4B). On the
contrary, a triple mutant GRK2/K567E;R578E;R587Q-GFP, in
which mutations that disrupt both Gbg and PtdIns(4,5)P2 binding
were introduced, no translocations were observed (Figure S4B).
These results are in agreement with the observations of coordi-
nated interactions of GRK2 with Gbg and PtdIns(4,5)P2 in
C mediating GRK2 membrane recruitment (Pitcher et al., 1995).
To address whether the inability of GRK2/R587Q to accelerate
GCD is due to its reduced membrane translocation, we tethered
wild-type GRK2-GFP and GRK2/R587Q-GFP to the membrane
by fusing them with Src-myristoylation signal (myrGRK2-GFP
and myrGRK2/R587Q-GFP, respectively) (Figure 4C). GCD rates
were 1.3 ± 0.5 s (n = 8) and 23.9 ± 5.4 s (n = 5), for myrGRK2-GFP
and myrGRK2/R587Q-GFP, respectively (p < 0.05). Moreover,
five cells expressing myrGRK2/R587Q-GFP did not display
GCD at all. This supports the idea that failure of myrGRK2/
R587Q to accelerate GCD is due to its inability to chelate Gbg,
Figure 4. GRK2 Mutants with Impaired Gbg Binding Capability Fail to and not due to its impaired membrane targeting.
Accelerate GIRK Desensitization
(A) A cartoon that displays the structure of the complex of GRK2 with Gbg
(Tesmer et al., 2005). The locations of the different point-mutations that were GRK2 Does Not Cause Desensitization
used in (B) are marked in red. of Constituently Active GIRK Mutants
(B) A bar plot summarizing the desensitization rates (t, s) of GIRK currents, Because Gbg-GRK2 interactions seem to play an important role
measured from cells transfected with GIRK channel, A1R, and the various in mediating the enhancement of GCD, one possible scenario is
GRK2 mutants. that GRK2 is competing with the GIRK channel for Gbg on A1R-
(C) A bar plot comparing the effect of myristoylated GRK2 (myr-GRK2) and
activated release. To test this possibility, we examined the effect
myrGRK2/R587Q mutant on GIRK desensitization rate.
See also Figure S4. of GRK on constituently active, Gbg independent GIRK mutant
channels (Sadja et al., 2001), GIRK1/S170P;GIRK4/S176P (Fig-
ure 5A). To avoid saturation and to ensure high quality voltage
GRK2 interaction with Gaq is not required for GRK2 action on clamp, we recorded currents in 5.6 mM external K+ solution.
GIRK currents. Whole cell recordings of GIRK1/S170P;GIRK4/S176P show
The interactions between GRK2 and Gbg or phosphatidylino- high basal activity regardless of receptor activation (Figure S5)
sitol 4,5-bisphosphate (PtdIns(4,5)P2) have also been thoroughly (Sadja et al., 2001), with only a minor current induction on aden-
studied in vitro, with different point mutations in GRK2 osine application. In contrast to wt GIRK recordings, GRK2 failed
PH-domain (Carman et al., 2000; Sterne-Marr et al., 2003; to accelerate the GCD rates of the mutant channels (Figure 5B).
Touhara et al., 1995). Because both Gbg and PtdIns(4,5)P2 are Currents flowing through GIRK1/S170P;GIRK4/S176P channel
key players in the activation of GIRK channels (Huang et al., mutants without or with GRK2 cotransfection showed current
1998; Logothetis et al., 1987; Reuveny et al., 1994; Sui et al., levels of 95 ± 2%, n = 8 and 81 ± 4%, n = 10 (at 5 s of agonist
1998), the GRK2-mediated enhancement of GCD might involve application), respectively. This is in contrast to the significant
interference of the interactions with these two molecules. We GCD observed for the wild-type channel that had a reduction
thus compared GCD rates of control and GRK2 transfected cells, of the residual current from 94 ± 14%, n = 7 to only 21 ± 4%,
and compared them with cells coexpressing the various GRK2 n = 10 with GRK2 cotransfection at the same time point
mutants (Figure 4A): GRK2/R587Q (Carman et al., 2000) and (Figure 5B). These findings further point toward the possibility
GRK2/K663E;K665E;K667E (Touhara et al., 1995), that disrupt that GRK2-mediated GCD involves the competition between
the interactions of the kinase with Gbg, and GRK2/ the channel and GRK2 for Gbg subunit.

754 Cell 143, 750–760, November 24, 2010 ª2010 Elsevier Inc.
A Ade Ba++ C Ade Ba++ Figure 5. Constituently Active, Gbg-Inde-
CONTROL CONTROL pendent, but Not GIRK Mutants that Have
Higher Affinity to PtdIns(4,5)P2, Are Insensi-
tive to GRK2
(A) Typical traces of GIRK1/S170P;GIRK4/S176P
channel mutants, without (upper trace) or in the
presence of GRK2 (lower trace).
Ade Ba++ Ade Ba++ (B) A bar plot summarizing the residual current
+GRK2 +GRK2 (in % of total current) after agonist application
without (dark gray) and in the presence of GRK2
50pA 200pA (light gray).
10s 10s (C) Typical current traces of GIRK1/M223L;GIRK4/
I229L channel mutants, without (upper trace) or in
120
B CONTROL GRK2 D CONTROL GRK2 the presence of GRK2 (lower trace).
100 100 (D) A bar plot summarizing the residual current (in
% of induced current) after agonist application
% Current (@ 5 s)
% Current (@ 5 s)

80 80 from cell without (dark gray) and with GRK2


60 (light gray). In both case, desensitization was
60
measure 5 s after agonist application.
40 40 See also Figure S5.

20 20
0 0
WT GIRK1/S170P; WT GIRK1/M223L;
GIRK4/S176P GIRK4/I229L YFP showed a t of 2.6 ± 0.0 ns, n = 10,
in a fused dimer of YFP and mCherry
a subpopulation (92.9 ± 0.5%) of the
donor molecules displayed a much
In light of the results described above, we were interested to shorter lifetime (0.6 ± 0.0 ns) corresponding to a FRET efficiency
test whether PtdIns(4,5)P2 depletion from the channel may also of 76.7 ± 0.2%, n = 10 (Figure S6A). We set out to measure the
account for GRK2-mediated GCD. Therefore, we took an changes in FRET between N-terminally fused Gb1 with YFP
advantage of the previously described GIRK mutants that (YFP-Gb1) (Riven et al., 2006) and C-terminally fused GRK2
display enhanced PtdIns(4,5)P2 affinity, GIRK1/M223L;GIRK4/ with mCherry (GRK2-Cherry) (Figure 6A). On A1R activation
I229L (Koike-Tani et al., 2005; Zhang et al., 1999) (Figure 5C). YFP-Gb1 fluorescence decreased in the presence of GRK2-
Increasing GIRK channel affinity to PtdIns(4,5)P2 did not inhibit Cherry, in agreement with YFP fluorescence quenching by
the action of GRK2 on GCD rates, where GIRK1/M223L; mCherry due to FRET (Figure 6B). Fitting the fluorescence life-
GIRK4/I229L without or with GRK2 showed (at 5 s during agonist time decays of the donor over time revealed that, at rest, two
application) residual currents of 75 ± 4%, n = 13 and 31 ± 7%, donor subpopulations exist (Figure 6C). One subpopulation
n = 16, respectively. Wild-type GIRK without or with GRK2 (22.6 ± 0.9%, n = 8) contains YFP-Gb1 proteins that interact
showed residual currents of 88 ± 5%, n = 7 and 14 ± 4%, n = 10, with GRK2-Cherry and hence result in shorter fluorescent life-
respectively (Figure 5D). These results demonstrate that times of 0.6 ± 0.1 ns, n = 8. The remaining fraction consists of
GRK2-mediated acceleration of GCD does not occur by PtdIns free YFP-Gb1 proteins that display the characteristic monoexpo-
(4,5)P2 depletion from the channel. nential lifetime of YFP-Gb1 monomers (t-3.04 ns; see Fig-
ure S6B). After A1R activation, the relative fraction of YFP-Gb1
A1R Activation Increases the Fraction of GRK2-Bound subunits that interact with GRK2-Cherry increases, seen as an
Gbg Population increase in the relative fraction of the shorter lifetime constants
As shown above, mutations that impair GRK2-Gbg interaction (to 29.4 ± 1.6%, n = 8, p < 0.05) and as a decrease in the fraction
abolish the ability of GRK2 to accelerate GCD. To obtain further displaying long lifetime of the YFP (Figure 6C, D). The time
evidence that indeed GRK2 binds Gbg in the context of the course of the shift in relative fraction of short and long lifetimes
plasma membrane, we recorded dynamic FRET using fluores- (4.2 ± 0.7 s) resembles GCD rates and GRK2 translocations.
cence lifetime approach (FRET-FLIM), under TIRF microscopy. Similar correlation was seen when mOR was used, the rates of
In this method donor fluorescence lifetime is recorded continu- YFP-Gb1 association with GRK2-Cherry was similar to the
ously and shortening in donor lifetime is indicative of FRET. For GCD and to the GRK2-GFP translocation rates, with t average
this purpose we used YFP and mCherry as donor and acceptor, for binding increase of 69.5 ± 15.3 s (n = 6) (Figure S6C). These
respectively. This pair has the advantage of a significant overlap findings support the above observations that GRK2 action on
between donor emission and acceptor absorption, yet leaving an GCD is mediated through the binding of Gbg to GRK2.
acceptor-free donor fluorescence bandwidth for detection,
resulting in high FRET efficiencies (Goedhart et al., 2007) (Fig- DISCUSSION
ure S6A). YFP has a nearly monoexponential lifetime decay
(Figures S6A and S6B) (Kremers et al., 2006), making it suitable Desensitization is an important cellular mechanism that allows
for use as a donor for FLIM measurements. Although cytosolic cells to adapt to long-term external stimuli. In the case of

Cell 143, 750–760, November 24, 2010 ª2010 Elsevier Inc. 755
A B Figure 6. FLIM-FRET under TIRF Reveals
Ade
that A1R Activation Increases the Fraction
1000 of GRK2-Bound Gbg
950 (A) A cartoon showing the experimental scheme
900 used in the FLIM-FRET experiments.
850 (B) Time course of YFP-Gb1 emission after the

Counts
800 activation of A1R in the presence of GRK2-Cherry,
750 in agreement with YFP quenching by mCherry on
700 increase of FRET.
650 (C) YFP-Gb1 lifetime changes after A1R activation.
600 Yellow symbol depicts the FRET-free YFP-Gb1
0 50 100 150 200 250 300 (t of 3.0 ns fraction) and red symbol depicts the
C D Time (s) faster lifetime component (0.9 ns) that corre-
Ade Tau donor Increase binding (Normalized, %) Ade
80 120 sponds to a FRET interaction between YFP-Gb1
Tau fret
and GRK2-Cherry. This FRET interaction corre-
100
Relative fraction (%)

70 sponds to a FRET efficiency of 0.7.


80
60 (D) YFP-Gb1 GRK2-Cherry binding increases
60
after A1R activation (n = 9). Black line depicts
50 40
the fitting to a monoexponential function with a
20 t = 4.2 ± 0.7 s.
40
0 See also Figure S6.
30
-20
20 -40
0 50 100 150 200 250 300 0 20 40 60 80
Time (s) Time (s)

GPCR signaling pathways, desensitization is mediated by a the G protein subunits (Shui et al., 1998). Here, using electro-
decrease in the cellular response to a continuous GPCR stimula- physiological and fluorescence resonance energy transfer
tion by agonists, resulting in a decrease in receptor number at techniques, we unequivocally demonstrate that GRK2 is the
the plasma membrane. This process, that takes minutes to component of the G protein pathway that mediates this
hours, is mediated by phosphorylation of the receptor by GRK, short-term current decrease in the presence of the receptor
leading to clathrin-mediated endocytosis, in a process termed agonist. The molecular mechanism of this action will be
downregulation (Bunemann et al., 1999; Tsao and von Zastrow, discussed below.
2000). In addition to this well characterized process, other Based on our results, we suggest the following mechanism for
mechanisms are necessary for a more rapid control of GPCR- GRK2-mediated GCD (Figure 7): at rest, trimeric G-proteins are
mediated signaling, specifically when the signal is intended to bound to the nonactivated Gi/o-coupled GPCR and the channel
control changes in electrical responsiveness of cells. In this (Riven et al., 2006). After receptor activation by an agonist, the
study we have described a mechanism that is responsible for Gbg subunits dissociate from the Ga subunit to interact with
the termination of GPCR-mediated activation of GIRK channels, the Gbg-binding domains on the channel, and promote channel
which occurs within seconds. gating (opening). At the same time, GRK2 is recruited, either
In locus ceruleus neurons, Blanchet and Luscher (2002) within the two-dimensional space of the membrane (within
showed that prolonged activation of the mOR leads to inhibition 100 nm of the membrane space), or through the classical
of GIRK function. It was shown that whereas mOR-mediated cytosolic-to-plasma membrane translocation (Pitcher et al.,
presynaptic inhibition remained constant over time, postsyn- 1998). The former possibility may be aided by PtdIns(4,5)P2 or
aptic inhibition, mediated by GIRK activation, showed strong by other membrane associated proteins, including the GIRK1
desensitization of the response, indicating control over the channel subunit (Dhami et al., 2004; Li et al., 2003; Palczewski,
GIRK currents downstream of the receptors. This decrease in 1997; Rishal et al., 2005). This recruitment of GRK2, which is in
GIRK currents could be overcome by additional activation of our case a receptor-specific event, promotes the binding of
G protein pathways. As a possible model for their results, it the Gbg subunit to GRK2 or GRK3, but not GRK6 that lacks
was suggested that the receptor might activate Gbg scaven- Gbg binding capability, and thus reduces the availability of the
gers such as GRK2 and GRK3, to induce competitive inhibition Gbg subunits to the channel. To have this chelation capacity,
on GIRK activation. In a separate study using the same GRK2 has to have a higher or comparable affinity for Gbg than
neurons, it was shown that GCD was dependent on two molec- does the channel. Indeed, from binding studies it has been
ular pathways, the b-arrestin/GRK2 and the ERK1/2 pathways shown that Gbg subunits bind recombinant GIRK1 or GIRK4
(Dang et al., 2009). These findings suggested that GCD might subunits with dissociation constants of 125 nM and 50 nM,
involve modifications of the G protein pathway that serves to respectively (Krapivinsky et al., 1995), whereas Gbg affinity for
translate receptor activation to GIRK gating. In contrast, GCD GRK2 is 20 nM (Pitcher et al., 1992; Wu et al., 1998). Further
by muscarinic receptor stimulation has been attributed to evidence to support the idea that differential affinity to Gbg
a mechanism solely involved the GPCR phosphorylation- may mediate this action comes from experiments where
dependent and independent mechanisms by GRK2, and not GIRK4 was overexpressed in atrial myocytes (Bender et al.,

756 Cell 143, 750–760, November 24, 2010 ª2010 Elsevier Inc.
Figure 7. A Cartoon Describing the Mechanism by
Which GRK2 Is Negatively Regulating GIRK Channel
Function
On receptor stimulation by GPCR, the G protein trimer
undergoes activation characterized by the exchange of GDP
for GTP on the Ga subunit. This in turn leads to the dissociation
of the Gbg subunits to freely bind and activate the GIRK
channel. Concomitantly, the GPCR induces the recruitment
of GRK2 to the plasma membrane making it available to bind
Gbg subunits of the G protein. Due to the relative higher affinity
of GRK2 for Gbg and to the larger mass action, GRK2 is now
able to effectively compete for the available pool of Gbg with
the GIRK channel, leading to a gradual removal of the Gbg
subunits and to a channel closure (desensitization), still in the
presence of the receptor agonist. Channel activation precedes
the action of GRK2 mainly due to the preexisting trimeric G
proteins in the vicinity of the channels (Riven et al., 2006).

2001). In these experiments, GCD rates were greatly reduced, in addressed? It is interesting to note that receptors that were not
comparison to the GCD of GIRK1/4 heterotetramer, supporting able to support GRK2-mediated GCD, were also not able to
the idea that high affinity binding of Gbg may determine the recruit GRK2 to the plasma membrane, even though they all
extent of channel current desensitization. Removal of Gbg from release Gbg on activation to gate GIRK channels. This may
the channel by GRK to affect channel function may not require suggest that different receptors have differential mechanisms
the removal of all four Gbg subunits, due to the steep depen- to recruit GRK2 to the plasma membrane. The process of
dence of channel function on Gbg binding (Sadja et al., 2002). membrane recruitment of GRK proteins has been ascribed to
Removing only one Gbg dimer reduces the efficacy of gating a Gbg subunit-dependent mechanism (Pitcher et al., 1998;
by 70%. Finally, by using other means to chelate Gbg on the Pitcher et al., 1992). It is therefore not clear how only a subset
membrane, such as coexpression of phosducin, similar effects of receptors have the ability to recruit the kinase, where others,
on GCD can be achieved (Riven et al., 2006). In conclusion, the that also release Gbg to activate the GIRK channels, do not.
evidence provided above strongly points toward the possibility We have tried to address this issue and found that neither PLC
that the acceleration of GCD by GRK2 is due to competition inhibition by NCDC, treatment with pertussis toxin, or using
for Gbg dimers with the channel. dominant negative Gas mutant (Berlot, 2002) affected the ability
How may GRK2-mediated GCD be interpreted in light of of the receptor to recruit GRK2 to the membrane (see Figure S3).
previous suggested mechanisms? Few other mechanisms This may suggest of other still unknown mechanisms that
have been proposed in the past to explain GCD. It has been mediate this process by selective type of GPCRs, probably by
proposed that GIRK desensitization in cardiac cells might result a specific direct interaction of the intracellular loops of the
from simultaneous activation of M2R and M3R of the Gi/o and receptor with GRK2.
the Gq pathways by acetylcholine, respectively (Keselman How might the immediate desensitization be achieved? In
et al., 2007; Kobrinsky et al., 2000; Meyer et al., 2001). Whereas addition to cytosolic GRK that is recruited to the membrane on
the former leads to GIRK opening, the latter leads to GCD by receptor activation, a basal membranous subpopulation of
PLC-mediated PtdIns(4,5)P2 depletion. Evidently, GCD occurs GRK2 is observed by us and by others (Aragay et al., 1998;
also in simpler cases, where cross-talk between different GPCRs Garcia-Higuera et al., 1994; Murga et al., 1998). This subpopula-
pathways are probably not involved, and can be independent of tion can enable the immediate negative feedback of GIRK
PtdIns(4,5)P2 depletion as showed by the use of PLC inhibitors activation. We cannot rule out also the possibility that GRK is
or activators (Meyer et al., 2001; Sickmann and Alzheimer, precoupled to GIRK (Rishal et al., 2005) and undergoes an
2003). This was also true for our observations using NCDC, orientation/conformation change on activation, enabling its
a PLC inhibitor that does not block GIRK channel function (Sick- immediate competition with the channels for Gbg subunits.
mann et al., 2008). Furthermore, as shown above, mutations that There are many studies suggesting the existence of signaling
affect the affinity of the channel to PtdIns(4,5)P2 (Koike-Tani complex between GIRK and Gbg (Clancy et al., 2005; Doupnik,
et al., 2005; Zhang et al., 1999), are not affecting GRK2-mediated 2008; Nikolov and Ivanova-Nikolova, 2004; Riven et al., 2006).
channel desensitization. We thus suggest that changes in PtdIns The GIRK-Gbg precoupling, before GPCR activation, might
(4,5)P2 may only be an additional form of a much slower regula- enable the specificity of GPCR signaling cascade in an environ-
tion of channel function, mediated by the enzymatic activity of ment that may be populated by receptors of different types. Gbg
PLC (Kobrinsky et al., 2000). precoupled to GIRK undergo local rearrangement on GPCR
Our observations show that among four different receptors activation to immediately transduce GIRK gating independent
described in this study, GCD was tightly regulated by GRK2 in of diffusion rates (Riven et al., 2006). So if indeed the effector
currents induced by A1R and mOR, showing a very robust (GIRK) is a module precoupled to its ‘‘switch-on,’’ could it be
acceleration of GCD. On the contrary two other receptors, that it is also precoupled to its ‘‘switch-off’’? There is evidence
namely mGluR2 and M4R were not able to induce GCD in the that GRK2 and GIRK channel encompass a common signaling
presence of GRK2. How might this receptor selectivity be complex (Nikolov and Ivanova-Nikolova, 2004).

Cell 143, 750–760, November 24, 2010 ª2010 Elsevier Inc. 757
Our results add a unique aspect to emerging evidence for constrained to 3.0 ns (YFP-Gb1), and tda as well as the relative size for
phosphorylation-independent activity of the GRK family, from each exponential term was extracted from fitting result (Lleres et al., 2007;
Peter et al., 2005; Wallrabe and Periasamy, 2005; Yasuda et al., 2006).
the regulation of receptor numbers or uncoupling of the GPCR
Maximum likelihood estimation (MLE) method was used for fitting. Fit quality
from the G protein at the plasma membrane, to regulation of was examined both by c2 values and by the absence of systematic variations
intracellular enzymes (for reviews see Ferguson [2007] and Reiter of fit residuals.
and Lefkowitz [2006]). In all of these cases, there is no indication
of a direct involvement of the Gbg subunits of the G protein in Molecular Biology and Cell Culture
GRK action. GPCR/GRK2-dependent action on channel activity, Fusions to fluorescent proteins (EGFP, YFP and mCherry) were based on
or other effectors, forms a new mechanism for a short-term commercially available pCMV-XFP vectors (Clontech). In EGFP A206K point
negative feedback for GPCR function, that selectively regulate mutation was made to eliminate its week dimerization tendency (Zacharias
et al., 2002). Point mutations and deletion done in GIRK and GRK2 were
effector activity in the continued presence of receptor agonists.
carried out by polymerase chain reaction (PCR) and verified by sequencing.
This mechanism may not exclusively pertain to GIRK channels,
Nonfused GIRK and PTX-S1 subunits (Sadja and Reuveny, 2009) were all in
but can be relevant to all membrane associated Gbg regulated pcDNA3.1 (Invitrogen). C-terminal fusion of fluorescent proteins to GRK2 did
effectors (Dupre et al., 2009). Because drug therapies for many not affect its function. HEK293 cells were transiently transfected using
diseases are targeted to the receptor, a better understanding Metafectene (Biontex, Germany) with cDNAs encoding for the channel
of the pathway that links receptor to effector activation and subunits, the receptor of choice and GRK (wt, GFP-fused or mutant). In
regulation (in this case the GIRK channel), and finding new GRK2 silencing experiments GRK2 shRNA (0.1 mg) or nontarget control
(0.1 mg) was cotransfected with the channel and the receptor. Currents were
means to regulate these steps, might lead to therapies with
measured 24–48 hr posttransfection according to Raveh et al. (2008). The
better resistance to complications such as tolerance and HL-1 cells, a gift from Dr. William C. Claycomb, were maintained using the
side-effects. recommended protocols (Claycomb et al., 1998). For electrophysiology
experiments, cells were transferred to uncoated 24-mm glass coverslips on
EXPERIMENTAL PROCEDURES the day of the recording.

Patch-Clamp Recordings SUPPLEMENTAL INFORMATION


Membrane currents were recorded under voltage-clamp conditions using
whole-cell patch-clamp configuration with an Axopatch 200B (Axon Instru- Supplemental Information includes Extended Experimental Procedures
ments) patch-clamp amplifier. Patch pipettes were fabricated from borosili- and six figures and can be found with this article online at doi:10.1016/
cate glass capillaries (2–5 MU). Signals were analog filtered using a 1 kHz j.cell.2010.10.018.
low-pass Bessel filter. After patch formation in a low K+ bath solution, the
bath solution was changed to high K+ solution. Adenosine (100 mM), glutamate
ACKNOWLEDGMENTS
(100 mM), methionine enkephalin (ME, 100 nM), carbachol (100 mM), and Ba+2
(3 mM) were used to study induced and basal GIRK currents. GIRK currents
The authors like to thank Ruth Meller and Elisha Shalgi for technical help,
were measured as inward currents at a holding potential of 80 mV at room
and the Reuveny laboratory for helpful comments. We are grateful to
temperature. Data acquisition and analysis were done using pCLAMP 9
Drs. J.L. Benovic for GRK2 and GRK6, W.C. Claycomb for HL-1 cells,
software (Axon Instruments). To determine GCD kinetics, current traces
D.E. Logothetis for PtdIns(4,5)P2 GIRK mutants, C. Barlot for Gas mutant,
were fitted to a monoexponential decay function using Chebyshev method.
S. Nakanishi for the mGluR2, Z. Vogel for mOR, and R. Tsien for mCherry
Results are expressed as average ± standard error of the mean (SEM). Signif-
cDNAs. The work was supported in part by the Josef Cohn Center for
icant differences were considered when p < 0.05 using Student’s t test.
Biomembrane Research, The Israeli Science Foundation (ISF grant 207/09),
The Minerva Foundation, and the Human Frontier Science Program.
TIRF Microscopy
Fluorescence was measured using through the objective TIRF microscopy Received: April 12, 2010
(Riven et al., 2003) with a 60 3 1.45 N.A. TIRFM objective (Olympus, Japan) Revised: August 3, 2010
and TIRF condenser (TILL Photonics, Germany). Images were acquired with Accepted: October 11, 2010
Ixon+ EMCCD camera (Andor, Ireland) using Imaging Workbench 6 software Published: November 24, 2010
(Indec, USA). DF/F (%) was calculated from ROI that contained the whole
cell membrane area and was background subtracted. Time constant (t) for
REFERENCES
GRK2 translocations, was calculated by determining the time after agonist
application when fluorescence reached 63% of maximum.
Aragay, A.M., Ruiz-Gomez, A., Penela, P., Sarnago, S., Elorza, A., Jimenez-
Sainz, M.C., and Mayor, F., Jr. (1998). G protein-coupled receptor kinase 2
Fluorescence Lifetime Measurements (GRK2): mechanisms of regulation and physiological functions. FEBS Lett.
For fluorescence lifetime measurements (FLIM), 470 nm ps diode laser 430, 37–40.
(FWHM < 90 ps) was used, driven by a 40 MHz pulse controller, PDL 800-B.
Bender, K., Wellner-Kienitz, M.C., Inanobe, A., Meyer, T., Kurachi, Y., and Pott,
Single photons were collected using PMA-165P photon counter and
L. (2001). Overexpression of monomeric and multimeric GIRK4 subunits in rat
processed using TimeHarp 200 PC-board. Data was acquired and analyzed
atrial myocytes removes fast desensitization and reduces inward rectification
using SymPhoTime software (PicoQuant, Germany). Donor fluorescence
of muscarinic K(+) current (I(K(ACh))). Evidence for functional homomeric
was collected from single cells under TIRF configuration (Riven et al., 2003).
GIRK4 channels. J. Biol. Chem. 276, 28873–28880.
For all measurements, laser intensities were set such that signal count rate
will be <1% of laser pulse rate. IRF was reconstructed from lifetime measure- Berlot, C.H. (2002). A highly effective dominant negative alpha s construct
ment of YFP-Gb1 under TIRF using laser powers comparable to those used in containing mutations that affect distinct functions inhibits multiple Gs-coupled
the experiment. YFP-Gb1 monomer lifetime was monoexponential with t of receptor signaling pathways. J. Biol. Chem. 277, 21080–21085.
3.0 ns (Figure S6B). To extract lifetimes and relative intensities, donor fluores- Blanchet, C., and Luscher, C. (2002). Desensitization of mu-opioid receptor-
cence traces were binned to 1-s segments and IRF reconvoluted trace evoked potassium currents: initiation at the receptor, expression at the
was fitted to double-exponential fitting model. One t parameter, td, was effector. Proc. Natl. Acad. Sci. USA 99, 4674–4679.

758 Cell 143, 750–760, November 24, 2010 ª2010 Elsevier Inc.
Bunemann, M., Lee, K.B., Pals-Rylaarsdam, R., Roseberry, A.G., and Hosey, Kong, G., Penn, R., and Benovic, J.L. (1994). A beta-adrenergic receptor
M.M. (1999). Desensitization of G-protein-coupled receptors in the cardiovas- kinase dominant negative mutant attenuates desensitization of the beta
cular system. Annu. Rev. Physiol. 61, 169–192. 2-adrenergic receptor. J. Biol. Chem. 269, 13084–13087.
Carman, C.V., Parent, J.L., Day, P.W., Pronin, A.N., Sternweis, P.M., Krapivinsky, G., Krapivinsky, L., Wickman, K., and Clapham, D.E. (1995).
Wedegaertner, P.B., Gilman, A.G., Benovic, J.L., and Kozasa, T. (1999). G beta gamma binds directly to the G protein-gated K+ channel, IKACh.
Selective regulation of Galpha(q/11) by an RGS domain in the G protein- J. Biol. Chem. 270, 29059–29062.
coupled receptor kinase, GRK2. J. Biol. Chem. 274, 34483–34492. Kremers, G.J., Goedhart, J., van Munster, E.B., and Gadella, T.W., Jr. (2006).
Carman, C.V., Barak, L.S., Chen, C., Liu-Chen, L.Y., Onorato, J.J., Kennedy, Cyan and yellow super fluorescent proteins with improved brightness, protein
S.P., Caron, M.G., and Benovic, J.L. (2000). Mutational analysis of folding, and FRET Forster radius. Biochemistry 45, 6570–6580.
Gbetagamma and phospholipid interaction with G protein-coupled receptor Li, J., Xiang, B., Su, W., Zhang, X., Huang, Y., and Ma, L. (2003). Agonist-
kinase 2. J. Biol. Chem. 275, 10443–10452. induced formation of opioid receptor-G protein-coupled receptor kinase
(GRK)-G beta gamma complex on membrane is required for GRK2 function
Clancy, S.M., Fowler, C.E., Finley, M., Suen, K.F., Arrabit, C., Berton, F.,
in vivo. J. Biol. Chem. 278, 30219–30226.
Kosaza, T., Casey, P.J., and Slesinger, P.A. (2005). Pertussis-toxin-sensitive
Galpha subunits selectively bind to C-terminal domain of neuronal GIRK Lleres, D., Swift, S., and Lamond, A.I. (2007). Detecting protein-protein
channels: evidence for a heterotrimeric G-protein-channel complex. Mol. interactions in vivo with FRET using multiphoton fluorescence lifetime imaging
Cell. Neurosci. 28, 375–389. microscopy (FLIM). Curr. Protoc. Cytom. Chapter 12, Unit12.10.

Claycomb, W.C., Lanson, N.A., Jr., Stallworth, B.S., Egeland, D.B., Delcarpio, Logothetis, D.E., Kurachi, Y., Galper, J., Neer, E.J., and Clapham, D.E. (1987).
J.B., Bahinski, A., and Izzo, N.J., Jr. (1998). HL-1 cells: a cardiac muscle cell The beta gamma subunits of GTP-binding proteins activate the muscarinic K+
line that contracts and retains phenotypic characteristics of the adult cardio- channel in heart. Nature 325, 321–326.
myocyte. Proc. Natl. Acad. Sci. USA 95, 2979–2984. Massoulie, J., Pezzementi, L., Bon, S., Krejci, E., and Vallette, F.M. (1993).
Molecular and cellular biology of cholinesterases. Prog. Neurobiol. 41, 31–91.
Cordeaux, Y., Ijzerman, A.P., and Hill, S.J. (2004). Coupling of the human A1
adenosine receptor to different heterotrimeric G proteins: evidence for Meyer, T., Wellner-Kienitz, M.C., Biewald, A., Bender, K., Eickel, A., and Pott,
agonist-specific G protein activation. Br. J. Pharmacol. 143, 705–714. L. (2001). Depletion of phosphatidylinositol 4,5-bisphosphate by activation of
phospholipase C-coupled receptors causes slow inhibition but not desensiti-
Dang, V.C., Napier, I.A., and Christie, M.J. (2009). Two distinct mechanisms zation of G protein-gated inward rectifier K+ current in atrial myocytes. J. Biol.
mediate acute mu-opioid receptor desensitization in native neurons. Chem. 276, 5650–5658.
J. Neurosci. 29, 3322–3327.
Murga, C., Penela, P., Zafra, F., and Mayor, F., Jr. (1998). The subcellular and
Day, P.W., Tesmer, J.J., Sterne-Marr, R., Freeman, L.C., Benovic, J.L., and cellular distribution of G protein-coupled receptor kinase 2 in rat brain.
Wedegaertner, P.B. (2004). Characterization of the GRK2 binding site of Neuroscience 87, 631–637.
Galphaq. J. Biol. Chem. 279, 53643–53652.
Nikolov, E.N., and Ivanova-Nikolova, T.T. (2004). Coordination of membrane
Dhami, G.K., Dale, L.B., Anborgh, P.H., O’Connor-Halligan, K.E., Sterne-Marr, excitability through a GIRK1 signaling complex in the atria. J. Biol. Chem.
R., and Ferguson, S.S. (2004). G Protein-coupled receptor kinase 2 regulator 279, 23630–23636.
of G protein signaling homology domain binds to both metabotropic glutamate Nobles, M., Sebastian, S., and Tinker, A. (2010). HL-1 cells express an inwardly
receptor 1a and Galphaq to attenuate signaling. J. Biol. Chem. 279, 16614– rectifying K+ current activated via muscarinic receptors comparable to that in
16620. mouse atrial myocytes. Pflugers Arch. 460, 99–108.
Doupnik, C.A. (2008). GPCR-Kir channel signaling complexes: defining rules of Palczewski, K. (1997). GTP-binding-protein-coupled receptor kinases–two
engagement. J. Recept. Signal Transduct. Res. 28, 83–91. mechanistic models. Eur. J. Biochem. 248, 261–269.
Dupre, D.J., Robitaille, M., Rebois, R.V., and Hebert, T.E. (2009). The role of Peter, M., Ameer-Beg, S.M., Hughes, M.K., Keppler, M.D., Prag, S., Marsh, M.,
Gbetagamma subunits in the organization, assembly, and function of GPCR Vojnovic, B., and Ng, T. (2005). Multiphoton-FLIM quantification of the
signaling complexes. Annu. Rev. Pharmacol. Toxicol. 49, 31–56. EGFP-mRFP1 FRET pair for localization of membrane receptor-kinase
Ferguson, S.S. (2007). Phosphorylation-independent attenuation of GPCR interactions. Biophys. J. 88, 1224–1237.
signalling. Trends Pharmacol. Sci. 28, 173–179. Pierce, K.L., Premont, R.T., and Lefkowitz, R.J. (2002). Seven-transmembrane
receptors. Nat. Rev. Mol. Cell Biol. 3, 639–650.
Garcia-Higuera, I., Penela, P., Murga, C., Egea, G., Bonay, P., Benovic, J.L.,
and Mayor, F., Jr. (1994). Association of the regulatory beta-adrenergic Pitcher, J.A., Inglese, J., Higgins, J.B., Arriza, J.L., Casey, P.J., Kim, C.,
receptor kinase with rat liver microsomal membranes. J. Biol. Chem. 269, Benovic, J.L., Kwatra, M.M., Caron, M.G., and Lefkowitz, R.J. (1992). Role
1348–1355. of beta gamma subunits of G proteins in targeting the beta-adrenergic
receptor kinase to membrane-bound receptors. Science 257, 1264–1267.
Goedhart, J., Vermeer, J.E., Adjobo-Hermans, M.J., van Weeren, L., and
Pitcher, J.A., Touhara, K., Payne, E.S., and Lefkowitz, R.J. (1995). Pleckstrin
Gadella, T.W., Jr. (2007). Sensitive detection of p65 homodimers using
homology domain-mediated membrane association and activation of the
red-shifted and fluorescent protein-based FRET couples. PLoS ONE 2, e1011.
beta-adrenergic receptor kinase requires coordinate interaction with G beta
Huang, C.L., Feng, S., and Hilgemann, D.W. (1998). Direct activation of inward gamma subunits and lipid. J. Biol. Chem. 270, 11707–11710.
rectifier potassium channels by PIP2 and its stabilization by Gbetagamma.
Pitcher, J.A., Freedman, N.J., and Lefkowitz, R.J. (1998). G protein-coupled
Nature 391, 803–806.
receptor kinases. Annu. Rev. Biochem. 67, 653–692.
Keselman, I., Fribourg, M., Felsenfeld, D.P., and Logothetis, D.E. (2007). Raveh, A., Riven, I., and Reuveny, E. (2008). The use of FRET microscopy to
Mechanism of PLC-mediated Kir3 current inhibition. Channels (Austin) 1, elucidate steady state channel conformational rearrangements and G protein
113–123. interaction with the GIRK channels. Methods Mol. Biol. 491, 199–212.
Kobrinsky, E., Mirshahi, T., Zhang, H., Jin, T., and Logothetis, D.E. (2000). Reiter, E., and Lefkowitz, R.J. (2006). GRKs and beta-arrestins: roles in
Receptor-mediated hydrolysis of plasma membrane messenger PIP2 leads receptor silencing, trafficking and signaling. Trends Endocrinol. Metab. 17,
to K+-current desensitization. Nat. Cell Biol. 2, 507–514. 159–165.
Koike-Tani, M., Collins, J.M., Kawano, T., Zhao, P., Zhao, Q., Kozasa, T., Reuveny, E., Slesinger, P.A., Inglese, J., Morales, J.M., Iniguez-Lluhi, J.A.,
Nakajima, S., and Nakajima, Y. (2005). Signal transduction pathway for the Lefkowitz, R.J., Bourne, H.R., Jan, Y.N., and Jan, L.Y. (1994). Activation of
substance P-induced inhibition of rat Kir3 (GIRK) channel. J. Physiol. 564, the cloned muscarinic potassium channel by G protein beta gamma subunits.
489–500. Nature 370, 143–146.

Cell 143, 750–760, November 24, 2010 ª2010 Elsevier Inc. 759
Rishal, I., Porozov, Y., Yakubovich, D., Varon, D., and Dascal, N. (2005). Na+ ions depends on the presence of phosphatidylinositol phosphates. Proc.
Gbetagamma-dependent and Gbetagamma-independent basal activity of G Natl. Acad. Sci. USA 95, 1307–1312.
protein-activated K+ channels. J. Biol. Chem. 280, 16685–16694.
Tesmer, V.M., Kawano, T., Shankaranarayanan, A., Kozasa, T., and Tesmer,
Riven, I., Kalmanzon, E., Segev, L., and Reuveny, E. (2003). Conformational J.J. (2005). Snapshot of activated G proteins at the membrane: the
rearrangements associated with the gating of the G protein-coupled Galphaq-GRK2-Gbetagamma complex. Science 310, 1686–1690.
potassium channel revealed by FRET microscopy. Neuron 38, 225–235.
Torres, G.E., Gainetdinov, R.R., and Caron, M.G. (2003). Plasma membrane
Riven, I., Iwanir, S., and Reuveny, E. (2006). GIRK channel activation involves
monoamine transporters: structure, regulation and function. Nat. Rev.
a local rearrangement of a preformed G protein channel complex. Neuron 51,
Neurosci. 4, 13–25.
561–573.
Touhara, K., Koch, W.J., Hawes, B.E., and Lefkowitz, R.J. (1995). Mutational
Sadja, R., and Reuveny, E. (2009). Activation gating kinetics of GIRK channels
analysis of the pleckstrin homology domain of the beta-adrenergic receptor
are mediated by cytoplasmic residues adjacent to transmembrane domains.
kinase. Differential effects on G beta gamma and phosphatidylinositol
Channels (Austin) 3, 205–214.
4,5-bisphosphate binding. J. Biol. Chem. 270, 17000–17005.
Sadja, R., Smadja, K., Alagem, N., and Reuveny, E. (2001). Coupling
Gbetagamma-dependent activation to channel opening via pore elements in Tsao, P., and von Zastrow, M. (2000). Downregulation of G protein-coupled
inwardly rectifying potassium channels. Neuron 29, 669–680. receptors. Curr. Opin. Neurobiol. 10, 365–369.
Sadja, R., Alagem, N., and Reuveny, E. (2002). Graded contribution of the Violin, J.D., Ren, X.R., and Lefkowitz, R.J. (2006). G-protein-coupled receptor
Gbeta gamma binding domains to GIRK channel activation. Proc. Natl. kinase specificity for beta-arrestin recruitment to the beta2-adrenergic
Acad. Sci. USA 99, 10783–10788. receptor revealed by fluorescence resonance energy transfer. J. Biol. Chem.
Shui, Z., Khan, I.A., Tsuga, H., Haga, T., and Boyett, M.R. (1998). Role of 281, 20577–20588.
receptor kinase in short-term desensitization of cardiac muscarinic K+ chan- Wallrabe, H., and Periasamy, A. (2005). Imaging protein molecules using FRET
nels expressed in Chinese hamster ovary cells. J. Physiol. 507, 325–334. and FLIM microscopy. Curr. Opin. Biotechnol. 16, 19–27.
Sickmann, T., and Alzheimer, C. (2003). Short-term desensitization of
Wu, G., Benovic, J.L., Hildebrandt, J.D., and Lanier, S.M. (1998). Receptor
G-protein-activated, inwardly rectifying K+ (GIRK) currents in pyramidal
docking sites for G-protein betagamma subunits. Implications for signal
neurons of rat neocortex. J. Neurophysiol. 90, 2494–2503.
regulation. J. Biol. Chem. 273, 7197–7200.
Sickmann, T., Klose, A., Huth, T., and Alzheimer, C. (2008). Unexpected
Yasuda, R., Harvey, C.D., Zhong, H., Sobczyk, A., van Aelst, L., and Svoboda,
suppression of neuronal G protein-activated, inwardly rectifying K+ current
K. (2006). Supersensitive Ras activation in dendrites and spines revealed by
by common phospholipase C inhibitor. Neurosci. Lett. 436, 102–106.
two-photon fluorescence lifetime imaging. Nat. Neurosci. 9, 283–291.
Sterne-Marr, R., Tesmer, J.J., Day, P.W., Stracquatanio, R.P., Cilente, J.A.,
O’Connor, K.E., Pronin, A.N., Benovic, J.L., and Wedegaertner, P.B. (2003). Zacharias, D.A., Violin, J.D., Newton, A.C., and Tsien, R.Y. (2002). Partitioning
G protein-coupled receptor Kinase 2/G alpha q/11 interaction. A novel surface of lipid-modified monomeric GFPs into membrane microdomains of live cells.
on a regulator of G protein signaling homology domain for binding G alpha Science 296, 913–916.
subunits. J. Biol. Chem. 278, 6050–6058. Zhang, H., He, C., Yan, X., Mirshahi, T., and Logothetis, D.E. (1999). Activation
Sui, J.L., Petit-Jacques, J., and Logothetis, D.E. (1998). Activation of the of inwardly rectifying K+ channels by distinct PtdIns(4,5)P2 interactions.
atrial KACh channel by the betagamma subunits of G proteins or intracellular Nat. Cell Biol. 1, 183–188.

760 Cell 143, 750–760, November 24, 2010 ª2010 Elsevier Inc.
Sequence-Dependent Sorting
of Recycling Proteins by Actin-Stabilized
Endosomal Microdomains
Manojkumar A. Puthenveedu,1,* Benjamin Lauffer,2 Paul Temkin,2 Rachel Vistein,1 Peter Carlton,3 Kurt Thorn,4
Jack Taunton,5 Orion D. Weiner,4 Robert G. Parton,6 and Mark von Zastrow2,5
1Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA
2Department of Psychiatry
3Department of Physiology
4Department of Biochemistry and Biophysics
5Department of Cellular and Molecular Pharmacology

University of California at San Francisco, San Francisco, CA 94158, USA


6The University of Queensland, Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, St. Lucia,

Queensland 4072, Australia 8


*Correspondence: map3@andrew.cmu.edu
DOI 10.1016/j.cell.2010.10.003

SUMMARY (TfR), are sorted away from soluble proteins, largely by bulk
membrane flow back to the cell surface. This occurs via the
The functional consequences of signaling receptor formation and fission of narrow tubules that have a high ratio
endocytosis are determined by the endosomal sort- of membrane surface area (and therefore membrane proteins)
ing of receptors between degradation and recycling to volume (soluble contents) (Mayor et al., 1993). Several
pathways. How receptors recycle efficiently, in proteins have been implicated in the formation of these tubules
a sequence-dependent manner that is distinct from (Shinozaki-Narikawa et al., 2006; Cullen, 2008; Traer et al.,
2007), which provide a geometric basis to bulk recycling and
bulk membrane recycling, is not known. Here, in
explain how nutrient receptors can recycle leaving soluble
live cells, we visualize the sorting of a prototypical
nutrients behind to be utilized in the lysosome (Dunn and
sequence-dependent recycling receptor, the beta-2 Maxfield, 1992; Mayor et al., 1993; Maxfield and McGraw,
adrenergic receptor, from bulk recycling proteins 2004). Second, many membrane proteins are transported to
and the degrading delta-opioid receptor. Our results the lysosome to be degraded. This involves a process called
reveal a remarkable diversity in recycling routes at involution, where proteins are packaged into vesicles that bud
the level of individual endosomes, and indicate that off to the interior of the endosome and, in essence, converts
sequence-dependent recycling is an active process these proteins into being a part of the soluble contents (Piper
mediated by distinct endosomal subdomains and Katzmann, 2007). Involution has also been studied exten-
distinct from those mediating bulk recycling. We sively, and the machinery responsible, termed ESCRT complex,
identify a specialized subset of tubular microdo- identified (Hurley, 2008; Saksena et al., 2007; Williams and Urbé,
2007). Third, several other membrane proteins, such as many
mains on endosomes, stabilized by a highly localized
signaling receptors, escape the bulk recycling and degradation
but dynamic actin machinery, that mediate this sort-
pathways, and are instead recycled in a regulated manner (Ha-
ing, and provide evidence that these actin-stabilized nyaloglu and von Zastrow, 2008; Yudowski et al., 2009). This
domains provide the physical basis for a two-step requires a specific cis-acting sorting sequence present on the
kinetic and affinity-based model for protein sorting receptor’s cytoplasmic surface (Cao et al., 1999; Hanyaloglu
into the sequence-dependent recycling pathway. and von Zastrow, 2008). How receptors use these sequences
to escape the involution pathway and recycle, though they are
INTRODUCTION excluded from the default recycling pathway (Maxfield and
McGraw, 2004; Hanyaloglu et al., 2005), is a fundamental cell
Cells constantly internalize a large fraction of proteins from their biological question that is still unanswered.
surface and the extracellular environment. The fates of these Although it is clear that different recycling cargo can travel
internalized proteins in the endosome have a direct impact on through discrete endosomal populations (Maxfield and McGraw,
several critical functions of the cell, including its response to 2004), endosome-to-plasma membrane recycling from a single
environmental signals (Lefkowitz et al., 1998; Marchese et al., endosome is generally thought to occur via a uniform population
2008; Sorkin and von Zastrow, 2009). of tubules. Contrary to this traditional view, we identify special-
Internalized proteins have three main fates in the endosome. ized endosomal tubular domains mediating sequence-depen-
First, many membrane proteins, such as the transferrin receptor dent recycling that are kinetically and biochemically distinct

Cell 143, 761–773, November 24, 2010 ª2010 Elsevier Inc. 761
Figure 1. B2AR Is Enriched in Endosomal Tubular Domains Devoid of DOR
(A) HEK293 cells stably expressing FLAG-B2AR, labeled with fluorescently-tagged anti-FLAG antibodies, were followed by live confocal imaging before (left) and
after 5 min (right) of isoproterenol treatment. Arrows show internal endosomes.
(B) Example endosomes showing tubular domains enriched in B2AR (arrowheads) with one enlarged in the inset.
(C) Examples of DOR endosomes. DOR is smoothly distributed on the endosomal membrane and is not detected in tubules.
(D) Average fluorescence of B2AR (red circles) and TfR (green diamonds) calculated across multiple tubules (n = 123 for B2AR, 100 for TfR). B2AR shows a 50%
enrichment over the endosomal membrane, while TfR is not enriched. Each point denotes an individual tubule, the bar denotes the mean, and the gray dotted line
denotes the fluorescence of the endosomal membrane.
(E) An endosome containing both internalized B2AR and DOR, showing a tubule containing B2AR but no detectable DOR (arrowheads).
(F) Trace of linear pixel values across the same endosome, normalized to the maximum, confirms that the tubule is enriched for B2AR but not DOR.
(G) Linear pixel values of endosomal tubules averaged across 11 endosomes show specific enrichment of B2AR in tubules.
Error bars are SEM. See also Figure S1 and Movie S1 and Movie S2.

from the domains that mediate bulk recycling. These domains surface before isoproterenol or DADLE, their respective
are stabilized by a local actin cytoskeleton that is required and agonists, were added. After agonist addition, both B2AR
sufficient for receptor recycling. We propose that such special- (Figure 1A) and DOR (data not shown) were robustly internalized,
ized actin-stabilized domains provide the physical basis for over- and appeared in endosomes within 5 min (Figure 1A and Movie
coming a kinetic barrier for receptor entry into endosomal S1 available online). As a control, receptors did not internalize
tubules and for affinity-based concentration of proteins in the in cells not treated with agonists, but imaged for the same period
sequence-dependent recycling pathway. of time (Figure S1A). The B2AR-containing endosomes colocal-
ized with the early endosome markers Rab5 (Figure S1B) and
RESULTS EEA1 (data not shown), consistent with previous data.
Internalized B2AR (Figure 1B), but not DOR (Figure 1C), also
Visualization of Receptor Sorting in the Endosomes labeled tubules that extended from the main body of the
of Living Cells receptor. When receptor fluorescence was quantified across
The beta 2-adrenergic receptor (B2AR) and the delta opioid multiple B2AR-containing tubules, we saw that receptors were
receptor (DOR) provide excellent models for physiologically rele- enriched in these tubules compared to the rest of the endosomal
vant proteins that are sorted from each other in the endosome. limiting membrane (Figure 1D). The bulk recycling protein TfR, in
Although they share endocytic pathways, B2AR is recycled effi- contrast, was not enriched in endosomal tubules (Figure 1D).
ciently in a sequence-dependent manner while DOR is selec- This suggests that sequence-dependent recycling receptors
tively degraded in the lysosome (Cao et al., 1999; Whistler are enriched by an active mechanism in these endosomal
et al., 2002). To study the endosomal sorting of these cargo tubules.
molecules, we started by testing whether tubulation was These endosomal tubules were preferentially enriched for
involved in this process. Because such sorting has not been B2AR over DOR on the same endosome. In cells coexpressing
observed in vivo, we first attempted to visualize the dynamics FLAG-tagged B2AR and GFP-tagged DOR, we observed endo-
of receptor sorting in live HEK293 cells expressing fluorescently somes that contained both receptors within 5 min after coapply-
labeled B2AR or DOR receptors, using high-resolution confocal ing isoproterenol and DADLE. Notably, these endosomes
microscopy. Both receptors were observed mostly on the cell extruded tubules that contained B2AR but not detectable DOR

762 Cell 143, 761–773, November 24, 2010 ª2010 Elsevier Inc.
Figure 2. Membranes Derived from Endosomal Tubules Deliver B2AR to the Cell Surface
(A) Frames from a representative time lapse series showing scission of a vesicle that contains B2AR but not detectable DOR, from an endosomal tubule.
(B) An image plane close to the plasma membrane in cells coexpressing SpH-B2AR and FLAG-B2AR (labeled with Alexa555), exposed to isoproterenol for 5 min,
and imaged by fast dual-color confocal microscopy. Arrows denote the FLAG-B2AR-containing membrane derived from the endosomal tubule that fuses.
(C) Fluorescence trace of the B2AR-containing membranes from the endosome in movie S4, showing the spike in SpH-B2AR fluorescence (fusion) followed by
rapid loss of fluorescence.
Scale bars represent 1mm. See also Figure S1 and Movie S3 and Movie S4.

(e.g., in Figure 1E and in Movie S2). Fluorescence traces across domain of B2AR (SpH-B2AR) (Miesenböck et al., 1998). SpH-
the endosome and the tubule confirmed that DOR was not B2AR is highly fluorescent when exposed to the neutral pH at
detectable in these B2AR tubules, suggesting that B2AR was the cell surface, but is quenched in the acidic environments of
specifically sorted into these tubular domains (e.g., in Figure 1F). endosomes and intracellular vesicles. This allows the detection
When linear pixel values from multiple sorting events were quan- of individual fusion events of vesicles containing B2AR at the
tified, B2AR was enriched 50% in the endosomal domains from cell surface (Yudowski et al., 2009). In cells coexpressing SpH-
which tubules originate, compared to the endosomal membrane B2AR and B2AR labeled with a pH-insensitive fluorescent dye
outside these domains (Figure 1G). Thus, these experiments (Alexa-555), vesicles derived from the endosomal tubules traf-
resolve, for the first time, individual events that mediate sorting ficked to the cell surface and fused, as seen by a sudden
of two signaling receptors in the endosomes of live cells. increase in SpH fluorescence followed by loss of fluorescence
due to diffusion (Figure 2B, and Movie S4). A fluorescence trace
from movie S4 confirmed the fusion and loss of B2AR fluores-
B2AR-Containing Endosomal Tubules Deliver Receptors
cence (Figure 2C). Also, Rab4 and Rab11, which function in
to the Cell Surface
endosome-to-plasma membrane recycling (Zerial and McBride,
To test whether these tubules mediated recycling of B2AR, we
2001; Maxfield and McGraw, 2004), were localized to the
visualized direct delivery of receptors from these tubules to the
domains containing B2AR (Figure S1). Together, this indicates
cell surface. In endosomes containing internalized B2AR and
that the B2AR-containing endosomal tubules mediate delivery
DOR, these tubular domains pinched off vesicles that contained
of B2AR to the cell surface.
B2AR but not detectable levels of DOR (Figure 2A and Movie S3).
To reliably assess if these vesicles traveled to the surface and
fused with the plasma membrane, we combined our current B2AR-Containing Tubules Are Marked by a Highly
imaging with a method that we have used previously to visualize Localized Actin Cytoskeleton
individual vesicle fusion events mediating surface receptor We next examined whether the B2AR-containing microdomains
delivery (Yudowski et al., 2006). Briefly, we attached the pH- were biochemically distinct from the rest of the endosomal
sensitive GFP variant superecliptic pHluorin to the extracellular membrane. We first focused on actin, as the actin cytoskeleton

Cell 143, 761–773, November 24, 2010 ª2010 Elsevier Inc. 763
Figure 3. B2AR Tubules Are Marked by a Highly Localized Actin Cytoskeleton
(A) Cells coexpressing fluorescently labeled B2AR and actin-GFP exposed to isoproterenol for 5 min. The boxed area is enlarged in the inset, with arrowheads
indicating specific concentration of actin on B2AR endosomal tubules.
(B) Time lapse series from an example endosome with B2AR and coronin-GFP. Coronin is detectable on the endosomal tubule (arrows) and on the vesicle (arrow-
heads) that buds off the endosome.
(C) A trace of linear pixel values across the same endosome, normalized to maximum fluorescence, shows coronin on the endosomal domain and the vesicle.
(D) Example structured illumination image of a B2AR endosome showing specific localization of coronin to a B2AR tubule (arrowheads).
(E) Electron micrograph of an HRP-positive endosome (arrow) showing actin filaments (labeled with 9 nm gold, arrowheads) along a tubule. The right panel shows
an enlarged view.
See also Movie S5 and Movie S6.

is required for efficient recycling of B2AR but not of TfR (Cao this specific actin concentration on the tubule (n = 350). As with
et al., 1999; Gage et al., 2005), and as it has been implicated in actin, coronin-GFP (Uetrecht and Bear, 2006), an F-actin binding
endosome motility (Stamnes, 2002; Girao et al., 2008) and protein, also localized specifically to the B2AR-containing
vesicle scission at the cell surface (Yarar et al., 2005; Perrais tubules on endosomes (Figure 3B), confirming that this was
and Merrifield, 2005; Kaksonen et al., 2005). Strikingly, in cells a polymerized actin cytoskeleton. Coronin was also observed
coexpressing B2AR and actin-GFP, actin was concentrated on on the B2AR-containing vesicle that was generated by dynamic
the endosome specifically on the tubular domains containing scission of the B2AR tubule (Figure 3B and Movie S5). Fluores-
B2AR (Figure 3A). Virtually every B2AR tubule observed showed cence traces of the linear pixels across the tubule and the vesicle

764 Cell 143, 761–773, November 24, 2010 ª2010 Elsevier Inc.
confirmed that coronin pinched off with the B2AR vesicle two main families of Arp2/3 activators (Millard et al., 2004), on the
(Figure 3C). endosome (Figure 4G). Similarly, we did not see endosomal
We also used two separate techniques to characterize actin recruitment of activated Cdc42, as assessed by a previously
localization on these tubules beyond the 250 nm resolution characterized GFP-fusion reporter consisting of the GTPase
offered by conventional microscopy. First, we first imaged the binding domain of N-WASP (Benink and Bement, 2005)
localization of coronin on endosomes containing B2AR tubules (data not shown). All three proteins were readily detected at
using structured illumination microscopy (Gustafsson et al., lamellipodia and filopodia as expected, indicating that the
2008), which resolves structures at 100 nm spatial resolution. proteins were functional in these cells. While we cannot rule
3D stacks obtained using this high-resolution technique out a weak or transitory interaction of these activators with
confirmed that coronin was specifically localized on the endoso- Arp2/3 at the endosome, the lack of enrichment prompted us
mal tubule that contained B2AR (Figure 3D and Movie S6). to test for alternate Arp2/3 activators. Cortactin, an Arp- and
Second, we examined the morphology of actin on endosomal actin- binding protein present on endosomes, has been
tubules at the ultrastructural level by pre-embedding immunoe- proposed to be such an activator (Kaksonen et al., 2000; Millard
lectron microscopy. Actin was clearly labeled as filaments lying et al., 2004; Daly, 2004). Cortactin-GFP was clearly concentrated
along tubules extruded from endosomal structures (Figure 3E). at the base of the B2AR tubule on the endosome (Figure 4G), in
a pattern identical to Arp2/3. When quantified (>200 endosomes
Actin Is Dynamically Turned over on the B2AR- each), every B2AR tubule was marked by cortactin, while none of
Containing Endosomal Tubules the endosomes showed detectable N-WASP, WAVE-2, or
We then tested whether the actin filaments on these tubules Cdc42. Similarly, the WASH protein complex, which has been
were a stable structure or were dynamically turned over. When recently implicated in trafficking from the endosome (Derivery
cells expressing actin-GFP were exposed to latrunculin, a drug et al., 2009; Gomez and Billadeau, 2009; Duleh and Welch,
that prevents actin polymerization, endosomal actin fluores- 2010), was also clearly localized to B2AR tubules (Figure 4G).
cence became indistinguishable from the ‘‘background’’ cyto- Together, these data suggest that an Arp2/3-, cortactin- and
plasmic fluorescence within 16–18 s after drug exposure (e.g., WASH-based machinery mediates dynamic actin assembly on
in Figure 4A). When quantified across multiple cells, endosomal the endosome.
actin fluorescence showed an exponential loss after latrunculin
exposure, with a t1/2 of 3.5 s (99% Confidence Interval = 3.0 to B2AR-Containing Tubules Are a Specialized Subset
4.1 s) (Figure 4B), indicating that endosomal actin turned over of Recycling Tubules on the Endosome
quite rapidly. As a control, stress fibers, which are composed Since the traditional view is that the endosomal tubules that
of relatively stable capped actin filaments, were turned over mediate direct recycling to the plasma membrane are a uniform
more slowly in these same cells (e.g., in Figure S2A). Endosomal population, we next tested whether these tubules were the same
actin was lost in >98% of cells within 30 s after latrunculin, in as those that recycle bulk cargo. When B2AR recycling was visu-
contrast to stress fibers, which persisted for over 2 min in alized along with bulk recycling of TfR, endosomes containing
>98% of cells (Figure S2B). Rapid turnover of endosomal actin both cargo typically extruded three to four tubules containing
was also independently confirmed by fluorescence recovery TfR. Strikingly, however, only one of these contained detectable
after photobleaching (FRAP) studies. When a single endosomal amounts of B2AR (Example in Figure 5A, quantified in Figure 5B).
actin spot was bleached, the fluorescence recovered rapidly This was consistent with fast 3D confocal live cell imaging of
within 20 s (Figure 4C). As a control for more stable actin fila- B2AR in endosomes, which showed that most endosomes
ments, stress fibers showed little recovery of fluorescence after extruded only one B2AR containing tubule, with a small fraction
bleaching in this interval (Figure 4C). Exponential curve fits containing two. When quantified, only 24.4% of all TfR tubules
yielding a t1/2 of 8.26 s (99% CI = 7.65 to 8.97 s), consistent contained detectable B2AR (n = 358 tubules).
with rapid actin turnover (Figure 4D). In contrast, only part of
the fluorescence (30%) was recovered in stress fibers in the B2AR Tubules Are a Kinetically and Biochemically
same cells by 20 s, with curve fits yielding a t1/2 of 50.35 s Distinct from Bulk Recycling Tubules
(99% CI = 46.05 to 55.54 s). These results indicate that actin is When the lifetimes of tubules were quantified, the majority
dynamically assembled on the B2AR recycling tubules. (>80%) of B2AR tubules lasted more than 30 s. In contrast, the
Considering the rapid turnover of actin, we next explored the majority of TfR tubules devoid of B2AR lasted less than 30 s
machinery responsible for localizing actin at the tubule. (Figures 5B and 5C, Movie S8). Each endosome extruded
The Arp2/3 complex is a major nucleator of dynamic actin poly- several tubules containing TfR, only a subset (30%) of which
merization that has been implicated in polymerization-based en- were marked by actin, coronin, or cortactin (Figures 5D and
dosome motility (Stamnes, 2002; Girao et al., 2008; Pollard, 5E, arrows). Time-lapse movies indicated that the highly tran-
2007). Arp3, an integral part of the Arp2/3 complex useful for sient TfR-containing tubules were extruded from endosomal
visualizing this complex in intact cells (Merrifield et al., 2004), domains that were lacking cortactin (Figure 5E, arrows), while
was specifically concentrated at the base of the B2AR tubules the relatively stable B2AR containing tubules were marked by
on the endosome (e.g., in Figure 4E and fluorescence trace in cortactin (Figure 5E, arrowheads). Importantly, the relative
Figure 4F, Movie S7). Every B2AR tubule observed had a corre- stability of the subset of tubules was conferred by the actin cyto-
sponding Arp3 spot at its base (n = 200). Surprisingly, however, skeleton, as disruption of actin using latrunculin virtually abol-
we did not see N-WASP and WAVE-2, canonical members of the ished the stable fraction of TfR tubules (Figures 5B and 5C).

Cell 143, 761–773, November 24, 2010 ª2010 Elsevier Inc. 765
Figure 4. Actin on B2AR Tubules Is Dynamic and Arp2/3-Nucleated
(A) Cells expressing actin-GFP imaged live after treatment with 10 mM latrunculin for the indicated times, show rapid loss of endosomal actin. A time series of the
boxed area, showing several endosomal actin loci, is shown at the lower panel.
(B) The change in endosomal and cytoplasmic actin fluorescence over time after latrunculin normalized to initial endosomal actin fluorescence (n = 10). One-
phase exponential curve fits (solid lines) show a t1/2 of 3.5 s for actin loss (R2 = 0.984, d.f = 23, Sy.x = 2.1 for endosomal actin, R2 = 0.960, d.f = 23, Sy.x =
1.9 for cytoplasmic). Endosomal and cytoplasmc actin fluorescence becomes statistically identical within 15 s after latrunculin. Error bars denote SEM.
(C) Time series showing FRAP of representative examples of endosomal actin (top) and stress fibers (bottom).
(D) Kinetics of FRAP of actin (mean ± s.e.m) quantified from 14 endosomes and 17 stress fibers. One-phase exponential curve fits (lines), show a t1/2 of 8.26 s for
endosomal actin (R2 = 0.973, d.f = 34, Sy.x = 4.8) and 50.35 s for stress fibers (R2 = 0.801, d.f = 34, Sy.x = 3.9).

766 Cell 143, 761–773, November 24, 2010 ª2010 Elsevier Inc.
Together, these results suggest that sequence-dependent sequence-dependent concentration of B2AR into these tubules.
recycling of B2AR is mediated by specialized tubules that are Consistent with this, B2AR was no longer concentrated in endo-
kinetically and biochemically distinct from the bulk recycling somal tubules when endosomal actin was acutely removed
tubules containing only TfR. using latrunculin (e.g., in Figure 6A). When the pixel fluorescence
along the limiting membrane of multiple endosomes was quanti-
A Kinetic Model for Sorting of B2AR into a Subset fied, B2AR was distributed more uniformly along the endosomal
of Endosomal Tubules membrane in the absence of actin (Figures 6B and 6C). We
The relative stability of B2AR tubules suggested a simple model, further confirmed this by comparing the variance in B2AR fluo-
based on kinetic sorting, for how sequence-dependent cargo rescence along the endosomal perimeter, irrespective of their
was sorted into a specific subset of tubules and excluded from orientation. B2AR fluorescence was significantly more uniform
the transient TfR-containing bulk-recycling tubules. We hypoth- in endosomes without actin (Figure 6D), indicating that actin
esized that B2AR diffuses more slowly on the endosomal was required for endosomes to concentrate B2AR in microdo-
membrane relative to bulk recycling cargo. The short lifetimes mains. Less than 20% of endosomes showed B2AR-containing
of the bulk-recycling tubules would then create a kinetic barrier tubules in the absence of endosomal actin, in contrast to control
for B2AR entry, while this barrier would be overcome in the cells where over 75% of endosomes showed B2AR-containing
subset of tubules stabilized by actin. tubules (Figure 6E). Further, cytochalasin D, a barbed-end
To test the key prediction of this model, that B2AR diffuses capping drug that prevents further actin polymerization but
more slowly than TfR on the endosomal membrane, we directly does not actively cause depolymerization, also inhibited B2AR
measured the diffusion rates of B2AR and TfR using FRAP. When entry into tubules (Figure 6E) and B2AR surface recycling
B2AR or TfR was bleached on a small part of the endosomal (Figure S4A). Neither TfR tubules on endosomes (Figure 6E)
membrane, B2AR fluorescence took significantly longer to nor TfR recycling (Figure S4B) was inhibited by actin depolymer-
recover than TfR (Figure 5F). When quantified, the rate of ization, consistent with a role for actin specifically in sequence-
recovery of fluorescence of B2AR (t1/2 = 25.77 s, 99% CI 23.45 dependent recycling of B2AR (Cao et al., 1999). Further, deple-
to 28.6 s) was 4 times slower than that of TfR (t1/2 = 6.21 s, tion of cortactin using siRNA (Figure 6F) also inhibited B2AR
99% CI 5.49 to 7.17 s), indicating that B2AR diffuses significantly entry into tubules (Figures 6G and 6H). This inhibition was
slower on the endosomal membrane than TfR (Figures 5F and specific to cortactin depletion, as it was rescued by exogenous
5G). Neither B2AR or TfR recovered within the time analyzed expression of cortactin (Figure 6H). Together, these results indi-
when the whole endosome was bleached (Figure 5H), confirming cate that a localized actin cytoskeleton concentrates sequence-
that the recovery of fluorescence was due to diffusion from the dependent recycling cargo into a specific subset of recycling
unbleached part of the endosome and not due to delivery of tubules on the endosome.
new receptors via trafficking. Further, B2AR on the plasma
membrane diffused much faster than on the endosome (t1/2 = B2AR Sorting into the Recycling Subdomains
6.45 s, 99% CI 5.62 to 7.66 s), comparable to TfR, suggesting Is Mediated by Its C-Terminal PDZ-Interacting Domain
that B2AR diffusion was slower specifically on the endosome We next asked whether this actin-dependent concentration of
(Figure 5H). receptors into endosomal tubules depended on the PDZ-inter-
We next tested whether the diffusion of B2AR into endosomal acting sequence present in the B2AR cytoplasmic tail that medi-
tubules was slower than that of TfR, by using the rate of increase ates sequence-dependent recycling (Cao et al., 1999; Gage
of B2AR fluorescence as an index of receptor entry into tubules. et al., 2005). To test if the sequence was required, we used
B2AR fluorescence continuously increased throughout the dura- a mutant B2AR (B2AR-ala) in which the recycling sequence
tion of the tubule lifetimes (Figure S3A). Further, in a single tubule was specifically disrupted by the addition of a single alanine
containing TfR and B2AR, TfR fluorescence reached its (Cao et al., 1999). Unlike B2AR, internalized B2AR-ala was not
maximum at a markedly faster rate than that of B2AR (Fig- able to enter the tubular domains in the endosome (e.g., in
ure S3B). Together, these results suggest that slow diffusion of Figure 6I, quantified in Figure 6J), or recycle to the cell surface
B2AR on the endosome and stabilization of recycling tubules (Figure S4). To test if this sequence was sufficient, we used
by actin can provide a kinetic basis for specific sorting of a chimeric DOR construct with the B2AR-derived recycling
sequence-dependent cargo into subsets of endosomal tubules. sequence fused to its cytoplasmic tail, termed DOR-B2 (Gage
et al., 2005), which recycles much more efficiently than DOR
Local Actin Assembly Is Required for B2AR Entry (Figure S4). In contrast to DOR, which showed little concentra-
into the Subset of Tubules tion in endosomal tubules, DOR-B2 entered tubules (Figures 6I
Because actin stabilizes the B2AR-containing subset of tubules, and 6J) and recycled in an actin-dependent manner similar to
the model predicts that endosomal actin would be required for B2AR (Figure S4D). Together, these results indicate that the

(E) Example endosomes in live cells coexpressing B2AR and Arp3-GFP showing Arp3 at the base of B2AR tubules (arrowhead in the inset).
(F) Trace of linear pixel fluorescence of B2AR and Arp3 shows Arp3 specifically on the endosomal tubule.
(G) Example endosomes from cells coexpressing B2AR and N-WASP-, WAVE2-, cortactin-, or WASH-GFP. N-WASP and WAVE2 were not detected on endo-
somes, while cortactin and WASH were concentrated at the B2AR tubules (arrowheads).
Scale bars represent 1 mm. See also Figure S2 and Movie S7.

Cell 143, 761–773, November 24, 2010 ª2010 Elsevier Inc. 767
Figure 5. B2AR Is Enriched Specifically in a Subset of Endosomal Tubules that Are Stabilized by Actin
(A) A representative example of an endosome with two tubules containing TfR, only one of which is enriched for B2AR.
(B) The number of tubules with B2AR, TfR, and TfR in the presence of 10 mM latrunculin, per endosome per min, binned into lifetimes less than or more than 30 s,
quantified across 28 endosomes and 281 tubules.
(C) The percentages of B2AR, TfR, and TfR + latrunculin tubules with lifetimes less than or more than 30 s, normalized to total number of tubules in each case.
(D) An example endosome containing TfR and coronin, showing that coronin is present on a subset of the TfR tubules. Arrowheads indicate a TfR tubule that is
marked by coronin, and arrows show a TfR tubule that is not.
(E) Time lapse series showing TfR-containing tubules extruding from endosomal domains without detectable cortactin. Arrowheads indicate a relatively stable
TfR tubule that is marked by coronin, and arrows denote rapid transient TfR tubules without detectable cortactin.
(F) Frames from a representative time lapse movie showing FRAP of B2AR (top row) or TfR (bottom row). The circles mark the bleached area of the endosome. TfR
fluorescence recovers rapidly, while B2AR fluorescence recovers slowly.
(G) Fluorescence recovery of B2AR (red circles) and TfR (green diamonds) on endosomes quantified from 11 experiments. Exponential fits (solid lines) show that
B2AR fluorescence recovers with a t1/2 of 25.77 s (R2 = 0.83, d.f = 37, Sy.x = 6.3), while TfR fluorescence recovers with a t1/2 of 6.21 s (R2 = 0.91, d.f = 30, Sy.x = 7.1).

768 Cell 143, 761–773, November 24, 2010 ª2010 Elsevier Inc.
PDZ-interacting recycling sequence on B2AR was both required selective control without affecting the recycling of constitutively
and sufficient to mediate concentration of receptors in the actin- cycling nutrient receptors. Further, such physical separation
stabilized endosomal tubular domains. might also reflect the differences in molecular requirements
As PDZ-domain interactions have been established to indi- that have been observed between bulk and sequence-depen-
rectly link various integral membrane proteins to cortical actin dent recycling (Hanyaloglu and von Zastrow, 2007).
(Fehon et al., 2010), we tested whether linking DOR to actin Endosome-associated actin likely plays a dual role in endoso-
was sufficient to drive receptor entry into endosomal tubules. mal sorting, both of which contribute to sequence-dependent
Remarkably, fusion of the actin-binding domain of the ERM entry of cargo selectively into special domains. First, by stabi-
protein ezrin (Turunen et al., 1994) to the C terminus of DOR lizing the specialized endosomal tubules relative to the much
was sufficient to localize the receptor (termed DOR-ABD) to more dynamic tubules that mediate bulk recycling, the local actin
endosomal tubules (Figure 6J). The surface recycling of B2AR, cytoskeleton could allow sequence-dependent cargo to
DOR-B2, and DOR-ABD were dependent on the presence of overcome a kinetic barrier that limits their entry into the bulk
an intact actin cytoskeleton (Figure S4), consistent with previous pathway. Supporting this, we show that most endosomal tubules
publications (Cao et al., 1999; Gage et al., 2005; Lauffer et al., are highly transient, lasting less than a few seconds (Figures 5B
2009). Further, transplantation of the actin-binding domain was and 5C), which allows enough time for entry of the fast-diffusing
also sufficient to specifically confer recycling to a version of bulk recycling cargo, but not the slow-diffusing sequence-
B2AR lacking its native recycling signal (Figure S4F). These dependent cargo (Figures 5F and 5G), into these tubules.
results indicate that the concentration of B2AR in the actin-stabi- A subset of these tubules representing the sequence-dependent
lized recycling tubules is mediated by linking receptors to the recycling pathway is stabilized by the presence of an actin cyto-
local actin cytoskeleton through PDZ interactions. skeleton (Figures 5B and 5C). This stabilization allows time for
B2AR to diffuse into these tubules (Figure S3), which eventually
DISCUSSION pinch off membranes that can directly fuse with the plasma
membrane (Figure 2). Interestingly, inhibition of actin caused
Even though endocytic receptor sorting was first appreciated a decrease in the total number of tubules by approximately
over two decades ago (e.g., Brown et al., 1983; Farquhar, 25% (Figure 5B), suggesting that the actin cytoskeleton plays
1983; Steinman et al., 1983), our understanding of the principles a role in maintaining the B2AR-containing subset of tubules,
of this process has been limited. A major reason for this has been and not just in the sorting of B2AR into these tubules.
the lack of direct assays to visualize signaling receptor sorting in Second, a local actin cytoskeleton could provide the
the endosome. Here we directly visualized, in living cells, endo- machinery for active concentration of recycling proteins like
somal sorting between two prototypic members of the largest the B2AR, which interact with actin-associated sorting proteins
known family of signaling receptors for which sequence-specific (ERM and ERM-binding proteins) through C-terminal sequences
recycling is critical for physiological regulation of cell signaling (Weinman et al., 2006; Wheeler et al., 2007; Lauffer et al., 2009;
(Pippig et al., 1995; Lefkowitz et al., 1998; Xiang and Kobilka, Fehon et al., 2010), in specialized recycling tubules. Consistent
2003). We resolve sorting at the level of single trafficking events with this, the C-terminal sequence on B2AR was both required
on individual endosomes, and define a kinetic and affinity-based and sufficient for sorting to the endosome and for recycling,
model for how sequence-dependent receptors are sorted away and a distinct actin-binding sequence was sufficient for both
from bulk-recycling and degrading proteins. receptor entry into tubules and recycling (Figure 6 and Figure S4).
By analyzing individual sorting and recycling events on single PDZ-interacting sequences have been identified on several
endosomes, we demonstrate a remarkable diversity in recycling signaling receptors, including multiple GPCRs, with different
pathways emanating from the same organelle (Scita and Di specificities for distinct PDZ-domain proteins (Weinman et al.,
Fiore, 2010). The traditional view has been that recycling to the 2006). Further, actin-stabilized subsets of tubules were present
plasma membrane is mediated by a uniform set of endosomal even in the absence of B2AR in the endosome. We propose
tubules from a single endosome. In contrast to this view, we that, using a combination of kinetic and affinity-based sorting
demonstrate that the recycling pathway is highly specialized, principles, discrete Actin-Stabilized SEquence-dependent
and that specific cargo can segregate into specialized subsets Recycling Tubule (ASSERT) domains could thus mediate effi-
of tubules that are biochemically, biophysically, and functionally cient sorting of sequence-dependent recycling cargo away
distinct. Receptor recycling plays a critical role in controlling the from both degradation and bulk recycling pathways that diverge
rate of cellular re-sensitization to signals (Lefkowitz et al., 1998; from the same endosomes.
Sorkin and von Zastrow, 2009), and recent data suggest that Our results, therefore, uncover an additional role for actin poly-
the sequence-dependent recycling of signaling receptors is merization in endocytic sorting, separate from its role in endo-
selectively controlled by signaling pathways (Yudowski et al., some motility. It will be interesting to investigate the mechanism
2009). The physical separation between bulk and sequence- and signals that control the nucleation of such a spatially local-
dependent recycling that we demonstrate here allows for such ized actin cytoskeleton on the endosome. The lack of obvious

(H) Fluorescence recovery of B2AR (blue triangles) and TfR (green diamonds) on endosomes when the whole endosome was bleached, or of B2AR on the cell
surface (red circles) quantified from 12 experiments. B2AR fluorescence on the surface recovers with a t1/2 of 6.49 s (R2 = 0.94, d.f = 27, Sy.x = 8.1).
Error bars denote SEM. Scale bars represent 1 mm. See also Figure S3 and Movie S8.

Cell 143, 761–773, November 24, 2010 ª2010 Elsevier Inc. 769
Figure 6. B2AR Enrichment in Tubules Depends on Endosomal Actin and a PDZ-Interacting Sequence on the B2AR Cytoplasmic Domain
(A) Representative fields from B2AR-expressing cells exposed to isoproterenol showing B2AR endosomes before (top panel) or after (bottom panel) exposure to
10 mM latrunculin for 5 min. Tubular endosomal domains enriched in B2AR (arrowheads) are lost upon exposure to latrunculin.
(B) Schematic of measurement of endosomal B2AR fluorescence profiles in the limiting membrane. The profile was measured in a clockwise manner starting from
the area diametrically opposite the tubule (an angle of 0 ).
(C) B2AR concentration along the endosomal membrane, calculated from fluorescence profiles of 20 endosomes, normalized to the average endosomal B2AR
fluorescence. In the presence of latrunculin, B2AR enrichment in tubules is abolished, and B2AR fluorescence shows little variation along the endosomal
membrane.
(D) Variance in endosomal B2AR fluorescence values measured before and after latrunculin. B2AR distribution becomes more uniform after latrunculin.
(E) The percentages of endosomes extruding B2AR-containing tubules, calculated before (n = 246) and after (n = 106) treatment with latrunculin, or before
(n = 141) and after (n = 168) cytochalasin-D, show a significant reduction after treatment with either drug. As a control, the percentages of endosomes extruding
TfR-containing tubules before (n = 317) and after (n = 286), respectively, are shown.
(F) Cortactin immunoblot showing reduction in protein levels after siRNA.
(G) Representative fields from B2AR-containing endosomes in cells treated with control and cortactin siRNA. Arrowheads denote endosomal tubules in the
control siRNA-treated cells.
(H) Percentages of endosomes extruding B2AR tubules calculated in control siRNA-treated cells (n = 210), cortactin siRNA-treated cells (n = 269), and cortactin
siRNA-treated cells expressing an siRNA-resistant cortactin (n = 250).
(I) Representative examples of endosomes from agonist-exposed cells expressing B2AR, B2AR-ala, DOR, or DOR-B2. Arrowheads denote receptor-containing
tubules on B2AR and DOR-B2 endosomes.
(J) The percentage of endosomes with tubular domains containing B2AR, B2AR-ala, DOR, DOR-B2, or DOR-ABD (n = 246, 302, 137, 200, and 245, respectively)
were quantified.
Scale bars represent 1 mm; and error bars represent SEM. See also Figure S4.

770 Cell 143, 761–773, November 24, 2010 ª2010 Elsevier Inc.
concentration of the canonical Arp2/3 activators, WASP and mere), or an Andor Revolution XD Spinning disk system on a Nikon Ti micro-
WAVE, suggests a novel mode of actin nucleation involving cor- scope. A 488 nm Ar laser and a 568 nm Ar/Kr laser (Melles Griot), or 488 nm
and 561 nm solid-state lasers (Coherent) were used as light sources. Cells
tactin. Cortactin can act as a nucleation-promoting factor for
were imaged in Opti-MEM (GIBCO) with 2% serum and 30 mM HEPES
Arp2/3, at least in vitro (Ammer and Weed, 2008), and can (pH 7.4), maintained at 37 C using a temperature-controlled incubation
interact with dynamin (Schafer et al., 2002; McNiven et al., chamber. Time lapse images were acquired with a Cascade II EM-CCD
2000), which makes it an attractive candidate for coordinating camera (Photometrics) driven by MicroManager (www.micro-manager.org)
actin dynamics on membranes. Interestingly, inhibition of or an Andor iXon+ EM-CCD camera using iQ (Andor). The same lasers were
WASH, a recently described Arp regulator that is present on used as sources for bleaching in FRAP experiments. Structured illumination
microscopy was performed as described earlier (Gustafsson et al., 2008).
B2AR tubules, has been reported to result in an increase in endo-
somal tubules (Derivery et al., 2009). Although its role in
sequence-dependent recycling remains to be tested, this Electron Microscopy
suggests the presence of multiple actin-associated proteins EM studies were carried out using MDCK cells because they are amenable to
with distinct functions on the endosome. a previously described pre-embedding processing that facilitates detection of
cytoplasmic actin filaments (Ikonen et al., 1996; Parton et al., 1991), and
The simple kinetic and affinity-based principle that we
because they contain morphologically similar endosomes to HEK293 cells.
propose likely provides a physical basis for sequence-depen- Cells were grown on polycarbonate filters (Transwell 3412; Costar, Cam-
dent sorting of internalized membrane proteins between essen- bridge, MA) for 4 days as described previously (Parton et al., 1991). To allow
tially opposite fates in distinct endosomal domains. Proteins that visualization of early endosomes and any associated filaments a pre-embed-
bind sequence-dependent degrading receptors and are required ding approach was employed. Cells were incubated with HRP (Sigma type II,
for their degradation (Whistler et al., 2002; Marley and von 10mg/ml) in the apical and basolateral medium for 10min at 37 C and then
washed, perforated, and immunogold labeled with a rabbit anti- actin anti-
Zastrow, 2010) might act as scaffolds and provide a similar
body, a gift of Professor Jan de Mey (Strasbourg), followed by 9nm protein
kinetic barrier to prevent them from accessing the rapid bulk-re-
A-gold. HRP visualization and epon embedding was as described previously
cycling tubules. Entry of these receptors into the involution (Parton et al., 1991; Ikonen et al., 1996).
pathway might then be accelerated by their association with
the well-characterized ESCRT-associated domains on the vacu-
Image and Data Analysis
olar portion of endosomes (Hurley, 2008; Saksena et al., 2007;
Acquired image sequences were saved as 16-bit tiff stacks, and quantified
Williams and Urbé, 2007), complementary to the presently iden- using ImageJ (http://rsb.info.nih.gov/ij/). For estimating receptor enrichment,
tified ASSERT domains on a subset of endosomal tubules. a circular mask 5 px in diameter was used to manually select the membrane
Such diversity at the level of individual trafficking events to the at the base of the tubule or membranes derived from endosomes. Fluores-
same destination from the same organelle raises the possibility cence values measured were normalized to that of the endosomal membrane
that there exists yet further specialization among the pathways devoid of tubules. An area of the coverslip lacking cells was used to estimate
background fluorescence. For estimating linear pixel values along the tubules,
that mediate exit out of the endosome, including in the degrada-
a line selection was drawn along the tubule and across the endosome, and the
tive pathway and the retromer-based pathway to the trans-Golgi
Plot Profile function used to measure pixel values. For obtaining the average
network. Importantly, the physical separation in pathways that value plot across multiple sorting events, the linear pixels were first normalized
we report here potentially allows for cargo-mediated regulation to the diameter of the endosome and then averaged. To generate pixel values
as a mode for controlling receptor recycling to the plasma along the endosomal limiting membranes, the Oval Profile plugin, with 60
membrane. Such a mechanism can provide virtually an unlimited segments, was used after manually selecting the endosomal membrane using
level of selectivity in the post-endocytic system using minimal an oval ROI. Lifetimes of tubules were calculated by manually tracking the
extension and retraction of tubules over time-lapse series. Microsoft Excel
core trafficking machineries, as has been observed for endocy-
was used for simple data analyses and graphing. Curve fits of data were per-
tosis at the cell surface (Puthenveedu and von Zastrow, 2006). formed using GraphPad Prism. All P-values are from two-tailed Mann-Whitney
As the principles of such sorting depend critically on kinetics, tests unless otherwise noted.
the high-resolution imaging used here to analyze domain kinetics
and biochemistry, and to achieve single-event resolution in living
SUPPLEMENTAL INFORMATION
cells, provides a powerful method to elucidate biologically
important sorting processes in the future. Supplemental Information includes four figures and eight movies and can be
found with this article online at doi:10.1016/j.cell.2010.10.003.
EXPERIMENTAL PROCEDURES

Constructs and Reagents ACKNOWLEDGMENTS


Receptor constructs and stably transfected HEK293 cell lines are described
previously (Gage et al., 2005; Lauffer et al., 2009) Transfections were per- The majority of the imaging was performed at the Nikon Imaging Center at
formed using Effectene (QIAGEN) according to manufacturer’s instructions. UCSF. We thank David Drubin, Matt Welch, John Sedat, Aylin Hanyaloglu,
For visualizing receptors, FLAG-tagged receptors were labeled with M1 anti- Aaron Marley, and James Hislop for essential reagents and valuable help.
bodies (Sigma) conjugated with Alexa-555 (Invitrogen) as described (Gage M.A.P. was supported by a K99/R00 grant DA024698, M.v.Z. by an R37 grant
et al., 2005), or fusion constructs were generated where receptors were DA010711, and O.D.W. by an RO1 grant GM084040, all from the NIH. J.T. is an
tagged on the N-terminus with GFP. Latrunculin and Cytochalasin D (Sigma) investigator of the Howard Hughes Medical Institute.
were used at 10 mM final concentration.
Received: October 31, 2009
Live-Cell and Fluorescence Imaging Revised: April 7, 2010
Cells were imaged using a Nikon TE-2000E inverted microscope with a 1003 Accepted: September 27, 2010
1.49 NA TIRF objective (Nikon) and a Yokagawa CSU22 confocal head (Sola- Published: November 24, 2010

Cell 143, 761–773, November 24, 2010 ª2010 Elsevier Inc. 771
REFERENCES Lefkowitz, R.J., Pitcher, J., Krueger, K., and Daaka, Y. (1998). Mechanisms of
beta-adrenergic receptor desensitization and resensitization. Adv. Pharmacol.
Ammer, A.G., and Weed, S.A. (2008). Cortactin branches out: roles in regu- 42, 416–420.
lating protrusive actin dynamics. Cell Motil. Cytoskeleton 65, 687–707. Marchese, A., Paing, M.M., Temple, B.R., and Trejo, J. (2008). G protein-
Brown, M.S., Anderson, R.G., and Goldstein, J.L. (1983). Recycling receptors: coupled receptor sorting to endosomes and lysosomes. Annu. Rev. Pharma-
the round-trip itinerary of migrant membrane proteins. Cell 32, 663–667. col. Toxicol. 48, 601–629.
Benink, H.A., and Bement, W.M. (2005). Concentric zones of active RhoA and Marley, A., and von Zastrow, M. (2010). Dysbindin promotes the post-endo-
Cdc42 around single cell wounds. J. Cell Biol. 168, 429–439. cytic sorting of G protein-coupled receptors to lysosomes. PLoS ONE 5,
Cao, T.T., Deacon, H.W., Reczek, D., Bretscher, A., and von Zastrow, M. e9325.
(1999). A kinase-regulated PDZ-domain interaction controls endocytic sorting Maxfield, F.R., and McGraw, T.E. (2004). Endocytic recycling. Nat. Rev. Mol.
of the b2-adrenergic receptor. Nature 401, 286–290. Cell Biol. 5, 121–132.
Cullen, P.J. (2008). Endosomal sorting and signalling: an emerging role for Mayor, S., Presley, J.F., and Maxfield, F.R. (1993). Sorting of membrane
sorting nexins. Nat. Rev. Mol. Cell Biol. 9, 574–582. components from endosomes and subsequent recycling to the cell surface
Daly, R.J. (2004). Cortactin signalling and dynamic actin networks. Biochem. J. occurs by a bulk flow process. J. Cell Biol. 121, 1257–1269.
382, 13–25. McNiven, M.A., Kim, L., Krueger, E.W., Orth, J.D., Cao, H., and Wong, T.W.
Derivery, E., Sousa, C., Gautier, J.J., Lombard, B., Loew, D., and Gautreau, A. (2000). Regulated interactions between dynamin and the actin-binding protein
(2009). The Arp2/3 activator WASH controls the fission of endosomes through cortactin modulate cell shape. J. Cell Biol. 151, 187–198.
a large multiprotein complex. Dev. Cell 17, 712–723. Merrifield, C.J., Qualmann, B., Kessels, M.M., and Almers, W. (2004). Neural
Duleh, S.N., and Welch, M.D. (2010). WASH and the Arp2/3 complex regulate Wiskott Aldrich Syndrome Protein (N-WASP) and the Arp2/3 complex are re-
endosome shape and trafficking. Cytoskeleton (Hoboken) 67, 193–206. cruited to sites of clathrin-mediated endocytosis in cultured fibroblasts. Eur.
Dunn, K.W., and Maxfield, F.R. (1992). Delivery of ligands from sorting endo- J. Cell Biol. 83, 13–18.
somes to late endosomes occurs by maturation of sorting endosomes. Miesenböck, G., De Angelis, D.A., and Rothman, J.E. (1998). Visualizing secre-
J. Cell Biol. 117, 301–310. tion and synaptic transmission with pH-sensitive green fluorescent proteins.
Farquhar, M.G. (1983). Intracellular membrane traffic: pathways, carriers, and Nature 394, 192–195.
sorting devices. Methods Enzymol. 98, 1–13. Millard, T.H., Sharp, S.J., and Machesky, L.M. (2004). Signalling to actin
Fehon, R.G., McClatchey, A.I., and Bretscher, A. (2010). Organizing the cell assembly via the WASP (Wiskott-Aldrich syndrome protein)-family proteins
cortex: the role of ERM proteins. Nat. Rev. Mol. Cell Biol. 11, 276–287. and the Arp2/3 complex. Biochem. J. 380, 1–17.
Gage, R.M., Matveeva, E.A., Whiteheart, S.W., and von Zastrow, M. (2005). Parton, R.G., Dotti, C.G., Bacallao, R., Kurtz, I., Simons, K., and Prydz, K.
Type I PDZ ligands are sufficient to promote rapid recycling of G Protein- (1991). pH-induced microtubule-dependent redistribution of late endosomes
coupled receptors independent of binding to N-ethylmaleimide-sensitive in neuronal and epithelial cells. J. Cell Biol. 113, 261–274.
factor. J. Biol. Chem. 280, 3305–3313. Perrais, D., and Merrifield, C.J. (2005). Dynamics of endocytic vesicle creation.
Girao, H., Geli, M.I., and Idrissi, F.Z. (2008). Actin in the endocytic pathway: Dev. Cell 9, 581–592.
from yeast to mammals. FEBS Lett. 582, 2112–2119. Piper, R.C., and Katzmann, D.J. (2007). Biogenesis and function of multivesic-
Gomez, T.S., and Billadeau, D.D. (2009). A FAM21-containing WASH complex ular bodies. Annu. Rev. Cell Dev. Biol. 23, 519–547.
regulates retromer-dependent sorting. Dev. Cell 17, 699–711. Pippig, S., Andexinger, S., and Lohse, M.J. (1995). Sequestration and
Gustafsson, M.G., Shao, L., Carlton, P.M., Wang, C.J., Golubovskaya, I.N., recycling of beta 2-adrenergic receptors permit receptor resensitization.
Cande, W.Z., Agard, D.A., and Sedat, J.W. (2008). Three-dimensional resolu- Mol. Pharmacol. 47, 666–676.
tion doubling in wide-field fluorescence microscopy by structured illumination. Pollard, T.D. (2007). Regulation of actin filament assembly by Arp2/3 complex
Biophys. J. 94, 4957–4970. and formins. Annu. Rev. Biophys. Biomol. Struct. 36, 451–477.
Hanyaloglu, A.C., and von Zastrow, M. (2007). A novel sorting sequence in the Puthenveedu, M.A., and von Zastrow, M. (2006). Cargo regulates clathrin-
beta2-adrenergic receptor switches recycling from default to the Hrs-depen- coated pit dynamics. Cell 127, 113–124.
dent mechanism. J. Biol. Chem. 282, 3095–3104.
Saksena, S., Sun, J., Chu, T., and Emr, S.D. (2007). ESCRTing proteins in the
Hanyaloglu, A.C., and von Zastrow, M. (2008). Regulation of GPCRs by endo-
endocytic pathway. Trends Biochem. Sci. 32, 561–573.
cytic membrane trafficking and its potential implications. Annu. Rev. Pharma-
col. Toxicol. 48, 537–568. Schafer, D.A., Weed, S.A., Binns, D., Karginov, A.V., Parsons, J.T., and
Cooper, J.A. (2002). Dynamin2 and cortactin regulate actin assembly and fila-
Hanyaloglu, A.C., McCullagh, E., and von Zastrow, M. (2005). Essential role of
ment organization. Curr. Biol. 12, 1852–1857.
Hrs in a recycling mechanism mediating functional resensitization of cell
signaling. EMBO J. 24, 2265–2283. Scita, G., and Di Fiore, P.P. (2010). The endocytic matrix. Nature 463, 464–473.

Hurley, J.H. (2008). ESCRT complexes and the biogenesis of multivesicular Shinozaki-Narikawa, N., Kodama, T., and Shibasaki, Y. (2006). Cooperation of
bodies. Curr. Opin. Cell Biol. 20, 4–11. phosphoinositides and BAR domain proteins in endosomal tubulation. Traffic
7, 1539–1550.
Ikonen, E., Parton, R.G., Lafont, F., and Simons, K. (1996). Analysis of the role
of p200-containing vesicles in post-Golgi traffic. Mol. Biol. Cell 7, 961–974. Sorkin, A., and von Zastrow, M. (2009). Endocytosis and signalling: intertwin-
ing molecular networks. Nat. Rev. Mol. Cell Biol. 10, 609–622.
Kaksonen, M., Peng, H.B., and Rauvala, H. (2000). Association of cortactin
with dynamic actin in lamellipodia and on endosomal vesicles. J. Cell Sci. Stamnes, M. (2002). Regulating the actin cytoskeleton during vesicular trans-
113, 4421–4426. port. Curr. Opin. Cell Biol. 14, 428–433.
Kaksonen, M., Toret, C.P., and Drubin, D.G. (2005). A modular design for the Steinman, R.M., Mellman, I.S., Muller, W.A., and Cohn, Z.A. (1983). Endocy-
clathrin- and actin-mediated endocytosis machinery. Cell 123, 305–320. tosis and the recycling of plasma membrane. J. Cell Biol. 96, 1–27.
Lauffer, B.E., Chen, S., Melero, C., Kortemme, T., von Zastrow, M., and Var- Traer, C.J., Rutherford, A.C., Palmer, K.J., Wassmer, T., Oakley, J., Attar, N.,
gas, G.A. (2009). Engineered protein connectivity to actin mimics PDZ-depen- Carlton, J.G., Kremerskothen, J., Stephens, D.J., and Cullen, P.J. (2007).
dent recycling of G protein-coupled receptors but not its regulation by Hrs. SNX4 coordinates endosomal sorting of TfnR with dynein-mediated transport
J. Biol. Chem. 284, 2448–2458. into the endocytic recycling compartment. Nat. Cell Biol. 9, 1370–1380.

772 Cell 143, 761–773, November 24, 2010 ª2010 Elsevier Inc.
Turunen, O., Wahlström, T., and Vaheri, A. (1994). Ezrin has a COOH-terminal Williams, R.L., and Urbé, S. (2007). The emerging shape of the ESCRT
actin-binding site that is conserved in the ezrin protein family. J. Cell Biol. 126, machinery. Nat. Rev. Mol. Cell Biol. 8, 355–368.
1445–1453. Xiang, Y., and Kobilka, B.K. (2003). Myocyte adrenoceptor signaling path-
Uetrecht, A.C., and Bear, J.E. (2006). Coronins: the return of the crown. Trends ways. Science 300, 1530–1532.
Cell Biol. 16, 421–426. Yarar, D., Waterman-Storer, C.M., and Schmid, S.L. (2005). A dynamic actin
cytoskeleton functions at multiple stages of clathrin-mediated endocytosis.
Weinman, E.J., Hall, R.A., Friedman, P.A., Liu-Chen, L.Y., and Shenolikar, S.
Mol. Biol. Cell 16, 964–975.
(2006). The association of NHERF adaptor proteins with g protein-coupled
Yudowski, G.A., Puthenveedu, M.A., and von Zastrow, M. (2006). Distinct
receptors and receptor tyrosine kinases. Annu. Rev. Physiol. 68, 491–505.
modes of regulated receptor insertion to the somatodendritic plasma
Wheeler, D., Sneddon, W.B., Wang, B., Friedman, P.A., and Romero, G. membrane. Nat. Neurosci. 9, 622–627.
(2007). NHERF-1 and the cytoskeleton regulate the traffic and membrane Yudowski, G.A., Puthenveedu, M.A., Henry, A.G., and von Zastrow, M. (2009).
dynamics of G protein-coupled receptors. J. Biol. Chem. 282, 25076–25087. Cargo-mediated regulation of a rapid Rab4-dependent recycling pathway.
Whistler, J.L., Enquist, J., Marley, A., Fong, J., Gladher, F., Tsuruda, P., Mur- Mol. Biol. Cell 20, 2774–2784.
ray, S.R., and Von Zastrow, M. (2002). Modulation of postendocytic sorting of Zerial, M., and McBride, H. (2001). Rab proteins as membrane organizers. Nat.
G protein-coupled receptors. Science 297, 615–620. Rev. Mol. Cell Biol. 2, 107–117.

Cell 143, 761–773, November 24, 2010 ª2010 Elsevier Inc. 773
Mechanisms Determining the Morphology
of the Peripheral ER
Yoko Shibata,1,2 Tom Shemesh,3 William A. Prinz,4 Alexander F. Palazzo,1,5 Michael M. Kozlov,3,*
and Tom A. Rapoport1,2,*
1Howard Hughes Medical Institute
2Department of Cell Biology
Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
3Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel
4Laboratory of Cell Biochemistry and Biology, National Institute of Diabetes and Digestive and Kidney Disorders,

National Institute of Health, Bethesda, MD 02892, USA


5Department of Biochemistry, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada

*Correspondence: michk@post.tau.ac.il (M.M.K.), tom_rapoport@hms.harvard.edu (T.A.R.)


DOI 10.1016/j.cell.2010.11.007

SUMMARY the tubular ER network (Hu et al., 2008, 2009; Shibata et al.,
2008; Voeltz et al., 2006), but essentially nothing is known about
The endoplasmic reticulum (ER) consists of the how ER sheets are generated. In addition, it is unknown whether
nuclear envelope and a peripheral network of tubules proteins specifically segregate into ER sheets and whether there
and membrane sheets. The tubules are shaped by is a functional significance to the existence of different ER
the curvature-stabilizing proteins reticulons and morphologies.
DP1/Yop1p, but how the sheets are formed is ER tubules are characterized by high membrane curvature in
cross-section and are shaped by two families of curvature-stabi-
unclear. Here, we identify several sheet-enriched
lizing proteins, the reticulons and DP1/Yop1p (Voeltz et al.,
membrane proteins in the mammalian ER, including
2006). Members of both families are ubiquitously expressed in
proteins that translocate and modify newly synthe- all eukaryotic cells. These proteins localize to the ER tubules,
sized polypeptides, as well as coiled-coil membrane and their depletion leads to the loss of tubules. Conversely, the
proteins that are highly upregulated in cells with overexpression of certain isoforms results in long, unbranched
proliferated ER sheets, all of which are localized by tubules. Purified members of the two families deform reconsti-
membrane-bound polysomes. These results indicate tuted proteoliposomes into tubules (Hu et al., 2008). Together,
that sheets and tubules correspond to rough and these results indicate that the reticulons and DP1/Yop1p are
smooth ER, respectively. One of the coiled-coil both necessary and sufficient for ER tubule formation. These
proteins, Climp63, serves as a ‘‘luminal ER spacer’’ two protein families do not share sequence homology, but
and forms sheets when overexpressed. More univer- both have a conserved domain containing two long hydrophobic
segments that sit in the membrane as hairpins (Voeltz et al.,
sally, however, sheet formation appears to involve
2006). These hairpins may stabilize the high curvature of tubules
the reticulons and DP1/Yop1p, which localize to
in cross-section by forming a wedge in the lipid bilayer. In addi-
sheet edges and whose abundance determines the tion, oligomerization of these proteins may generate arc-like
ratio of sheets to tubules. These proteins may scaffolds around the tubules (Shibata et al., 2008).
generate sheets by stabilizing the high curvature of The peripheral ER sheets vary in size but always consist of two
edges. closely apposed membranes whose distance is approximately
the same as the diameter of the tubules (30 nm in yeast
INTRODUCTION [Bernales et al., 2006] and 50 nm in mammals). Consequently,
the edges of sheets have a similarly high curvature as the cross-
How the characteristic shape of a membrane-bound organelle is section of tubules. In ‘‘professional’’ secretory cells, such as
generated is a fundamental question in cell biology. We have plasma B cells or pancreatic cells, the ER sheets extend
started to address this question for the endoplasmic reticulum throughout the entire cell and are studded with membrane-
(ER), an organelle that has a particularly intriguing morphology. bound ribosomes. They are stacked tightly with regular
It is a continuous membrane system that is comprised of the distances between the membranes on both the cytoplasmic
nuclear envelope as well as of a peripheral network of tubules and luminal sides (Fawcett, 1981). By contrast, cells that do
and sheets (Baumann and Walz, 2001; Shibata et al., 2009; not secrete many proteins contain mostly tubular ER. These
Voeltz et al., 2002). Both the tubules and sheets are dynamic, observations have led to the idea that ER sheets correspond to
i.e., they are continuously forming and collapsing. Previous rough ER (Shibata et al., 2006), the region of the ER that contains
work has identified proteins that are responsible for shaping membrane-bound ribosomes, i.e., ribosomes associated with

774 Cell 143, 774–788, November 24, 2010 ª2010 Elsevier Inc.
the translocons, the sites of translocation and modification of component forming the protein-conducting channel in the ER,
newly synthesized secretory and membrane proteins. On the but due to its tagging with GFP and overexpression,
other hand, ER tubules would correspond to smooth ER (Shibata GFP-Sec61b is not associated with the translocon and distrib-
et al., 2006), the ER region devoid of ribosomes, which may be utes throughout the ER (Shibata et al., 2008). Antibodies recog-
specialized in lipid metabolism or Ca2+ signaling. While these nizing the luminal chaperones BiP and Grp94 (anti-KDEL) also
ideas are attractive, the tubular ER clearly contains membrane- stained the entire ER (Figure 1C, middle). The integral membrane
bound ribosomes, and a segregation of rough ER proteins into proteins calnexin and Bap31 showed a similar ubiquitous local-
sheets has not yet been demonstrated. ization as overexpressed GFP-Sec61b (Figure 1B and Figure S1
Several mechanisms of ER sheet formation have been consid- available online). These results suggest that many luminal and
ered. One possibility is that integral membrane proteins would membrane ER proteins do not localize to a specific ER domain,
form bridges across the luminal space of the ER (Senda and consistent with the continuity of the membrane system.
Yoshinaga-Hirabayashi, 1998; Shibata et al., 2009). A second Next, we tested the endogenous localization of components of
possibility is that proteins form flat cytoplasmic or luminal the translocon. In contrast to overexpressed GFP-Sec61b,
scaffolds, as suggested for the formation of flat Golgi cisternae endogenous Sec61b was found concentrated in ER sheets
(Short et al., 2005). It has also been proposed that the membrane when compared to the localization of the luminal ER proteins
association of ribosomes could directly be responsible for the BiP and GRP94 (Figure 1C), although some weak staining of
generation of ER sheets (Puhka et al., 2007). Finally, given that the tubular network and nonspecific staining of the cytoplasm
the reticulons and DP1/Yop1p generate high curvature were also seen. Because endogenous Sec61b is contained in
membranes, one might imagine that they generate sheets by the Sec61 complex, these data suggest that translocons are
stabilizing the sheet edges, bringing the apposing membranes enriched in ER sheets. This is supported by the localization of
in close proximity (Shibata et al., 2009). endogenous TRAPa, a component tightly associated with the
Here, we show that rough ER proteins partition into ER sheets. ribosome-bound Sec61 complex (Ménétret et al., 2008); TRAPa
This includes both proteins involved in translocation and modifi- was strongly enriched in the peripheral ER sheets (Figure 1D).
cation of newly synthesized polypeptides, as well as coiled-coil Finally, Dad1, a component of the translocon-associated
membrane proteins that are highly upregulated in cells contain- oligosaccharyl transferase complex that glycosylates nascent
ing proliferated ER sheets. Membrane-bound polysomes are secretory and membrane proteins, also showed a similar locali-
required for the segregation of these rough ER proteins into zation; GFP-tagged Dad1 that was stably expressed in Dad1-
sheets, and one of the coiled-coil proteins, Climp63, serves as deficient cells at a level just sufficient to sustain viability (Nikonov
a luminal ER spacer. However, neither the polysomes nor the et al., 2002) showed a clear preference for ER sheets, in contrast
coiled-coil proteins are essential for sheet formation per se. to calreticulin in the same cell (Figure 1E). Together, these data
Instead, a major mechanism of sheet formation appears to indicate that translocon components are enriched in ER sheets.
involve the reticulons and DP1/Yop1p proteins, which can To identify additional sheet-segregating proteins that could
stabilize the high membrane curvature at sheet edges. Our potentially be required for sheet formation, we reasoned that
results suggest that, in many cells, their abundance is the major such proteins would be abundant in highly secretory cells that
determinant of ER morphology. contain proliferated ER sheets. We therefore identified by mass
spectrometry the most abundant, integral ER membrane
RESULTS proteins in dog pancreatic rough microsomes. The 25 most
abundant proteins include translocon components, such as
Segregation of Proteins into ER Sheets subunits of the oligosaccharyl transferase complex, signal
The different morphologies of the ER imply that, despite the peptidase, SRP receptor, components of the TRAP complex,
continuity of the membrane system, some proteins are likely and the Sec61 complex (Table S1). Of interest, the list also
enriched in certain domains. So far, the only proteins known includes p180 and Climp63. Kinectin, which is sequence related
with a specific localization are the tubule-preferring reticulons, to p180, is somewhat less abundant. All of these proteins have
DP1/Yop1p, and atlastins/Sey1p (Hu et al., 2009; Shibata a single transmembrane segment and an extended coiled-coil
et al., 2008; Voeltz et al., 2006). These proteins localize to tubules domain, which is located on the luminal side of the ER membrane
even when highly overexpressed. By contrast, other overex- in the case of Climp63 and on the cytoplasmic side in the case of
pressed ER proteins distribute indiscriminately throughout the p180 and kinectin (Figure S2A). The molecular function of these
entire ER, making it impossible to draw conclusions about their coiled-coil proteins is not well understood. Climp63 has been
endogenous localizations. We therefore first tested whether implicated in the interaction of ER membranes with microtubules
several endogenous ER proteins segregate into different ER (Klopfenstein et al., 1998). P180 was originally proposed to be
domains using immunofluorescence and confocal microscopy a ribosome receptor (Savitz and Meyer, 1990); it also interacts
in BSC1 cells. As expected, the luminal ER protein calreticulin, with microtubules (Ogawa-Goto et al., 2007) and is now thought
which is involved in the folding of glycoproteins, was found in to play a role in the differentiation of certain monocytic cells (Be-
peripheral ER sheets, which are mostly located close to the nyamini et al., 2009). Kinectin was initially identified as a receptor
nucleus, as well as in the tubular ER network and the nuclear for the molecular motor kinesin (Toyoshima et al., 1992).
envelope (Figure 1A). Calreticulin almost perfectly colocalized Another way to identify potential sheet-segregating proteins is
with GFP-tagged Sec61b, stably overexpressed in the same to analyze components that are upregulated during the differen-
cell. Endogenous Sec61b is part of the Sec61 complex, the tiation of immature B cells to IgG-secreting plasma cells, which

Cell 143, 774–788, November 24, 2010 ª2010 Elsevier Inc. 775
Figure 1. Localization of Proteins to Different ER Domains
(A) The localization of endogenous luminal ER protein calreticulin is compared with that of the stably overexpressed membrane protein GFP-Sec61b using
confocal microscopy in BSC1 cells. Calreticulin was detected with specific antibodies by indirect immunofluorescence (left) and Sec61b by GFP fluorescence
(middle). The right panel shows a merged image. Scale bar, 10 mm.
(B) As in (A) but comparing the localization of the ER membrane protein calnexin with that of GFP-Sec61b.
(C) The localization of endogenous Sec61b is compared to that of the endogenous ER luminal proteins BiP and GRP94 (anti-KDEL), using indirect immunoflu-
orescence with specific antibodies and confocal microscopy.
(D) As in (A) but comparing the localization of the translocon membrane protein TRAPa with that of GFP-Sec61b. Also note that TRAPa is noticeably depleted from
the nuclear envelope.
(E) The localization of stably expressed GFP-Dad1 in a BHK cell line lacking endogenous Dad1 is compared with that of endogenous calreticulin.
All insets show a magnified view of the boxed areas highlighting the junctions between ER sheets and tubules. See also Figure S1.

involves massive ER sheet proliferation. To identify mRNAs 25 most upregulated mRNAs coding for ER membrane proteins
whose abundance is greatly increased, we sorted through (Table S2) includes components of the translocon, of the
published microarray data (Luckey et al., 2006). The list of the unfolded protein response, and of the ER protein degradation

776 Cell 143, 774–788, November 24, 2010 ª2010 Elsevier Inc.
machinery. It also includes Climp63 and p180 (their mRNAs are Climp63 Serves as a ‘‘Luminal ER Spacer’’
upregulated by a factor of 19–26; kinectin mRNA was not To test for a possible role of the coiled-coil membrane proteins in
analyzed). Together with the mass spectrometry data, these ER morphology, we performed RNAi experiments. The depletion
results raise the possibility that the coiled-coil membrane of Climp63, p180, and kinectin (Figure S5A) either individually or
proteins Climp63, p180, and kinectin localize to ER sheets. together did not abolish the existence of ER sheets (Figure 2D
Because these proteins have no known function in protein trans- versus 2E). Nevertheless, these proteins have an effect on ER
location or modification, they are also candidates for being morphology, as the sheets in depleted cells spread throughout
involved in sheet formation. the cytoplasm (Figures S5B and S5C), similarly to what is
Next, we tested whether the coiled-coil proteins are enriched observed when cells are treated with puromycin or pactamycin
in ER sheets, using immunofluorescence and confocal micros- (Figure S3). It thus seems that the coiled-coil membrane proteins
copy. At endogenous levels, all three proteins indeed segregated are not required for sheet formation per se may but function in
to ER sheets, whereas in the same cells, calreticulin distributed segregating sheet domains close to the cell nucleus.
throughout the entire ER (Figures 2A–2C). P180-GFP overex- Thin-section electron microscopy of COS7 cells confirmed
pressed at moderate levels was also enriched in ER sheets (Fig- that peripheral ER sheets persist after puromycin treatment or
ure S2B). Thus, in addition to the translocon proteins, at least depletion of Climp63, p180, and kinectin (compare Figures 4B
three other abundant integral membrane proteins are enriched and 4C with 4A). No bulging of the two membrane sheets was
in ER sheets. All three proteins were noticeably depleted from observed, but of interest, the luminal width was significantly
the nuclear envelope (Figure 2 and Figure S2), as reported previ- reduced in triple knockdown cells (from 45–50 nm to 25–30 nm;
ously for Climp63 (Klopfenstein et al., 1998). Figure 4E). A similar effect was seen when Climp63 alone was
depleted (Figures 4D and 4E), whereas single or double knock-
A Role for Polysomes in Protein Enrichment down of p180 and kinectin had no obvious phenotype (Figure 4E
in ER Sheets and data not shown). These results indicate that Climp63 serves
Because translocon-associated proteins were found enriched in to maintain the normal luminal width of peripheral ER sheets,
ER sheets and are also generally associated with ribosomes, we likely by forming bridges through their luminal coiled-coil
tested whether the sheet-preferring proteins are localized by domains (Klopfenstein et al., 2001). Consistent with a luminal
their association with membrane-bound translating ribosomes. spacer function, organisms that lack Climp63, including
We treated tissue culture cells with puromycin, a drug that Drosophila S2 cells (Figure 4E), silkworm (Senda and Yoshi-
releases nascent polypeptide chains from ribosomes and naga-Hirabayashi, 1998), and S. cerevisiae (Bernales et al.,
disassembles polysomes; the localization of endogenous 2006), all appear to have narrower ER sheets than mammals. It
sheet-preferring proteins was subsequently analyzed by immu- should be noted that the distance between the inner and outer
nofluorescence. TRAPa moved into the tubular network (Fig- nuclear membranes was unaffected by Climp63 depletion and
ure 3A). Quantification shows that, in untreated cells, TRAPa is was the same in mammalian and insect cells (Figure 4E), consis-
enriched in sheets, as compared to the general ER marker tent with the absence of this protein from the nuclear envelope.
GFP-Sec61b, but 15 min after puromycin addition, TRAPa was
almost equally abundant in sheets and tubules (Figure 3E). The Linking the Formation of ER Sheets and Tubules
disassembly of the polysomes did not abolish the ER sheets, The overexpression of Climp63 led to a dramatic proliferation of
which in fact occupied a larger surface in many cells (Figure S3A). ER sheets; we observed a good correlation between the expres-
To rule out the possibility that inhibition of translation causes the sion level of a FLAG-tagged version of Climp63 in COS7 cells
redistribution of TRAPa, we performed control experiments with and the generation of sheets, an effect that is most strikingly
cycloheximide, a drug that inhibits the elongation of polypeptide seen in three-dimensional (3D) reconstructions of the ER (Figures
chains but leaves polysomes intact. Cycloheximide inhibited 5A and 5B; quantification in 5C). In thin-section electron micros-
protein synthesis as effectively as puromycin (Figure S4), but copy, prominent membrane structures were seen that consisted
TRAPa stayed in ER sheets (Figures 3B and 3E). All of the other of anastomosing sheets containing membrane-bound ribo-
tested ER sheet-preferring proteins behaved in the same way as somes (Figure 5D). The sheets had a constant luminal width of
TRAPa (Figures 3C–3E). On the other hand, the localization of 50 nm, and at the highest expression levels, the luminal protein
calnexin and Bap31, membrane proteins that did not segregate calreticulin was displaced from areas of Climp63 localization
into ER sheets, remained unchanged after treatment with either (Figure S6), consistent with Climp63 filling the luminal space.
puromycin or cycloheximide, as was also the case for the luminal We also observed organized smooth ER (OSER) structures in
protein calreticulin (Figure 3E and Figures S3B and S3C). Pacta- which the membranes were tightly stacked and the internal
mycin, an inhibitor of translation initiation, which allows ribo- membranes were devoid of ribosomes (Figure S7). Although
somes to run off the mRNAs, had a similar effect as puromycin these structures are likely caused by oligomerization of the
on sheet-segregating proteins, i.e., they were no longer concen- cytoplasmic GFP tag (Snapp et al., 2003), they differ from normal
trated in sheets (Figure S3D). Again, the sheets did not disappear OSERs by having a constant luminal spacing of 50 nm.
but often occupied a larger area of the cell (Figure S3E). These Given that ER sheet proliferation was also observed when the
results indicate that polysomes concentrate sheet domains curvature-stabilizing reticulons are depleted in mammalian cells
and localize certain membrane proteins to ER sheets, likely by RNAi (Anderson and Hetzer, 2008) or when the reticulons and
because these proteins have a direct or indirect affinity for Yop1p are lacking in S. cerevisiae (Voeltz et al., 2006), we tested
membrane-bound polysomes. whether Climp63 and the reticulons have opposing effects on ER

Cell 143, 774–788, November 24, 2010 ª2010 Elsevier Inc. 777
Figure 2. Membrane Proteins Enriched in ER Sheets
(A) The endogenous localization of the membrane protein Climp63 is compared with that of the luminal ER protein calreticulin in COS7 cells, using indirect
immunofluorescence with specific antibodies. The far-right panel shows a merged image. Junctions between peripheral ER sheets and tubules are highlighted
in the magnified view of the boxed area (inset). Scale bar, 10 mm.
(B) As in (A) but comparing the localization of kinectin (KTN) and calreticulin.
(C) As in (A) but comparing the localization of p180 and calreticulin.
(D) Climp63, p180, and kinectin were depleted in COS7 cells by RNAi (C/P/K siRNA), and Climp63, TRAPa, and calreticulin were visualized using indirect
immunofluorescence with specific antibodies. Scale bar, 10 mm.
(E) As in (D) but with cells transfected with control siRNA oligonucleotides.
See also Figure S2 and Figure S5.

sheet formation. Indeed, when the reticulon Rtn4b was overex- tion in Figures 5G and 5H). Concomitant with the decrease in
pressed in COS7 cells, peripheral ER sheets became diminished sheet structures, the normal tubular network was gradually re-
with increasing expression levels (Figures 5E and 5F; quantifica- placed with long, unbranched tubules (quantification in Figure 5I).

778 Cell 143, 774–788, November 24, 2010 ª2010 Elsevier Inc.
When Climp63 and Rtn4a were both highly overexpressed, the A Model for the Generation of ER Sheets and Tubules
normal ER morphology was almost restored (Figure 5J). Taken To test whether the curvature-stabilizing proteins alone could
together, these results indicate that Climp63 and the curva- explain the relative amounts of sheets and tubules in a cell, we
ture-promoting proteins undergo a ‘‘tug-of-war’’ that determines developed a simple theoretical model. We assume that the
the amount of membrane partitioning into these domains. reticulons and DP1/Yop1p localize exclusively to tubules and
sheet edges, generating and stabilizing these high curvature
membranes by forming oligomeric scaffolds that are shaped
Curvature-Stabilizing Proteins Localize to Sheet Edges
as rigid arcs. Based on previous estimates, the energetically
Because the reticulons and DP1/Yop1p localize to tubules, one
optimal distance between the arcs is assumed to be 40 nm (Hu
might expect that they are also found at sheets edges because
et al., 2008). The edge membrane can be seen as a half-cylinder,
these have a similarly high membrane curvature as tubules in
whose axis bends in the sheet plane forming the sheet circumfer-
cross-section. Indeed, in many cells, the endogenous reticulons
ence. The protein-driven formation of a sheet edge enables the
localized to the edges of sheets, as demonstrated by immunoflu-
two membranes of a sheet to adopt planar shapes (Figure 7A).
orescence using antibodies recognizing both Rtn4a and 4b (Fig-
A tubule forms by self-folding of a part of the edge into
ure 6A). Similar observations were made in plant cells (Sparkes
a complete cylinder and therefore represents an edge extension
et al., 2010). In Climp63-overexpressing cells with proliferated
(Figure 7A). We assume negligible bulging between the arc-like
sheets, Rtn4a/b lined the edges of essentially all sheets in an
scaffolds, as supported by previous results (Hu et al., 2008),
even more striking manner (Figure 6B).
and a diameter of 30 nm for both sheet edges and tubules (Fig-
To test whether the curvature-stabilizing proteins generally
ure 4) (Bernales et al., 2006).
localize to sheet edges, we tested the localization of a reticulon
Our model calculates for a given membrane surface area the
in S. cerevisiae. We expressed Rtn1p-GFP from the chromo-
total length of the tubules and the shape and dimensions of the
some together with ssRFP-HDEL, a general, luminal ER marker.
sheets in dependence of the number of curvature-stabilizing
Indeed, peripheral ER sheets were generally lined by Rtn1p-GFP
proteins, Nc. We characterize the edge length by a parameter
(Figure 6C). The edge localization of Rtn1p-GFP was even more
G = Le/ Le0, wherein Le0 is the circumference of a flat circular
obvious in cells where ER sheet proliferation was induced by
disc with the same overall membrane area (i.e., G = 1 for a flat
deletion of the genes encoding the tubule-shaping protein
disc). G is proportional to Nc (Supplemental Information). In our
Yop1p and the GTPase Sey1p (Figure 6D) (Hu et al., 2009).
calculations, we assume that Nc is at least large enough to
Similar results were obtained when ER sheets were induced by
generate a circular sheet (G R 1).
deletion of OPI1 (Figure 6E) (Schuck et al., 2009). Thus, as in
For each G value, we computed the overall membrane shape
mammalian cells, the reticulons localize to the edges of periph-
by minimizing the energy of the edge bending in the sheet plane
eral ER sheets. These results indicate that the reticulons stabilize
(see Experimental Procedures and Supplemental Information).
the high curvature of both tubules in cross-section and of sheet
The top view of the shapes is presented in Figure 7B. Starting
edges.
from the circular disc configuration at G = 1 (Figure 7B, blue
line), the sheet shape elongates with increasing G (and Nc) (light
A Role for Curvature-Stabilizing Proteins blue line) and then acquires a flattened dumbbell appearance
in Sheet Formation with a narrowing neck (aqua and yellow lines) and, finally, at
Given the localization of the curvature-generating proteins to G2, splits into two droplet-like sheets with a short tubule
sheet edges, we considered the possibility that they can between them (orange line). Further increase of G results in
generate sheets by bringing the apposing membranes into close tubule elongation and a decrease in the sizes of the two sheets
proximity. In this model, the ratio of sheets and tubules would be (Figure 7B, dark red line). Eventually, the whole membrane
determined by the relative amounts of lipids and curvature- converts into a tubule (not shown in Figure 7B). Thus, the curva-
stabilizing proteins. Indeed, the sheet proliferation seen upon ture-producing proteins alone can generate both sheets, and
OPI1 deletion in S. cerevisiae (Figure 6E) is likely caused by an tubules and their abundance determines the relative amounts
increase in phospholipid synthesis; Opi1p normally inhibits the of the two membrane domains.
transcription factors Ino2p and Ino4p, which control many Next, we extended the model to include the effect of proteins
phospholipid synthesis enzymes (Ambroziak and Henry, 1994; enriched in sheets. We assume that polysome-bound Climp63,
Carman and Henry, 2007). To test whether expression of a curva- p180, and kinectin, as well as translocons, can diffuse
ture-stabilizing protein would convert the sheets into tubules, we throughout the sheets but cannot move into high curvature
used opi1D cells that express Rtn1p-GFP from the chromosome membrane areas, i.e., sheet edges and tubules. The number of
as well as the luminal ER marker ssRFP-HDEL. The overexpres- all of these ‘‘sheet proteins’’ together is denoted by Ns. The sheet
sion of untagged Rtn1p from a CEN plasmid led to a partial proteins may be considered as generating an ‘‘osmotic pres-
conversion of sheets into tubules (Figure 6F; quantification in sure’’ on the sheet edges, a force that resists the shrinkage of
Figure 6H). When untagged Rtn1p was expressed at a still higher a sheet domain. The magnitude of this effect is determined by
level from a 2 m plasmid, the sheet-to-tubule ratio converted the interplay between the effective ‘‘osmotic pressure’’
back to about the level seen in wild-type cells (Figures 6G produced by the sheet proteins and the effective stretching
and 6H). These data support the idea that the abundance of elasticity of the edge, the latter being determined by the curva-
the reticulons determines the relative amounts of sheets and ture-stabilizing proteins (see Experimental Procedures and
tubules in the cell. Supplemental Information). Our model does not take into

Cell 143, 774–788, November 24, 2010 ª2010 Elsevier Inc. 779
Figure 3. Polysome-Dependent Membrane Protein Enrichment in ER Sheets
(A) The localization of the translocon component TRAPa is compared with that of stably expressed GFP-Sec61b after 15 min of treatment with puromycin (PURO).
The far-right panel shows a merged image. Junctions between peripheral ER sheets and tubules are highlighted in the magnified view of the boxed area (inset).
Scale bar, 10 mm.
(B) As in (A) but after 15 min of treatment with cycloheximide (CHX).
(C) As in (A) but comparing the localization of Climp63 with calreticulin after puromycin treatment.
(D) As in (C) but after cycloheximide treatment.
(E) Quantification of sheet enrichment of different ER proteins in untreated cells (blue bars) and in cells treated with puromycin (PURO; green) or cycloheximide
(CHX; red). The ratio of the average fluorescence intensity in sheets versus tubules was determined for calnexin (CNX), Bap31, calreticulin (CRT), TRAPa, and
kinectin and was divided by the sheet-to-tubule fluorescence ratio for stably expressed GFP-Sec61b, a protein that shows no preference for either ER domain.

780 Cell 143, 774–788, November 24, 2010 ª2010 Elsevier Inc.
Figure 4. Climp63 Affects the Luminal Width of Peripheral ER Sheets
(A) Rough ER sheets in a COS7 cell visualized by thin-section electron microscopy. Scale bar, 0.5 mm.
(B) As in (A) but after treatment with puromycin (PURO) for 15 min.
(C) As in (A) but after RNAi-depletion of Climp63, p180, and kinectin (C/P/K siRNA).
(D) As in (A) but after RNAi-depletion of Climp63.
(E) Quantification of the luminal width of peripheral ER sheets and the nuclear envelope (NE) in differently treated COS7 cells. For comparison, Drosophila S2R+
cells were also analyzed. Shown are the means and standard errors of n cells analyzed for each sample. Kinectin, KTN.

account that Climp63 affects sheet formation by serving as a Ns at a constant Nc converts two small sheet areas connected
luminal spacer, and it does not make any assumptions about by a narrow tubule into a larger sheet area. Thus, the model reca-
the specific roles of p180 and kinectin. pitulates the experimental observation of a tug-of-war between
We computed the G values and membrane configurations for sheet-promoting Climp63 and curvature-stabilizing proteins.
different values of Nc and Ns (Figure 7C). The colored lines on the
bottom plane of the diagram represent the relationship between DISCUSSION
Nc and Ns for a given shape of the system, with the colors
corresponding to the shapes as in Figure 7B. Figure 7C demon- Our results indicate that several mechanisms shape peripheral
strates that an increase of Nc at a given Ns results in larger G ER sheets. The most basic and universal mechanism appears
(blue to red transition) and thus in more tubules, whereas an to involve the previously identified curvature-stabilizing proteins,
increase of Ns at a given Nc results in lower G, i.e., more sheets. the reticulons and DP1/Yop1p. These proteins would stabilize
This is further illustrated in Figure 7D, in which the increase of not only the high curvature of narrow tubules, but also the

A similar analysis was done for GFP-Dad1 and Climp63 but with calreticulin as reference. Shown are the means and standard errors of data obtained from 7 to
30 cells for each condition.
See also Figure S3 and Figure S4.

Cell 143, 774–788, November 24, 2010 ª2010 Elsevier Inc. 781
Figure 5. Climp63 and Reticulon Overexpression Change the Abundance of Sheets and Tubules
(A) FLAG-Climp63 overexpressed at relatively high levels in a COS7 cell was visualized by indirect immunofluorescence using FLAG antibodies. A 3D image was
generated from a complete series of z sections (step size 0.25 mm) taken with a confocal microscope. Scale bar, 10 mm.
(B) As in (A) but in a cell expressing FLAG-Climp63 at the highest observed levels.
(C) Quantification of the effect of Climp63 overexpression on ER sheet abundance. Shown are the percentages of cells with normal reticular ER (blue bars), of cells
with both large sheets and reticular ER (red), and of cells with large ER sheets lacking reticular ER (green) at different expression levels of FLAG-Climp63. The cells
were divided into five groups according to their expression levels, as determined by overall average fluorescence intensity.
(D) Thin-section electron micrograph of a COS7 cell overexpressing GFP-Climp63. The inset shows an enlargement of the boxed region. Scale bar, 0.5 mm.
(E) HA-Rtn4b (red) was expressed in COS7 cells at relatively low levels and was localized with HA antibodies by indirect immunofluorescence and confocal
microscopy. Endogenous Climp63 (green) was localized in the same cells with specific antibodies.
(F) As in (G) but with the highest observed expression level of HA-Rtn4b. Note that Climp63 appears in bright punctae and in the nuclear envelope.

782 Cell 143, 774–788, November 24, 2010 ª2010 Elsevier Inc.
curvature of sheet edges, a mechanism that is sufficient to keep optimize the size of the luminal space of peripheral ER sheets,
the two membranes of a sheet closely apposed. The reticulons such that sufficient luminal chaperones can be accommodated
and DP1/Yop1p probably stabilize high curvature by two mech- and the sheets are packed into a minimal space.
anisms, ‘‘hydrophobic insertion/wedging’’ and ‘‘scaffolding’’ Our analysis also identified two other coiled-coil membrane
(Shibata et al., 2009). The conserved segments of these proteins proteins, p180 and kinectin, with a potential role in shaping ER
may form a wedge in the lipid bilayer that occupies more space in sheets. These proteins are enriched in sheets and abundant
the cytoplasmic leaflet than in the luminal leaflet. Oligomerization in cells with proliferated ER sheets. Overexpression of p180
of these proteins may generate scaffolds around curved has been reported to induce sheets in S. cerevisiae and in a
membranes, which may take the shape of open arcs, given monocytic cell line (Becker et al., 1999; Benyamini et al., 2009),
that they can localize to sheet edges. Our theoretical model although in our own experiments and those of others, the effects
demonstrates that the reticulons and DP1/Yop1p alone can were smaller (Ueno et al., 2010 and data not shown). The deple-
generate both tubules and sheets, with their abundance deter- tion of p180 and kinectin had no effect on ER sheet morphology.
mining the ratio of these domains. Consistent with the proposed Although the precise role of these proteins remains to be estab-
dual role of the reticulons and DP1/Yop1p in tubule and sheet lished, all coiled-coil membrane proteins could stabilize sheets
formation, they localize to both tubules and sheet edges, their simply by being excluded from high-curvature regions, as shown
depletion leads to increased sheet areas, and their overexpres- by our theoretical considerations. They may be considered as
sion converts sheets into tubules. generating an ‘‘osmotic pressure,’’ a force that counteracts the
In S. cerevisiae, the amount of membrane surface and the shrinkage of sheet domains. Consistent with experimental
abundance of the reticulons and Yop1p appear to be the deci- observations for Climp63, the coiled-coil proteins are predicted
sive factors determining the ratio of peripheral ER sheets and to be in a tug-of-war with the reticulons and DP1/Yop1p, with
tubules. Generating more lipid increases the sheet area, whereas the former shifting the balance toward sheets and the latter
increasing the abundance of the curvature-stabilizing proteins toward tubules. In this model, it does not actually matter how
increases the number of tubules. The observation of sheets in proteins are excluded from tubules and sheet edges. Given
cells lacking the reticulons and Yop1p may be explained by that all identified sheet-promoting proteins contain extended
the presence of other low-abundance curvature-promoting coiled-coil domains, they all have the propensity to oligomerize,
proteins or by the association of the cortical ER with the plasma which may contribute to their exclusion from high-curvature
membrane. Although we cannot exclude the existence of sheet- regions.
promoting proteins in yeast, the current data are consistent with The coiled-coil membrane proteins are not essential for sheet
a model in which curvature-stabilizing proteins are the major formation per se, as is obvious from our observation that their
determinant of peripheral ER morphology. depletion by RNAi does not abolish ER sheets. This suggests
Our data suggest that, in mammalian cells, there are several that, like in yeast, the reticulons and DP1/Yop1p may provide
additional factors that determine the morphology of peripheral the basic mechanism by which both sheets and tubules are
ER sheets. This includes the coiled-coil membrane protein generated. Consistent with this hypothesis, Climp63, p180,
Climp63, which serves as a luminal spacer. After its depletion, and kinectin are not known in lower organisms, in contrast to
the luminal width of the sheets decreases from 50 to 30 nm, the reticulons and DP1/Yop1p, which are present in all
a spacing that is also seen in organisms that lack the protein. eukaryotes.
Climp63 is highly upregulated in mammalian cells with prolifer- All sheet-enriched proteins tested, including translocon
ated ER sheets, and it induces sheets at the expense of tubules components and the coiled-coil membrane proteins, appear
when overexpressed in tissue culture cells. Thus, at high to be concentrated by membrane-bound polysomes; upon
concentrations, Climp63 appears to generate sheets all by itself, polysome disassembly, all of these proteins distribute equally
and the lack of extensive sheet edges may make the contribution between sheets and tubules throughout the cell. Thus, these
of the curvature-stabilizing proteins less important. However, proteins must have a direct or indirect affinity for membrane-
with luminal spacers alone, one would expect bulging of the bound polysomes. Indeed, several of the tested sheet-prefer-
sheet edges, in contrast to our observations (Figure 4), indicating ring proteins are known to be associated with membrane-
that the curvature-stabilizing proteins may have a role even in bound translating ribosomes, including components of the
cells with proliferated ER sheets. Climp63’s function may be to Sec61 complex, the TRAP complex, the oligosaccharyl

(G) Quantification of the peripheral ER sheet area relative to the total ER area for different expression levels of HA-Rtn4b. The areas of ER sheets and tubules were
determined from the fluorescence of Climp63 and Rtn4b, respectively, after subtraction of background. The cells were divided into five groups according to their
expression levels of HA-Rtn4b, as determined by overall average fluorescence intensity, and the mean and standard error were calculated for each group.
(H) Quantification of the effect of Rtn4b overexpression on ER sheet morphology, as determined by Climp63 staining. Shown are the percentages of cells with
normal ER sheets (blue bars), of cells with disc-like ER sheets (red), and of cells with punctae (green) at different expression levels of Rtn4b. The cells were divided
into five groups according to their expression levels.
(I) Quantification of the effect of Rtn4b overexpression on ER tubule morphology, as determined by HA-Rtn4b staining. Shown are the percentages of cells with
normal reticular ER (blue bars), of cells with an abnormally dense ER network (red), and of cells with unbranched, long tubules (green) at different expression levels
of Rtn4b. The cells were divided into five groups according to their expression levels.
(J) Myc-Rtn4a and FLAG-Climp63 were both highly expressed in COS7 cells. The far-right panel shows a merged image. Note that the ER morphology is
almost normal.
See also Figure S6 and Figure S7.

Cell 143, 774–788, November 24, 2010 ª2010 Elsevier Inc. 783
Figure 6. The Reticulons Localize to the Edges of ER Sheets
(A) The localization of endogenous Rtn4a and 4b is compared with that of Climp63 using indirect immunofluorescence with specific antibodies in COS7 cells. The
insets show enlargements of the boxed region. Arrows point to reticulons lining the sheets. The far-right panel shows merged images. Scale bar, 10 mm.
(B) As in (A) but with cells overexpressing FLAG-Climp63.
(C) Rtn1p-GFP (green) and ssRFP-HDEL (red) were coexpressed in wild-type S. cerevisiae cells, and the cortical ER was visualized by fluorescence microscopy.
Scale bar, 5 mm.
(D) As in (C) except that the cells had proliferated ER sheets caused by deletion of SEY1 and YOP1 (sey1Dyop1D).
(E) As in (C) except the cells had proliferated ER sheets caused by deletion of OPI1 (opi1D). The cells also contained an empty vector as a control for panels (F) and (G).
(F) As in (E) except that untagged Rtn1p was expressed under the endogenous promoter from a CEN plasmid.
(G) As in (E) except that untagged Rtn1p was expressed under the endogenous promoter from a 2 m plasmid.
(H) Quantification of the experiments in (C) and (E–G). The relative area of ER sheets was determined from the area of ssRFP-HDEL fluorescence that did not coloc-
alize with Rtn1p-GFP fluorescence and was divided by the total area of ssRFP-HDEL fluorescence. 14 to 38 cells were analyzed per condition, and the means and
standard errors were calculated.

784 Cell 143, 774–788, November 24, 2010 ª2010 Elsevier Inc.
Figure 7. Modeling of the Effect of Curvature-Stabilizing and Sheet-Promoting Proteins on ER Morphology
(A) The reticulons and DP1/Yop1p (yellow arcs) are assumed to localize exclusively to tubules and sheet edges, generating and stabilizing these high-curvature
membranes. Stabilization of sheet edges enables the upper and lower membranes of the sheet to adopt planar shapes.
(B) Top view of membrane shapes computed by the theoretical model for increasing G values. The computation was performed for a total membrane area cor-
responding to 1 mm radius of the initial disc-like shape, a 15 nm cross-section radius of the tubules and edges, and a 40 nm optimal distance between the arc-like
proteins at the edge (see Supplemental Information). Change of G from 1 to 2.1(blue to red) corresponds to increasing the number of curvature-stabilizing proteins
Nc from 140 to 290.
(C) G values and membrane shapes were calculated for different numbers of curvature-stabilizing and sheet-promoting proteins, Nc and Ns. The colors corre-
spond to the membrane shapes shown in Figure 7B. The colored lines on the bottom plane of the diagram represent the relationship between Nc and Ns for a given
shape of the system.
(D) G values and membrane shapes were computed for different Ns values at Nc = 290. The shapes refer to Ns = 0, 500, and 1000.

transferase complex, and p180 (Görlich and Rapoport, 1993). Our results indicate that ER sheets correspond to rough
These proteins stay bound to ribosomes upon detergent solu- ER and tubules to smooth ER. We propose that the assembly
bilization of rough ER membranes, but they can be released of translating membrane-bound ribosomes into polysomes
from the ribosomes by puromycin/high salt treatment. Climp63 concentrates the associated membrane-proteins, including
and kinectin are not bound to detergent-solubilized translocons Climp63, p180, and kinectin. Their concentration might facilitate
(data not shown), so how they are recruited remains to be their higher-order oligomerization, which may be required for
clarified. their exclusion from high-curvature areas and thus for their

Cell 143, 774–788, November 24, 2010 ª2010 Elsevier Inc. 785
sheet-promoting function. Once sheets are formed, the how these factors collaborate with the identified membrane-
membrane binding of polysomes would be facilitated. Poly- shaping principles.
somes often form spirals that could have an inherent preference
for associating with ER sheets (Christensen and Bourne, 1999); EXPERIMENTAL PROCEDURES
whereas individual ribosomes or small polysomes can bind
Mammalian Tissue Culture and Transfections
to narrow tubules, it is unlikely that each ribosome of a large
BSC1 cells stably expressing GFP-Sec61b and COS7 cells were grown in
polysome could be efficiently arranged on a narrow tubule. The DMEM containing 10% fetal bovine serum at 37 C and 5% CO2 and were
binding of large polysomes could therefore be restricted to passaged every 2–3 days. GFP-Dad1 BHK cells (M3/18; Nikonov et al., 2002)
membrane sheets. The assembly of membrane-bound poly- were maintained in 10% CO2 at 39.5 C to degrade endogenous Dad1. For trans-
somes would concentrate more coiled-coil membrane proteins, lation inhibition experiments, cells were treated with 200 mM cycloheximide,
and these in turn would generate more sheet area by the 200 mM puromycin, or 100 nM pactamycin in complete media for 15 min.
To deplete Climp63, kinectin, and p180, COS7 cells were plated onto
‘‘osmotic effect,’’ allowing more polysomes to bind, and so on,
acid-washed coverslips at 20% confluency and were transfected with
a mechanism that would ultimately lead to a segregated rough 120 nM total siRNA using Oligofectamine (Invitrogen). After1.5 days, cells
ER domain. This model is consistent with the observation that were retransfected with the same amount of siRNA oligonucleotides and
the disassembly of polysomes or the depletion of Climp63 then processed for immunofluorescence 1.5 days afterward. Experiments
increases the mobility of translocons in the plane of the with control siRNA oligonucleotides (QIAGEN) were done in parallel using
membrane (Nikonov et al., 2007; Nikonov et al., 2002). It also the same conditions. Transient DNA transfections were performed using
Lipofectamine 2000 (Invitrogen). See Supplemental Information for a list of
agrees with our results showing that the disassembly of poly-
DNA and siRNA constructs.
somes leads to the spreading of ER sheets similar to that
seen upon depletion of the sheet-promoting proteins. Our model Indirect Immunofluorescence and Confocal Microscopy
explains the classic observation that, in many cells, membrane- Indirect immunofluorescence with mammalian cells was done as described
bound ribosomes are not randomly distributed throughout the (Shibata et al., 2008). Cells grown on acid-washed coverslips were fixed
ER but, rather, concentrated in a separate membrane domain, with 4% paraformaldehyde (EMS), permeabilized with 0.1% Triton X-100
(Pierce), and immunostained with various primary antibodies and then washed
the rough ER. An active sorting of proteins into the rough ER is
in PBS and probed with various fluorophore-conjugated secondary anti-
consistent with previous cell fractionation experiments, which bodies. See Supplemental Information for a list of antibodies used.
demonstrated that general ER proteins indiscriminately Images were captured using a Yokogawa spinning-disk confocal on a Nikon
distribute throughout the ER, whereas translocon-associated TE2000U inverted microscope with a 603 or 1003 Plan Apo NA 1.4 objective
proteins are enriched in the rough ER (Hinman and Phillips, lens, a Hamamatsu ORCA ER-cooled CCD camera, and MetaMorph software.
1970; Kreibich et al., 1978; Vogel et al., 1990). All analyses/quantifications were done on raw 16 bit images using MetaMorph.
For presentation, brightness levels were adjusted across the entire image and
The nuclear envelope is a prominent ER domain whose struc-
were changed from 16 to 8 bits using Adobe Photoshop. Quantification was
ture is determined independently of the peripheral ER. Although
performed as described in Supplemental Information.
the reticulons have been implicated in the assembly of the
nuclear envelope and in the insertion of nuclear pores (Anderson Thin-Section Electron Microscopy
and Hetzer, 2008; Dawson et al., 2009), they are nearly absent Thin-section EM experiments were performed as described previously
from the nuclear envelope, and their depletion or overexpression (Shibata et al., 2008) except that cells were fixed directly in culture plates.
Quantification was performed as described in Supplemental Information.
has no significant effect on this domain’s morphology. Similarly,
DP1/Yop1p or the coiled-coil membrane proteins Climp63, Microscopy and Image Quantification of S. cerevisiae Cells
p180, and kinectin are also nearly absent from the nuclear enve- Yeast strains and constructs used are described in Supplemental Information.
lope and have no obvious effect on its structure. Of interest, Yeast cells were imaged live in complete medium at room temperature using
TRAPa was also depleted from the nuclear envelope, raising an Olympus BX61 microscope, UPlanApo 100 3 /1.35 lens, Qimaging Retiga
the possibility that translocons are preferentially located in EX camera, and IVision version 4.0.5 software. To calculate relative peripheral
sheet amounts, cortical ER images of cells expressing ssRFP-HDEL and
peripheral ER sheets. Thus, distinct mechanisms may determine
Rtn1-GFP were taken. Images were thresholded above background, and the
the formation and function of the sheet-like domains of the percentage of sheet area was calculated for each cell as the percentage of
nuclear envelope and peripheral ER. area of ssRFP-HDEL that did not overlap with Rtn1-GFP using Metamorph
In summary, our results lead to a simple model, according to software. Means and standard errors were calculated using Microsoft Excel.
which the basic morphological elements of the peripheral ER, For presentation, brightness levels were adjusted across the entire image,
the tubules and sheets, are generated by the curvature- changed from 16 to 8 bits, and cropped using Adobe Photoshop.
stabilizing proteins. Superimposed on this mechanism, mem-
Identification of Abundant Coiled-Coil Membrane Proteins
brane-bound polysomes and associated coiled-coil membrane Mass spectrometry of dog pancreatic microsomal proteins and identification
proteins may cooperate to form segregated rough ER sheets in of mRNAs coding for ER membrane proteins that are upregulated during
mammalian cells, domains that are functionally specialized in B cell differentiation (Luckey et al., 2006) were performed as described in
protein translocation. Other factors probably contribute to the the Supplemental Information.
morphology of the peripheral ER. Microtubules keep the
mammalian ER under tension and stabilize membrane tubules, Modeling of Sheet versus Tubule Generation
To compute the membrane configurations (the length of the tubule as well as
but they could also potentially form an additional scaffold that
the areas and shapes of the sheets) in dependence of the numbers of the
stabilizes sheets, as suggested by the fact that both Climp63 curvature-producing, Nc, and the sheet-promoting proteins, Ns, we minimize
and p180 are microtubule-binding proteins (Klopfenstein et al., the system energy, Ftot, for the given total membrane area, Atot. The total
1998; Ogawa-Goto et al., 2007). It will be interesting to elucidate energy Ftot consists of three contributions: the effective stretching energy of

786 Cell 143, 774–788, November 24, 2010 ª2010 Elsevier Inc.
the edge, Fs; the energy of the effective osmotic pressure of the sheet- Anderson, D.J., and Hetzer, M.W. (2008). Reshaping of the endoplasmic
promoting proteins, Fp; and the energy of edge bending in the sheet plane, Fb. reticulum limits the rate for nuclear envelope formation. J. Cell Biol. 182,
h i2 911–924.
The energy Fs is given by Fs = 12kB TNc ðLe NNccðll00 + la ÞÞ , wherein, Le is the total
Baumann, O., and Walz, B. (2001). Endoplasmic reticulum of animal cells and
length of the edge including the tubules; l0 is the energetically preferred
its organization into structural and functional domains. Int. Rev. Cytol. 205,
distance between the arc-like proteins measured along the edge; la is the width
149–214.
of one protein arc; and kB Tz4$1021 Joule is the product of the Boltzmann
constant and the absolute temperature. According to this expression, the Becker, F., Block-Alper, L., Nakamura, G., Harada, J., Wittrup, K.D., and
length of the edge in a stress-free state is Le = Nc ðl0 + la Þ, and the effective Meyer, D.I. (1999). Expression of the 180-kD ribosome receptor induces
rigidity of the edge stretching-compression with respect to Le is membrane proliferation and increased secretory activity in yeast. J. Cell Biol.
kstr = kB T$Nc . Based on previous estimates, we take l0 = 40nm and la = 4nm 146, 273–284.
(Hu et al., 2008). Benyamini, P., Webster, P., and Meyer, D.I. (2009). Knockdown of p180
The osmotic pressure energy Fp is given by Fp = kB T$Ns $lnðNs b=Aflat Þ, eliminates the terminal differentiation of a secretory cell line. Mol. Biol. Cell
wherein Aflat is the flat area available to the sheet proteins and b is the area 20, 732–744.
of one sheet protein. The area Aflat is related to the total area and length of Bernales, S., McDonald, K.L., and Walter, P. (2006). Autophagy counterbal-
the edge by Aflat = 12ðAtot  a$Le Þ, wherein a is the membrane area absorbed ances endoplasmic reticulum expansion during the unfolded protein
by a unit length of the edge, which can be estimated as a = p$Re z50nm response. PLoS Biol. 4, e423.
(Re z15nm is the radius of the edge cross-section), and Atot is the total
Carman, G.M., and Henry, S.A. (2007). Phosphatidic acid plays a central role in
membrane area.
the transcriptional regulation of glycerophospholipid synthesis in Saccharo-
The energy Fb is given by Fb = 12B#c2e dLe , wherein ce is the in-plane curvature
myces cerevisiae. J. Biol. Chem. 282, 37293–37297.
of the edge, and B is the modulus of the edge in-plane bending, which can be
estimated using the membrane bending modulus kz20kB T (Helfrich, 1973) as Christensen, A.K., and Bourne, C.M. (1999). Shape of large bound polysomes
BzpRe kz900kB T$nm. The integration is performed over the whole edge in cultured fibroblasts and thyroid epithelial cells. Anat. Rec. 255, 116–129.
length, including the tubules. Dawson, T.R., Lazarus, M.D., Hetzer, M.W., and Wente, S.R. (2009).
Estimates supported by numerical computations show that the total length ER membrane-bending proteins are necessary for de novo nuclear pore
of the edge Le and the corresponding value of the parameter G are determined formation. J. Cell Biol. 184, 659–675.
by the energies Fs and Fp and are largely independent of Fb. At the same time, Fawcett, D.W. (1981). The Cell (Philadelphia: W.B. Saunders).
the system configuration resulting from minimization of Fb depends of the
Görlich, D., and Rapoport, T.A. (1993). Protein translocation into proteolipo-
parameter G. Therefore, we determine the system configuration in two steps.
somes reconstituted from purified components of the endoplasmic reticulum
First, we minimize the sum of Fc + Fs with respect to Le for every set of
membrane. Cell 75, 615–630.
numbers Nc and Ns and determine the corresponding function G (Nc, Ns).
Second, for every value of G (Nc, Ns), we minimize Fb with respect to the system Helfrich, W. (1973). Elastic properties of lipid bilayers: theory and possible
shape and find the equilibrium configuration. experiments. Z. Naturforsch. C 28, 693–703.
The Supplemental Information gives a more detailed discussion of the Hinman, N.D., and Phillips, A.H. (1970). Similarity and limited multiplicity of
model. membrane proteins from rough and smooth endoplasmic reticulum. Science
170, 1222–1223.
SUPPLEMENTAL INFORMATION Hu, J., Shibata, Y., Voss, C., Shemesh, T., Li, Z., Coughlin, M., Kozlov, M.M.,
Rapoport, T.A., and Prinz, W.A. (2008). Membrane proteins of the endoplasmic
Supplemental Information includes Extended Experimental Procedures, reticulum induce high-curvature tubules. Science 319, 1247–1250.
seven figures, and two tables and can be found with this article online at Hu, J., Shibata, Y., Zhu, P.P., Voss, C., Rismanchi, N., Prinz, W.A., Rapoport,
doi:10.1016/j.cell.2010.11.007. T.A., and Blackstone, C. (2009). A class of dynamin-like GTPases involved in
the generation of the tubular ER network. Cell 138, 549–561.
ACKNOWLEDGMENTS
Klopfenstein, D.R., Kappeler, F., and Hauri, H.P. (1998). A novel direct interac-
tion of endoplasmic reticulum with microtubules. EMBO J. 17, 6168–6177.
We thank C. Denison, J. Minsteris, and S. Gygi for mass spectrometry analysis;
J. Baughman for microarray analysis; A. Condon and A. Boye-Doe for Klopfenstein, D.R., Klumperman, J., Lustig, A., Kammerer, R.A., Oorschot, V.,
technical assistance; J. Iwasa for help with illustrations; G. Kreibich, and Hauri, H.P. (2001). Subdomain-specific localization of CLIMP-63 (p63) in
K. Ogawa-Goto, L. Lu, and R. Yan for materials; the Nikon Imaging Center the endoplasmic reticulum is mediated by its luminal a-helical segment.
and the Electron Microscopy facility at HMS for microscopy assistance; and J. Cell Biol. 153, 1287–1300.
R. Klemm and A. Osborne for critical reading of the manuscript. Y.S. was sup- Kreibich, G., Ulrich, B.L., and Sabatini, D.D. (1978). Proteins of rough micro-
ported by the American Heart Association and is a Howard Hughes Medical somal membranes related to ribosome binding. I. Identification of ribophorins
Institute postdoctoral fellow. W.A.P. is supported by the Intramural Research I and II, membrane proteins characteristics of rough microsomes. J. Cell Biol.
Program of the National Institute of Diabetes and Digestive and Kidney 77, 464–487.
Diseases. T.A.R. is a Howard Hughes Medical Institute Investigator. M.M.K. Luckey, C.J., Bhattacharya, D., Goldrath, A.W., Weissman, I.L., Benoist, C.,
is supported by the Israel Science Foundation (ISF) and the Marie Curie and Mathis, D. (2006). Memory T and memory B cells share a transcriptional
network ‘‘Virus Entry.’’ program of self-renewal with long-term hematopoietic stem cells. Proc. Natl.
Acad. Sci. USA 103, 3304–3309.
Received: May 19, 2010
Ménétret, J.F., Hegde, R.S., Aguiar, M., Gygi, S.P., Park, E., Rapoport, T.A.,
Revised: September 3, 2010
and Akey, C.W. (2008). Single copies of Sec61 and TRAP associate with a non-
Accepted: October 26, 2010
translating mammalian ribosome. Structure 16, 1126–1137.
Published: November 24, 2010
Nikonov, A.V., Hauri, H.P., Lauring, B., and Kreibich, G. (2007). Climp-63-
REFERENCES mediated binding of microtubules to the ER affects the lateral mobility of
translocon complexes. J. Cell Sci. 120, 2248–2258.
Ambroziak, J., and Henry, S.A. (1994). INO2 and INO4 gene products, positive Nikonov, A.V., Snapp, E., Lippincott-Schwartz, J., and Kreibich, G. (2002).
regulators of phospholipid biosynthesis in Saccharomyces cerevisiae, form Active translocon complexes labeled with GFP-Dad1 diffuse slowly as large
a complex that binds to the INO1 promoter. J. Biol. Chem. 269, 15344–15349. polysome arrays in the endoplasmic reticulum. J. Cell Biol. 158, 497–506.

Cell 143, 774–788, November 24, 2010 ª2010 Elsevier Inc. 787
Ogawa-Goto, K., Tanaka, K., Ueno, T., Tanaka, K., Kurata, T., Sata, T., and Irie, Short, B., Haas, A., and Barr, F.A. (2005). Golgins and GTPases, giving identity
S. (2007). p180 is involved in the interaction between the endoplasmic retic- and structure to the Golgi apparatus. Biochim. Biophys. Acta 1744, 383–395.
ulum and microtubules through a novel microtubule-binding and bundling Snapp, E.L., Hegde, R.S., Francolini, M., Lombardo, F., Colombo, S., Pedraz-
domain. Mol. Biol. Cell 18, 3741–3751. zini, E., Borgese, N., and Lippincott-Schwartz, J. (2003). Formation of stacked
Puhka, M., Vihinen, H., Joensuu, M., and Jokitalo, E. (2007). Endoplasmic ER cisternae by low affinity protein interactions. J. Cell Biol. 163, 257–269.
reticulum remains continuous and undergoes sheet-to-tubule transformation Sparkes, I., Tolley, N., Aller, I., Svozil, J., Osterrieder, A., Botchway, S., Mueller,
during cell division in mammalian cells. J. Cell Biol. 179, 895–909. C., Frigerio, L., and Hawes, C. (2010). Five Arabidopsis reticulon isoforms
Savitz, A.J., and Meyer, D.I. (1990). Identification of a ribosome receptor in the share endoplasmic reticulum location, topology, and membrane-shaping
rough endoplasmic reticulum. Nature 346, 540–544. properties. Plant Cell 22, 1333–1343.
Schuck, S., Prinz, W.A., Thorn, K.S., Voss, C., and Walter, P. (2009). Toyoshima, I., Yu, H., Steuer, E.R., and Sheetz, M.P. (1992). Kinectin, a major
Membrane expansion alleviates endoplasmic reticulum stress independently kinesin-binding protein on ER. J. Cell Biol. 118, 1121–1131.
of the unfolded protein response. J. Cell Biol. 187, 525–536. Ueno, T., Tanaka, K., Kaneko, K., Taga, Y., Sata, T., Irie, S., Hattori, S., and
Senda, T., and Yoshinaga-Hirabayashi, T. (1998). Intermembrane bridges Ogawa-Goto, K. (2010). Enhancement of procollagen biosynthesis by p180
within membrane organelles revealed by quick-freeze deep-etch electron through augmented ribosome association on the endoplasmic reticulum in
microscopy. Anat. Rec. 251, 339–345. response to stimulated secretion. J. Biol. Chem. 285, 29941–29950.
Shibata, Y., Hu, J., Kozlov, M.M., and Rapoport, T.A. (2009). Mechanisms Voeltz, G.K., Prinz, W.A., Shibata, Y., Rist, J.M., and Rapoport, T.A. (2006).
shaping the membranes of cellular organelles. Annu. Rev. Cell Dev. Biol. 25, A class of membrane proteins shaping the tubular endoplasmic reticulum.
329–354. Cell 124, 573–586.
Shibata, Y., Voeltz, G.K., and Rapoport, T.A. (2006). Rough sheets and smooth Voeltz, G.K., Rolls, M.M., and Rapoport, T.A. (2002). Structural organization of
tubules. Cell 126, 435–439. the endoplasmic reticulum. EMBO Rep. 3, 944–950.
Shibata, Y., Voss, C., Rist, J.M., Hu, J., Rapoport, T.A., Prinz, W.A., and Voeltz, Vogel, F., Hartmann, E., Görlich, D., and Rapoport, T.A. (1990). Segregation of
G.K. (2008). The reticulon and DP1/Yop1p proteins form immobile oligomers in the signal sequence receptor protein in the rough endoplasmic reticulum
the tubular endoplasmic reticulum. J. Biol. Chem. 283, 18892–18904. membrane. Eur. J. Cell Biol. 53, 197–202.

788 Cell 143, 774–788, November 24, 2010 ª2010 Elsevier Inc.
Abortive HIV Infection Mediates
CD4 T Cell Depletion and Inflammation
in Human Lymphoid Tissue
Gilad Doitsh,1 Marielle Cavrois,1,5 Kara G. Lassen,1,5 Orlando Zepeda,1 Zhiyuan Yang,1 Mario L. Santiago,1,4
Andrew M. Hebbeler,1 and Warner C. Greene1,2,3,*
1Gladstone Institute of Virology and Immunology, 1650 Owens Street, San Francisco, CA 94158, USA
2Department of Medicine
3Department of Microbiology and Immunology

University of California, San Francisco, CA 94143, USA


4Present address: University of Colorado, Denver, Aurora, CO 80045, USA
5These authors contributed equally to this work

*Correspondence: wgreene@gladstone.ucsf.edu
DOI 10.1016/j.cell.2010.11.001

SUMMARY Various mechanisms have been proposed to contribute to


the death of these bystander CD4 T cells including the action
The mechanism by which CD4 T cells are depleted of host-derived factors like tumor necrosis factor-a, Fas ligand
in HIV-infected hosts remains poorly understood. and TRAIL (Gandhi et al., 1998; Herbeuval et al., 2005), and viral
In ex vivo cultures of human tonsil tissue, CD4 factors like HIV-1 Tat, Vpr, and Nef released from infected cells
T cells undergo a pronounced cytopathic response (Schindler et al., 2006; Westendorp et al., 1995). Considerable
following HIV infection. Strikingly, >95% of these interest has also focused on the role of gp120 and gp41 Env
protein in indirect cell death, although it is not clear whether
dying cells are not productively infected but instead
death signaling involves gp120 binding to its chemokine
correspond to bystander cells. We now show that
receptor or gp41-mediated fusion. It is also unclear whether
the death of these ‘‘bystander’’ cells involves abor- such killing is caused by HIV-1 virions or by infected cells
tive HIV infection. Inhibitors blocking HIV entry or expressing Env.
early steps of reverse transcription prevent CD4 Most studies have focused on death mechanisms acting prior
T cell death while inhibition of later events in the viral to viral entry. Less is known about the fate of HIV-1-infected CD4
life cycle does not. We demonstrate that the nonper- T cells that do not express viral genes, in particular naive CD4
missive state exhibited by the majority of resting CD4 T cells in tissue that are refractory to productive HIV infection
tonsil T cells leads to accumulation of incomplete (Glushakova et al., 1995; Kreisberg et al., 2006). In these cells,
reverse transcripts. These cytoplasmic nucleic acids infection is aborted after viral entry, as reverse transcription is
activate a host defense program that elicits a coordi- initiated but fails to reach completion (Kamata et al., 2009;
Swiggard et al., 2004; Zack et al., 1990; Zhou et al., 2005).
nated proapoptotic and proinflammatory response
Human lymphoid aggregated cultures (HLACs) prepared from
involving caspase-3 and caspase-1 activation. While
tonsillar tissue closely replicate the conditions encountered by
this response likely evolved to protect the host, it HIV in vivo and thus form an attractive, biologically relevant
centrally contributes to the immunopathogenic system for studying HIV-1 infection (Eckstein et al., 2001).
effects of HIV. Lymphoid organs are the primary sites of HIV replication and
contain more than 98% of the body’s CD4 T cells. Moreover,
INTRODUCTION events critical to HIV disease progression occur in lymphoid
tissues, where the network of cell-cell interactions mediating
Despite extensive efforts over the past quarter century, the the immune response deteriorates and ultimately collapses.
precise mechanism by which HIV-1 causes progressive deple- Primary cultures of peripheral blood cells do not fully mimic
tion of CD4 T cells remains debated. Both direct and indirect the cytokine milieu, the cellular composition of lymphoid tissue,
cytopathic effects have been proposed. When immortalized nor the functional relationships that are undoubtedly important
T cell lines are infected with laboratory-adapted HIV-1 strains, in HIV pathogenesis. Finally, HLACs can be infected with
direct CD4 T cell killing predominates. Conversely, in more a low number of viral particles in the absence of artificial mito-
physiological systems, such as infection of lymphoid tissue gens, allowing analysis of HIV cytopathicity in a natural and
with primary HIV-1 isolates, the majority of dying cells appear preserved environment. In this study, we used the HLAC system
as uninfected ‘‘bystander’’ CD4 T cells (Finkel et al., 1995; Jekle to explore the molecular basis for HIV-induced killing of CD4
et al., 2003). T cells.

Cell 143, 789–801, November 24, 2010 ª2010 Elsevier Inc. 789
Figure 1. Massive Depletion of CD4 T Cells in HLACs Containing Small Number of Productively Infected Cells
Kinetics of spreading viral infection versus depletion of CD4 T cells after infection of HLACs with a replication-competent HIV reporter virus encoding GFP.
CD4 downregulation in GFP-positive cells likely represents the combined action of the HIV Nef, Vpu, and Env proteins expressed by this virus. Ratios of viable
CD4 versus CD8 T cells in HIV-infected and uninfected cultures are also shown. Flow cytometry plots represent live-gated cells, based on the forward-scatter
versus side-scatter profile of the complete culture. These data are the representative results of six independent experiments utilizing tonsil cells from six different
donors.

RESULTS was selected because tonsillar tissue contains a high per-


centage of CD4 T cells expressing CXCR4 (90%–100%).
Selective Depletion of CD4 T Cells by X4-Tropic HIV-1 Productively infected GFP-positive cells appeared in small
To explore depletion of CD4 T cells by HIV-1, HLACs made from numbers 3 days after infection, peaked on days 6–9, and
freshly dissected human tonsillar tissues were infected with decreased until day 12, when few CD4 T cells remained in the
a GFP reporter virus (NLENG1), prepared from the X4-tropic culture (Figure 1). Fluorescence-linked antigen quantification
NL4-3 strain of HIV-1. This reporter produces fully replication- (FLAQ) assay of HIV-1 p24 (Hayden et al., 2003) confirmed the
competent viruses. An IRES inserted upstream of the Nef gene accumulation of viral particles in the medium between day 3
preserves Nef expression and supports LTR-driven GFP expres- and days 8 to 9, when a plateau was reached (data not shown).
sion (Levy et al., 2004), allowing simultaneous quantification of Interestingly, when HIV-1 p24 levels plateaued no more than
the dynamics of HIV-1 infection and T cell depletion. NL4-3 1.5% of all cells (about 5% of CD4 T cells) were GFP-positive.

790 Cell 143, 789–801, November 24, 2010 ª2010 Elsevier Inc.
Figure 2. CD4 T Cell Depletion in HIV-1-Infected HLACs Predominantly Involves Nonproductively Infected Cells
(A) Experimental strategy to assess indirect cell killing in HIV-1-infected human lymphoid cultures. Fresh human tonsil tissue from a single donor is processed into
HLAC, and then separated into two fractions. One fraction is challenged with HIV-1 and cultured for 6 days, allowing viral spread. On day 5, the uninfected fraction
is treated with AZT (5 mM) and labeled with CFSE (1 mM). On day 6, the infected and CFSE-labeled cultures are mixed and cocultured in the presence of AZT.
Because of its site of action, AZT does not block viral output from the HIV-infected cells but prevents productive infection of CFSE-labeled cells. After 6 days
of coculturing, the number of viable CFSE-positive cells is determined by flow cytometry.
(B) Flow cytometry analysis of the mixed HLACs. Indirect killing is determined by gating on live CFSE-positive cells in the mixed cultures. Effector cells are either
infected or uninfected cells.
(C) Extensive depletion of nonproductively infected CD4 T cells by HIV-1. CFSE-labeled cells mixed with uninfected or infected cells were cultured in the presence
of 5 mM AZT alone or together with 250 nM AMD3100. Data represent live CFSE-positive cells 6 days after coculture with infected or uninfected effector cells. The
absence of productive infection in the CFSE-positive cells was confirmed by internal p24 staining and monitoring GFP expression following infection with the
NLENG1 HIV-1 reporter virus (data not shown).
(D) Preferential depletion of nonproductively infected CD4 T cells by HIV-1. The absolute numbers of viable CFSE-positive CD4 and CD8 T cells and B cells were
determined. Percentages are normalized to the number of viable CFSE-positive cells cocultured with uninfected cells in the presence of AZT, as depicted by an
asterisk. Error bars represent standard deviations of three samples from the same donor. This experiment is representative of more than 10 independent
experiments with more than 10 donors of tonsillar tissues.
See also Figure S1.

However, although the number of CD4 T cells was not markedly Extensive Depletion of Nonproductively Infected CD4 T
altered in infected cultures through six days, the culture was Cells in HLACs
almost completely devoid of CD4 T cells by day 9. CD8 T cells To determine if indirect killing (formerly indicated as ‘‘bystander’’)
were not depleted in infected cultures, and CD4 T cells were of CD4 T cells accounted for most of the observed cellular
not depleted in uninfected cultures. These findings reveal depletion, we took advantage of a reported experimental
marked and selective depletion of CD4 T cells in HLAC cultures. strategy (Jekle et al., 2003) that unambiguously distinguishes
However, due to the nature of the assay, we could not definitively between the death of productively and nonproductively infected
conclude whether the principal mechanism of depletion involved cells (Figure 2A). After 6 days of coculture, survival analysis
direct or indirect effects of HIV-1. of CFSE-labeled cells by flow cytometry (Figure 2B) showed

Cell 143, 789–801, November 24, 2010 ª2010 Elsevier Inc. 791
Figure 3. HIV-1 Fusion Is Necessary to Induce Killing of Nonproductively Infected Cells
(A and C) Concentrations of T20 that block viral infection. HLACs were infected with the indicated clones of HIV-1 in the presence of the indicated concentrations
of T20 or no drugs. One hour before incubation with the virus, cells were pretreated with T20 or left untreated. At 12 hr, cells were washed extensively and cultured
under the same conditions. On day 9, the viral concentration was determined using a p24gag FLAQ assay. The amount of p24gag accumulated in the absence of
drugs by each viral clone (A) or by SKY (C) was defined as 100%.
(B and D) Effect of T20 on indirect killing. CFSE-labeled cells were cocultured with cells infected with the indicated viral clones in the presence of 5 mM AZT and the
indicated concentrations of T20. After 6 days, indirect killing in the mixed cultures was assessed. The number of viable CFSE-positive CD4 T cells cocultured with
uninfected cells in the presence of AZT was defined as 100% (data not shown). To allow successful initial infection we pseudotyped the GIA-SKY mutants with the
VSV-G envelope. NL4-3, WT lab-adapted virus; WEAU 16-8, primary virus; SIM, T20-resistant virus; GIA-SKY, T20-dependent virus; GIA and SKY, single-domain
mutant viruses. Representative data from three independent experiments with different donors are shown.
See also Figure S2.

extensive depletion of CD4 T cells in cultures mixed with HIV- HIV gp41-Mediated Fusion Is Necessary for Depletion
infected cells but not in those mixed with uninfected cells (Fig- of Nonproductively Infected CD4 T Cells
ure 2C). The relative proportion of CD8 T cells was not altered. Studies with AMD3100 and AZT indicated that indirect CD4 T cell
CD3+/CD8– T cells were similarly depleted, indicating that the killing is mediated by events occurring between gp120-CXCR4
loss was not an artifact of downregulated surface expression binding and reverse transcription. Engagement of the chemokine
of CD4 following direct infection. Loss of CFSE-labeled CD4 coreceptor induces conformational changes in gp41, resulting in
T cells was prevented by AMD3100, which blocks the engage- insertion of viral fusion peptide on gp41 into the target T cell
ment of gp120 with CXCR4, but not by the reverse transcriptase membrane. To determine if the gp120-CXCR4 interaction alone
inhibitor AZT. Thus, productive viral replication is not required for or later events involving viral fusion are required for indirect
CD4 T cell death. killing, we evaluated the effects of enfuvirtide (T20), a fusion
To estimate the absolute numbers of all CFSE-labeled cell inhibitor that blocks six-helix bundle formation by gp41, a prereq-
subsets, we added a standard number of fluorescent beads uisite for virion fusion and core insertion.
to the cell suspensions (Figure 2D). In contrast to the sharp We first determined the optimal concentrations of T20 that
decline in CD4 T cells, the absolute numbers of CD8 T and block viral infection (Figure 3A). In NL4-3-infected cells, T20 began
B cells were unaltered. Separating the HLAC into distinct to inhibit infection at concentrations > 2 mg/ml; complete inhibition
cell types revealed that cell death occurred in purified popula- required 10 mg/ml. In cells infected with a primary viral isolate,
tions of CD4 T cells suggesting that other cell types did not WEAU 16-8 (Figure S2), infection was completely inhibited by
mediate the killing. (Figure S1 available online). In all in- 0.1 mg/ml of T20. T20 did not inhibit infection with a T20-resistant
stances, CD4-specific killing was prevented by AMD3100 mutant, SIM (Rimsky et al., 1998), regardless of concentration.
but not AZT. Importantly, the extent of CD4 T cell depletion Next, we investigated the effect of T20 on indirect CD4 T cell
in the presence of AZT was similar to that observed when killing (Figure 3B). In the absence of T20, high levels of indirect
no antiviral drugs were added (Figure 2C and Figure 1, re- killing were observed. T20 concentrations that blocked infection
spectively). Together, these results suggest that indirect also greatly inhibited indirect killing. T20 did not inhibit indirect
killing is the predominant mechanism for CD4 T cell depletion killing in cultures containing SIM-infected cells. Thus, blocking
in HIV-infected HLACs. gp41-mediated fusion prevents indirect killing.

792 Cell 143, 789–801, November 24, 2010 ª2010 Elsevier Inc.
We then examined a T20-dependent mutant, GIA-SKY (Bald- in indirect killing. Therefore, the requirement for cell-cell interac-
win et al., 2004), which fuses only when T20 is present, but tion in indirect killing may be mediated either by effective delivery
cannot initiate a spreading infection in the absence of T20 (Fig- of HIV-1 virions or by cell-associated Env.
ure 3C). Consistent with its T20 dependency, in the presence To discriminate between virion-mediated and cell-associated
of 1 mg/ml T20, the GIA-SKY mutant readily replicated while Env induction of indirect killing, we tested the effects of HIV
growth was inhibited at higher or lower T20 concentrations. protease inhibitors. These inhibitors act during the budding
The single-domain mutants GIA and SKY exhibited a T20-resis- process, resulting in immature viral particles that cannot fuse
tance phenotype similar to that of SIM. with target cells (Wyma et al., 2004). We first assessed the effect
GIA-SKY-infected cells did not induce indirect killing of CD4 of protease inhibitors on viral maturation. NL4-3 viruses carrying
T cells in the absence of T20 (Figure 3D). Indirect killing was observed a b-lactamase-Vpr (BlaM-Vpr) reporter protein were produced
in cultures treated with 1 mg/ml T20 but was inhibited at higher or in 293T cells in the presence or absence of the HIV protease
lower concentrations. Since T20-dependent viruses were bound inhibitor amprenavir. We also produced a mutant virus, TR712,
to CXCR4 before T20 was added, these findings argue that encoding a form of gp41 lacking 144 of the 150 amino acids in
CXCR4 signaling is not sufficient to elicit indirect CD4 T cell killing. the C-terminal cytoplasmic tail. This deletion largely relieves
the impaired fusogenic properties of immature HIV-1 particles
Indirect Killing Requires a Close Interaction between (Wyma et al., 2004). Protein analysis of viral lysates showed
Uninfected and HIV-Infected Cells that the NL4-3 and TR712 virions appropriately cleaved gp160
Next we examined whether indirect killing requires close contact to generate gp120 in the presence and absence of amprenavir.
with HIV-infected cells or instead can be fully supported by However, in the presence of amprenavir, an uncleaved form of
virions accumulating in the supernatants of the infected histocul- p55 Gag polypeptide rather than the mature p24 CA protein
tures. We found that cell-free supernatants from HIV-infected accumulated in both NL4-3 and TR712 virions (Figure 4D). These
histocultures were much less efficient at inducing indirect killing results confirm that amprenavir treatment of virus producing
(Figure 4A). To exclude the possibility that the concentration of cells results in the accumulation of immature particles containing
virions in the supernatants was too low, we repeated this exper- normal levels of incorporated Env proteins.
iment using a 20-fold concentrated virion supernatants (1 mg To test the ability of these viruses to fuse with target cells, we
p24/ml) but failed to detect indirect CD4 T cell killing (Figure 4B). used an HIV virion-based fusion assay that measures b-lacta-
Together, these findings suggest that close cell-cell contact is mase (BlaM) activity delivered to target cells upon the fusion
likely required for indirect killing. of virions containing BlaM fused to the Vpr protein (BlaM-Vpr)
To further explore the potential requirement of close cell-cell (Cavrois et al., 2002). Immunoblotting for BlaM confirmed that
contact for indirect killing (Sherer et al., 2007; Sourisseau et al., NL4-3 and TR712 virions incorporated Blam-Vpr in the presence
2007), we repeated these assays using cells that had been washed or absence of amprenavir (Figure 4D).
daily with fresh RPMI to prevent accumulation of HIV-1 virions and Next, SupT1 cells were infected with mature or amprenavir-
soluble factors. Such cell washing did not affect the ability of the treated immature NL4-3 or TR712 virions containing BlaM-Vpr.
resultant infected cells to mediate indirect CD4 T cell killing (Fig- Immature NL4-3 viruses displayed a 90% decline in fusogenic
ure 4B), suggesting that virions released into the medium do not properties (Figure 4E). In contrast, immature TR712 retained
participate in indirect killing. We confirmed these findings using 40% fusion capacity, indicating that the impaired fusion is not
a transwell culture system. CSFE-labeled cells and HIV-infected a result of a defective BlaM enzyme. Thus, immature virions
cells were mixed or physically separated by a transwell insert with generated in the presence of amprenavir display greatly reduced
1 mm pores, which allows free diffusion of virions but not cells. Indi- ability to fuse with target cells. Importantly, protease inhibitors
rect killing was substantial in the mixed cultures but not in the trans- did not affect the function of Env proteins expressed on infected
well cultures (Figure 4C). Together, these findings indicate that cells and did not block cell-cell fusion (Figure S3C).
indirect killing requires close interaction between CFSE-labeled We next investigated the effect of protease inhibitors on
and HIV-1-infected cells, consistent with in vitro (Garg et al., indirect killing. Remarkably, three different protease inhibitors
2007; Holm and Gabuzda, 2005) and in vivo studies showing that inhibited indirect killing as efficiently as AMD3100 (Figure 4F).
apoptotic nonproductively infected cells in human lymph nodes These results indicated that HIV-1 virions, not HIV-infected cells,
often cluster near productively infected cells (Finkel et al., 1995). are responsible for indirect CD4 T cell killing. Additionally, reca-
pitulating the efficient viral delivery of close cell-cell interactions
Indirect Killing Requires Fusion of Virions by spinoculation of free virions resulted in extensive and selec-
from Nearby HIV-Producing Cells tive indirect killing of CD4 T cells while sparing CD8 T cells and
Indirect killing required gp41-mediated fusion and close interac- B cells (Figures S3A and S3B). Thus, although indirect killing in
tion with HIV-infected cells, suggesting that cell death may be lymphoid cultures requires a close interaction between nonpro-
caused by the fusion of HIV-1 virions to CD4 T cells, syncytia ductively and productively infected cells, this killing involves
formation, or hemifusion (mixing of lipids in the absence of fusion virions rather than cell-associated Env.
pore formation) mediated by Env present on HIV-infected cells
interacting with neighboring CD4 T cells. HIV-1 virions (Holm Nonpermissive CD4 T Cells Die from Abortive Infection
et al., 2004; Jekle et al., 2003; Vlahakis et al., 2001), cell-medi- Based on these findings, we hypothesized that ‘‘indirect killing’’
ated fusion (LaBonte et al., 2000; Margolis et al., 1995), and involves an abortive form of infection, like that which occurs in
hemifusion (Garg et al., 2007) have been proposed to be involved nonpermissive resting CD4 T cells. These naive CD4 T cells

Cell 143, 789–801, November 24, 2010 ª2010 Elsevier Inc. 793
Figure 4. Killing of Nonproductively Infected CD4 T Cells Requires Fusion of Virions from Nearby HIV-1-Producing Cells
(A) Supernatants from HIV-infected HLACs are less efficient at inducing indirect killing than mixing of HIV-infected and uninfected HLACs.
(B) HIV-1 virions released into the medium do not participate in indirect killing. Replacing the mixed culture with fresh RPMI every 24 hr did not impair indirect killing.
Challenging HLACs with supernatants containing 20-fold more histoculture-derived virions (1 mg p24/ml) than normally accumulated in mixed cultures containing
infected cells (50 ng p24/ml) did not induce indirect killing. Percentages are normalized to the number of viable CFSE-positive cells depicted by an asterisk.
(C) CFSE-labeled cells are not killed when HIV-infected HLAC is physically separated by a 1 mm –pore transwell system that allows free diffusion of HIV-1 parti-
cles. Values represent the levels of viable CFSE-positive cells after 6 days of culture in the presence of the indicated drugs. Red, HIV-infected cells; blue, unin-
fected cells; green, CFSE-labeled cells.
(D) Mature and immature viruses carry equivalent amounts of envelope protein and Blam-Vpr, but differ in their content of capsid and Gag precursor. NL4-3 and
TR712 viruses were generated in 293T cells with or without amprenavir, lysed and subjected to SDS-PAGE immunoblotting analysis for gp120, p55 Gag, p24 CA,
Blam-Vpr, and free Blam.
(E) Immature viruses have reduced capacity to enter cells. SupT1 cells were mock infected or infected with mature or immature NL4-3 or TR712 virions con-
taining Blam-Vpr. After loading of cells with CCF2 dye, fusion was analyzed by flow cytometry. Percentages are the fraction of cells displaying increased cleaved
CCF2 fluorescence, indicating virion fusion.
(F) Protease inhibitors inhibit indirect killing. CFSE-labeled cells were cocultured with NL4-3-infected or uninfected cells in the presence of AZT (5 mM) alone or
together with AMD3100 (250 nM). To the indicated cultures were added 5 mM of amprenavir, saquinavir, or indinavir. Percentages are normalized to the number
of viable CFSE-positive cells depicted by an asterisk. Error bars represent the SD obtained with three independent samples from the same donor.
See also Figure S3.

exhibit an early post-entry block to HIV-1 infection that can be 2009; Unutmaz et al., 1999; Zack et al., 1990). To test this
relieved by activation with phytohemagglutinin (PHA) and inter- hypothesis, we compared the killing of activated and nonacti-
leukin-2 (IL-2) (Kreisberg et al., 2006; Santoni de Sio and Trono, vated CFSE-labeled cells in HLACs.

794 Cell 143, 789–801, November 24, 2010 ª2010 Elsevier Inc.
Figure 5. Death of Abortively Infected CD4 T Cells Is Due to Impaired Reverse Transcription
(A) Status of mixed HLACs containing either resting or activated CFSE-labeled cells, 4 days after coculturing with effector cells. Activated CFSE-labeled cells
were stimulated with PHA and IL-2 48 hr before mixing, but not during coculturing with effector cells. To avoid direct killing of activated CFSE-labeled cells in
cultures with no drugs, cell killing was terminated and analyzed 4 days after coculturing.
(B) AZT renders activated CFSE-labeled CD4 T cells sensitive to indirect killing. Resting or activated CFSE-labeled cells were cocultured with effector cells in the
presence of no drugs, AZT (5 mM) alone, or AZT and AMD3100 (250 nM). Data are from two independent experiments performed with tonsil cells from two different
donors.
(C) AZT-induced killing is lost when AZT-resistant viruses are tested. Resting or activated CFSE-labeled cells were cocultured with cells infected with NL4-3 or
HIV-1 clones #629 and #964 in the presence of no drugs, AZT (0.5 mM) alone, or AZT and AMD3100 (250 nM). AZT-sensitive and AZT-resistant sub-clones are
depicted. Data are representative of three independent experiments with three different donors.
(D) NNRTIs prevent killing of abortive infected CD4 T cells. Resting or activated CFSE-labeled cells were cocultured with infected or uninfected effector cells, in
the presence of no drugs, AZT (5 mM), AMD3100 (250 nM), the NNRTIs efavirenz (100 nM), and nevirapine (1 mM), or the integration inhibitors raltegravir (30 mM)
and 118-D-24 (60 mM). Killing of resting CFSE-labeled CD4 T cells was blocked with equal efficiency by NNRTIs and AMD3100 (columns 15, 16), but not by
integration inhibitors (columns 17, 18). In combination, NNRTIs prevented cell death induced by AZT in activated CFSE-labeled cells (compare column 38 to
44 and 45). Data are representative of four independent experiments with four different donors.
The absolute numbers of CFSE-labeled CD8 T cells and B cells was unaltered in these experiments (data not shown). Percentages are normalized to the number
of viable CFSE-positive cells depicted by an asterisk.
See also Figure S4.

CFSE-labeled cells were activated with PHA and IL-2 two days contained a small percentage of cells expressing CD25 and
before mixing with effector cells, and contained a large CD69 (Figure 5A). Either in the presence or absence of AZT,
percentage of dividing CD25 and CD69 positive cells. Nonacti- killing of resting CFSE-labeled CD4 T cells was robust (Figure 5B,
vated (resting) CFSE-labeled cells did not divide and typically columns 4 and 5, and 16 and 17). In sharp contrast, activated

Cell 143, 789–801, November 24, 2010 ª2010 Elsevier Inc. 795
Figure 6. Cytoplasmic HIV-1 DNA Triggers Proapoptotic and Proinflammatory Responses in Abortively Infected CD4 T Cells
(A) Critical reactions in HIV-1 reverse transcription as detected by probes monitoring different regions within the Strong stop, Nef, and Env DNA fragments. RDDP,
RNA-dependent DNA polymerase. Adapted from S.J. Flint et al., Principles of Virology, 2000 ASM Press, Washington DC, with permission.
(B) NNRTIs prevent accumulation of DNA elongation products. The amount of viral DNA detected by a particular probe was calculated as a fold change relative
to cells treated with no drugs (i.e., calibrator). A bactin probe was used as an internal reference. Mean cycle threshold (Ct) values of calibrator samples are
depicted. CD4 T cells were infected with WT NL4-3 produced in 293T cells, or with a Dvif NL4-3 collected from supernatants of infected HLAC, as described
in Figure S4C. Data are representative of two independent experiments performed with cells from two different donors.
(C and D) Abortive HIV-1 infection generates a coordinated proapoptotic and proinflammatory response involving caspase-3 and caspase 1 activation. HLACs
were spinoculated with no virus or with NL4-3 and AZT (5 mM), Efavirenz (100 nM), and T20 (10 mg/ml), as indicated (see Figures S3A and S3B). After 3 days, cells
were assessed by flow cytometry for intracellular levels of proinflammatory cytokines, serine 37 phosporylated p53, and activated caspases as indicated.
Ethidium monoazide was used to exclude dead and necrotic cells from the annexinV binding analysis. Data are representative of three independent experiments
with three different donors.
(E) Death of abortively infected CD4 T cells requires caspase activation. CSFE-labeled cells were cocultured with effector cells in the presence of 20 mM of Z-VAD-
FMK, a general caspase inhibitor, or Z-FA-FMK, a negative control for caspase inhibitors. AZT (5 mM); AMD3100 (250 nM). Percentages are normalized to the number
of viable CFSE-positive cells depicted by an asterisk. Error bars represent standard error of the mean of three experiments from three different HLAC donors.
(F) Abortive HIV infection promotes the maturation and secretion of IL-1b in tonsillar CD4 T cells. Isolated tonsillar CD4 T cells were either untreated, or stimulated
with PMA (phorbol-12-myristate-12-acetate, 0.5 mM) and the potassium ionophore nigericin (10 mM), or spinoculated with or without NL4-3 in the presence of

796 Cell 143, 789–801, November 24, 2010 ª2010 Elsevier Inc.
CFSE-labeled CD4 T cells were not depleted in the absence of demonstrating that a certain degree of DNA synthesis is required
AZT, but were extensively depleted in cultures containing AZT to elicit the cytopathic response.
(Figure 5B, columns 10 and 11 and 22 and 23). Addition of This notion was further strengthened in findings obtained with
AMD3100 prevented the AZT-induced killing of activated vif-deficient (Dvif) HIV-1 particles where reverse transcription is
CFSE-labeled cells, excluding nonspecific toxic effects of AZT inhibited during strong-stop DNA synthesis due to incorporated
in the activated cells (Figure 5B, columns 12 and 24). APOBEC3G (A3G) (Bishop et al., 2008; Li et al., 2007). Abortively
The ability of AZT to promote indirect killing of activated CD4 infected CD4 T cells were not depleted by Dvif NL4-3-infected
T cells suggested that cell death is triggered by impaired reverse cells (Figures S4C and S4D), indicating that termination of
transcription. To investigate this possibility, we repeated the reverse transcription before the completion of strong-stop
experiment with two pairs of AZT-resistant HIV-1 clones, 629 DNA synthesis is not sufficient to generate a cytopathic
and 964 (Larder et al., 1989). We first determined that concentra- response. Other HIV-1 mutants containing substitutions in
tions of 0.5 mM AZT block viral replication in NL4-3-infected and RNase H and nucleocapsid that promote early defects in reverse
AZT-sensitive clones and achieve half maximal inhibitory effect transcription failed to elicit indirect CD4 T cell killing (Figures S4E
in AZT-resistant clones (Figures S4A and S4B). and S4F). Together, these findings indicate that accumulation of
When resting CFSE-labeled cells were used, the extent of reverse-transcribed DNA, rather than any inherent activity of the
killing by the AZT-resistant HIV-1 viruses was similar to that HIV-1 proteins, is the key factor that triggers the death response.
obtained with NL4-3 with or without AZT (Figure 5C resting
CFSE-positive cells), demonstrating a redundant function for Abortively Infected CD4 T Cells Commence
endogenous termination of reverse transcription and AZT. Alter- but Do Not Complete Reverse Transcription
natively, when activated CFSE-labeled cells were tested, AZT- We next examined the status of HIV-1 reverse transcription in
resistant HIV-1 clones did not deplete CFSE-labeled CD4 tonsillar CD4 T cells after infection. Specifically, we investigated
T cells in the presence of AZT (Figure 5C, columns 29 and 35). the effect on reverse transcription after treatment with NNRTIs,
such as efavirenz and nevirapine, which prevent the death of
Death of Abortively Infected CD4 T Cells Is Triggered abortively infected CD4 T cells, or with AZT or integrase inhibitor
by Premature Termination of Viral DNA Elongation (raltegravir) that do not prevent CD4 T cell death. Taqman-based
We next asked what stage of reverse transcription triggers abor- quantitative real-time PCR (QPCR) was used to quantify the
tive infection cell death. AZT inhibits DNA elongation but not synthesis of reverse transcription products in isolated CD4
early DNA synthesis (Arts and Wainberg, 1994). We therefore T cells from HLAC 16 hr after infection with NL4-3. We designed
examined whether blocking early DNA synthesis with nonnu- specific QPCR primers and probes (Table S1) to monitor
cleoside reverse transcriptase inhibitors (NNRTIs) would have sequential steps in reverse transcription including generation
the same effect as AZT. Impaired reverse transcription may of strong-stop DNA, first template exchange (Nef), and DNA
also lead to abortive integration, causing chromosomal DNA strand elongation (Env) (Figure 6A). Reverse transcription
breaks and a genotoxic response. To exclude this possibility, products corresponding to strong-stop DNA were similar in
we used integrase inhibitors. To discriminate between the cyto- untreated CD4 T cells or cells treated with AZT, NNRTIs, or
pathic response induced by endogenous termination of reverse raltegravir but were greatly reduced in cells treated with
transcription and the response induced by AZT, we separately AMD3100 or in cultures infected with Dvif NL4-3 where arrest
assessed resting and activated CFSE-labeled cells. occurs prior to the completion of strong-stop DNA synthesis
Remarkably, the NNRTIs, efavirenz and nevirapine, blocked (Figure 6B columns 1–8). In contrast, the accumulation of later
indirect killing of resting CD4 T cells as efficiently as AMD3100 reverse transcription products detected by the Nef and Env
(Figure 5D, columns 15 and 16). These findings suggested that probes were dramatically inhibited by the NNRTIs but not by
allosteric inhibition of reverse transcriptase induced by these raltegravir. Levels of Nef (Figure 6B, columns 10 and 11) and
NNRTI’s interrupts reverse transcription sufficiently early to Env (columns 18 and 19) DNA products were similar in untreated
abrogate the death response. In contrast, the integrase inhibitors cells and cells treated with AZT, indicating that reverse transcrip-
raltegravir and 118-D-24 did not prevent abortive infection killing tion in most tonsillar CD4 T cells naturally terminates during DNA
(Figure 5D, columns 17 and 18), suggesting that cell death chain elongation, coinciding with the block induced by AZT. The
involves signals generated prior to viral integration. NNRTIs minor inhibition detected by AZT is likely due to a small number
also protected activated CFSE-labeled cells from death induced of permissive CD4 T cells in the culture. These results show that
by AZT (Figure 5D, column 38 versus columns 44 and 45), abortively infected CD4 T cells accumulate incomplete reverse

AZT (5 mM), AMD3100 (250 nM), and efavirenz (100 nM) as indicated. After 3 days, half of the cells were lysed and subjected to SDS-PAGE immunobloting anal-
ysis. On day 5, the supernatants from the rest of the cells were collected and subjected to SDS-PAGE immunobloting analysis. The IL-1b antibody detects the
pro-IL-1b (37kD) and the mature cleaved form (17kD). Data are the representative results of five independent experiments using tonsillar CD4 T cells isolated
from five different donors.
(G) DNA reverse transcription intermediates induce an IFN-stimulatory antiviral innate immune response (ISD). ISRE-GFP reporters were transfected with 1 mg of
HIV-1 reverse transcription intermediate products as indicated by numbers (detailed description in Figure S5E), empty DNA plasmid, or polyinosinic:polycytidylic
acid [poly(I:C)], and were analyzed by flow cytometry after 48 hr. Data are representative of three independent experiments; error bars show the SD for three
independent samples from the same experiment.
See also Figure S5 and Figure S6.

Cell 143, 789–801, November 24, 2010 ª2010 Elsevier Inc. 797
transcription products representative of DNA strand elongation. the cytokine and cytopathic response observed in tonsillar
Blocking earlier steps of reverse transcription by NNRTIs or by CD4 T cells. We next synthesized the various HIV-1 reverse
genetic mutations like deletion of Vif or mutation of RNase H transcription intermediates and tested their ability to activate
restricts accumulation of such products, and prevents abortive the ISRE-GFP reporter. Interestingly, none of the RNA-contain-
infection-induced cell death (Figure S6A). ing oligonucleotides stimulated the ISRE-GFP reporter expres-
sion above baseline. In sharp contrast, ssDNA and dsDNA
DNA Reverse Transcription Intermediates Elicit oligonucleotides longer than 500 bases in length, which corre-
a Coordinated Proapoptotic and Proinflammatory sponded to reverse transcription intermediates produced during
Response in Abortively Infected CD4 T Cells DNA elongation, evoked a potent ISRE-GFP activation (Fig-
We next evaluated whether HIV-mediated indirect killing of CD4 ure 6G). Similarly, when cells were stimulated with poly(I:C), a
T cells is associated with deregulation of cytokine production or synthetic double-stranded RNA known to activate IRF3 via the
a DNA damage response. To facilitate a vigorous and synchro- RIG-I pathway elicited a comparable ISRE-GFP response. Taken
nized killing effect, HLACs were spinoculated with NL4-3 virions together, these findings indicate that reverse transcription
in the presence of various antiviral drugs. Interestingly, based on intermediates generated during DNA chain elongation induce
immunostaining after cytokine capture, abortively infected CD4 a coordinated proapoptotic and proinflamatory innate immune
T cells expressed IFN-b, and high levels of the proinflammatory response involving caspase-3 and caspase-1 activation in
interleukin 1b (IL–1b), but not tumor necrosis factor (TNFa) abortively infected CD4 T cells.
(Figure 6C). Phosphorylation of S37 p53 was not observed,
suggesting that abortive HIV-1 infection does not induce a DISCUSSION
DNA damage cascade. Abortively infected CD4 T cells also
displayed caspase-1 and caspase-3 activity along with appear- The mechanism through which HIV-1 kills CD4 T cells, a hallmark
ance of annexin V (Figure 6D). T20 and efavirenz but not AZT of AIDS, has been a topic of vigorous research and one of the
prevented activation of these caspases, indicating that apo- most pressing questions for the field over the last 28 years
ptosis was induced by abortive HIV-1 infection. Cell death was (Thomas, 2009). In this study, we investigated the mechanism
completely prevented by Z-VAD-FMK, a pan-caspase inhibitor, of HIV-1-mediated killing in lymphoid tissues, which carry the
suggesting that caspase activation is required for the observed highest viral burdens in infected patients. We used HLACs
cytopathic response (Figure 6E). Such mode of cytokine produc- formed with fresh human tonsil cells and an experimental strategy
tion and caspase activation was not observed in CD8 T or B cells that clearly distinguishes between direct and indirect mecha-
(Figures S5B and S5C). nisms of CD4 T cell depletion. We now demonstrate that indirect
We next examined whether abortive HIV-1 infection signals for cell killing involving abortive HIV infection of CD4 T cells accounts
the maturation and secretion of IL-1b. In cells, IL–1b activity is for the vast majority of cell death occurring in lymphoid tissues.
rigorously controlled. Cells can be primed to express inactive No more than 5% of the CD4 T cells are productively infected,
pro-IL-1b by various proinflammatory signals. However, the but virtually all the remaining CD4 T cells are abortively infected
release of bioactive IL-1b requires a second signal leading to ultimately leading to caspase-mediated cell death. Equivalent
activation of inflammasomes, cleavage of pro-IL-1b by caspase findings were observed in HLACs formed with fresh human
1 and secretion of the bioactive 17 kDa form of IL-1b (Schroder spleen (Figures S6B and S6C), indicating this mechanism of
and Tschopp, 2010). Interestingly, Western blot analysis re- CD4 T cell depletion can be generalized to other lymphoid
vealed high amounts of intracellular pro-IL-1b in untreated CD4 tissues.
T cells, suggesting that tonsillar CD4 T cells are primed to release The massive depletion of nonproductively infected CD4 T cells
proinflammatory mediators (Figure 6F). Stimulating the CD4 is in contrast to their survival after infection of intact blocks of
T cells with PMA and nigericin induced further accumulation tonsillar tissue in human lymphoid histoculture (HLH) (Grivel
of pro-IL-1b and promoted the maturation and release of the et al., 2003). This result probably reflects differences between
bioactive 17 kDa IL-1b into the supernatant. Remarkably, infec- the HLH and the HLAC experimental systems. In HLH, the com-
tion of CD4 T cells with NL4-3 in the presence of AZT similarly plex three-dimensional spatial cellular organization of lymphoid
resulted in maturation and release of the bioactive 17 kDa tissue is preserved, but cellular movement and interaction are
IL-1b into the supernatant. This response was completely pre- restricted, both of which are required for indirect killing. In
vented by efavirenz and AMD3100, suggesting that abortive HLAC, the tissue is dispersed, and cells are free to interact, result-
HIV-1 infection signals the maturation and release of bioactive ing in a rapid and robust viral spread. While the mechanism
IL-1b in these CD4 T cells. triggering indirect CD4 T cell death is certainly identical in both
To identify the nature of the nucleic acid species that trigger settings, HLH allows only a slow, nearly undetectable progres-
these responses, we used a recently described H35 rat hepato- sion of indirect CD4 T cell death. In HLAC, this process is accel-
cyte cell line containing an IFN-sensitive response element erated, allowing the outcome to be detected in a few days. Inter-
(ISRE) linked to GFP (Patel et al., 2009). H35 cells were first estingly, indirect killing was also less efficient when peripheral
infected with pseudotyped VSV-G HIV-1 virions. These virions blood cells were tested (data not shown). It is possible that cellular
induced GFP expression and cell death in the presence or factors specifically produced in lymphoid organs are required to
absence of AZT. Importantly, the expression of GFP and cell accelerate indirect killing of peripheral blood CD4 T cells.
death response were blocked by efavirenz but not raltegravir Several mechanisms have been proposed to explain indirect
(Figure S5D). Thus, the H35 system successfully reconstitutes CD4 T cell killing during HIV infection. Our finding that CD4

798 Cell 143, 789–801, November 24, 2010 ª2010 Elsevier Inc.
Figure 7. Consequences of Inhibiting Early Steps of HIV-1 Infection on CD4 T Cell Death
(A) The nonpermissive state of most CD4 T cells in lymphoid tissue leads to endogenous termination of reverse transcription during DNA chain elongation (i.e.,
‘‘killing zone’’). As a result, DNA intermediates accumulate in the cytoplasm and elicit a multifaceted proapoptotic and proinflammatory innate immune defense
programs, coordinated by IFN-stimulatory DNA (ISD) response, caspase-3, caspase-1, and IL-1b, to restrict viral spread. Different classes of antiretroviral drugs
act at different stage of the HIV life cycle. NNRTIs like efavirenz and nevirapine inhibit early steps of DNA synthesis and therefore prevent such response and the
consequence CD4 T cell death. AZT is less efficient at blocking DNA synthesis and therefore unable to abrogate this response.
(B) In permissive CD4 T cells reverse transcription proceeds, allowing HIV-1 to bypass the ‘‘killing zone’’ and move on to productive (or latent) infection.
Interrupting reverse transcription with AZT traps the virus in the ‘‘killing zone’’ and induces cell death. EFV, efavirenz; NVP, nevirapine; and RTGR, raltegravir.
See also Figure S6.

T cell death is blocked by entry and fusion inhibitors but not by defense (Rehwinkel and Reis e Sousa, 2010). Our results suggest
AZT, strongly suggested that such killing involves nonproductive that abortive HIV-1 infection also stimulates activation of
infection of CD4 T cells. Therefore, we focused on events that caspase-3, which is linked to apoptosis, and caspase-1, which
occur after HIV-1 entry. Our investigations demonstrate that promotes the processing and secretion of the proinflammatory
abortive viral DNA synthesis occurring in nonpermissive, quies- cytokines like IL–1b. It is certainly possible that pyroptosis
cent CD4 tonsil T cells, plays a key role in the cell death elicited in response to caspase-1 activation also contributes to
response. Conversely, in the small subset of permissive target the observed cytopathic response (Schroder and Tschopp,
cells, reverse transcription is not interrupted, minimizing the 2010). The release of inflammatory cytokines during CD4 T cell
accumulation and subsequent detection of such reverse death could also contribute to the state of chronic inflammation
transcription intermediates (Figure 7). that characterizes HIV infection. This inflammation may fuel
Interrupted or slowed reverse transcription may create persis- further viral spread by recruiting uninfected lymphocytes to the
tent exposure to cytoplasmic DNA products that elicit an antiviral inflamed zone. While this innate response was likely designed
innate immune response coordinated by activation of type I IFNs to protect the host, it is subverted in the case of HIV infection
(Stetson and Medzhitov, 2006). Such activation, termed and importantly contributes to the immunopathogenic effects
IFN-stimulatory DNA (ISD) response, may be analogous to the characteristic of HIV infection and AIDS.
type I IFN response triggered by the RIG-I-like receptor (RLR) Such antiviral pathways comprise an unrecognized cell-
family of RNA helicases that mediate a cell-intrinsic antiviral intrinsic retroviral detection system (Manel et al., 2010; Stetson

Cell 143, 789–801, November 24, 2010 ª2010 Elsevier Inc. 799
et al., 2008). Viral RNA in infected cells is recognized by QPCR reactions were performed in an ABI Prism 7900HT (Applied
members of the RIG-I-like family of receptors that detect specific Biosystems).
RNA patterns like uncapped 50 triphosphate (Rehwinkel and
ISRE-GFP H35 Reporter Cells, Microscopy, and Generation
Reis e Sousa, 2010). Although uncapped RNA intermediates
of Synthetic HIV-1 Reverse Transcription Intermediates
are generated by the HIV-1 RNase H, they contain a 50 mono-
H35 rat hepatic cells containing an ISRE-GFP reporter were maintained as
phosphate and therefore may be not recognized by the RIG-I described (Patel et al., 2009). For microscopy imaging, ISRE-GFP reporter
system (Figure 6G). In contrast to RNA receptors, intracellular H35 cells were infected with a replication competent VSV-G pseudotyped
sensing of viral DNA remains poorly understood. Consequently, NL4-3 and analyzed using an Axio observer Z1 microscope (Zeiss). Transfec-
it is unclear how HIV-1 DNA intermediates are detected in the tions and generation of synthetic HIV-1 reverse transcription intermediates are
cytoplasm of abortively infected CD4 T cells. AIM2 (absent in described in detail in Figure S5E and Extended Experimental Procedures.

melanoma 2) was recently identified as a cytoplasmic dsDNA


receptor that induces cell death in macrophages through activa- SUPPLEMENTAL INFORMATION
tion of caspase-1 in imflammasomes (Hornung et al., 2009). Our
Supplemental Information includes Extended Experimental Procedures,
preliminary investigations have not supported a role for AIM2 in six figures, and one table and can be found with this article online at
cell death induced by abortive HIV infection (data not shown), doi:10.1016/j.cell.2010.11.001.
suggesting the potential involvement of a different DNA-sensing
mechanism. We also have not identified a role for TLR9 and ACKNOWLEDGMENTS
MYD88 signaling in this form of cell death. Additional candidate
sensors recognizing cytoplasmic HIV-1 DNA are now under We thank David N. Levy for the NLENG1 plasmid; David Fenard for the NL4-3
study. variant plasmids SIM, GIA, GIA-SKY, and SKY; George M. Shaw for the WEAU
16-8 env clone; and Suraj J. Patel, Kevin R. King, and Martin L. Yarmush for the
In summary, both productive and nonproductive forms of HIV
H35 ISRE-GFP reporter cell line. The following reagents were obtained through
infection contribute to the pathogenic effects of this lentivirus. the AIDS Research and Reference Reagent Program, Division of AIDS, NIAID,
The relative importance of these different cell death pathways NIH: AMD3100, T-20, Saquinavir, Amprenavir, Indinavir, Nevirapine, Efavirenz,
might well vary with the stage of HIV infection. For example, and AZT-resistant HIV-1 clones #629 and #964. Special thanks to Dr. Eva
direct infection and death might predominate during acute Herker for assistance with fluorescence microscopy; to Dr. Stefanie Sowinski
infection where CCR5-expressing memory CD4 T cells in gut- for help with assessing inflammatory responses in primary immune cells; and
to Jason Neidleman for stimulating discussions and technical advice. We also
associated lymphoid tissue are effectively depleted. Conversely,
thank Marty Bigos for assistance with the flow cytometry; Stephen Ordway
the CXCR4-dependent indirect killing we describe in tonsil tissue and Gary Howard for editorial assistance; John C.W. Carroll and Alisha Wilson
may reflect later stages of HIV-induced disease where a switch for graphics; and Robin Givens and Sue Cammack for administrative assis-
to CXCR4 coreceptor usage occurs in approximately 50% of tance. Funding for this project was provided by the Universitywide AIDS
infected subjects. Research Program, F04-GIVI-210 (G.D.); the UCSF-GIVI Center for AIDS
The current study demonstrates how a cytopathic response Research, NIH/NIAID P30 AI027763 (M.C.); the Francis Goelet Fellowship
(K.G.L.); and the UCSF Medical Scientist Training Program, NIH/NIGMS T32
involving abortive viral infection of resting nonpermissive CD4
GM007618-32 (O.Z.).
T cells can lead not only to CD4 T cell depletion but also to the
release of proinflammatory cytokines. The ensuing recruitment Received: November 5, 2009
of new target cells to the site of inflammation may fuel a vicious Revised: May 7, 2010
cycle of continuing infection and CD4 T cell death centrally Accepted: October 29, 2010
contributing to HIV pathogenesis. Published: November 24, 2010

EXPERIMENTAL PROCEDURES REFERENCES

Culture and Infection of HLACs Arts, E.J., and Wainberg, M.A. (1994). Preferential incorporation of nucleoside
Human tonsil or splenic tissues were obtained from the National Disease analogs after template switching during human immunodeficiency virus
Research Interchange and the Cooperative Human Tissue Network and pro- reverse transcription. Antimicrob. Agents Chemother. 38, 1008–1016.
cessed as previously described (Jekle et al., 2003). For a detailed description Baldwin, C.E., Sanders, R.W., Deng, Y., Jurriaans, S., Lange, J.M., Lu, M., and
see Extended Experimental Procedures. Berkhout, B. (2004). Emergence of a drug-dependent human immunodefi-
ciency virus type 1 variant during therapy with the T20 fusion inhibitor. J.
FACS Analysis and Gating Strategy, Preparation of HIV-1 Virions, Virol. 78, 12428–12437.
and Virion-Based Fusion Assay Bishop, K.N., Verma, M., Kim, E.Y., Wolinsky, S.M., and Malim, M.H. (2008).
Data were collected on a FACS Calibur (BD Biosciences) and analyzed with APOBEC3G inhibits elongation of HIV-1 reverse transcripts. PLoS Pathog. 4,
Flowjo software (Treestar). HIV-1 viruses were generated by transfection of e1000231.
proviral DNA into 293T cells by the calcium phosphate method. Virion-based
Cavrois, M., De Noronha, C., and Greene, W.C. (2002). A sensitive and specific
fusion assay was performed as previously described (Cavrois et al., 2002).
enzyme-based assay detecting HIV-1 virion fusion in primary T lymphocytes.
Detailed protocols are provided in the supplemental experimental procedures.
Nat. Biotechnol. 20, 1151–1154.

Spinoculation and Taqman-Based QPCR Analysis Eckstein, D.A., Penn, M.L., Korin, Y.D., Scripture-Adams, D.D., Zack, J.A.,
of HIV-1-Infected CD4 T Cells Kreisberg, J.F., Roederer, M., Sherman, M.P., Chin, P.S., and Goldsmith,
The spinoculation method is described in detail in Figures S3A and S3B. M.A. (2001). HIV-1 actively replicates in naive CD4(+) T cells residing within
Isolation of HLAC CD4 T cells and QPCR protocol are described in detail human lymphoid tissues. Immunity 15, 671–682.
in supplemental experimental procedures. Primers and probes sequences Finkel, T.H., Tudor-Williams, G., Banda, N.K., Cotton, M.F., Curiel, T., Monks,
used to detect reverse transcription products are provided in Table S1. C., Baba, T.W., Ruprecht, R.M., and Kupfer, A. (1995). Apoptosis occurs

800 Cell 143, 789–801, November 24, 2010 ª2010 Elsevier Inc.
predominantly in bystander cells and not in productively infected cells of HIV- Margolis, L.B., Glushakova, S., Baibakov, B., and Zimmerberg, J. (1995).
and SIV-infected lymph nodes. Nat. Med. 1, 129–134. Syncytium formation in cultured human lymphoid tissue: fusion of implanted
Gandhi, R.T., Chen, B.K., Straus, S.E., Dale, J.K., Lenardo, M.J., and HIV glycoprotein 120/41-expressing cells with native CD4+ cells. AIDS Res.
Baltimore, D. (1998). HIV-1 directly kills CD4+ T cells by a Fas-independent Hum. Retroviruses 11, 697–704.
mechanism. J. Exp. Med. 187, 1113–1122. Patel, S.J., King, K.R., Casali, M., and Yarmush, M.L. (2009). DNA-triggered
Garg, H., Joshi, A., Freed, E.O., and Blumenthal, R. (2007). Site-specific innate immune responses are propagated by gap junction communication.
mutations in HIV-1 gp41 reveal a correlation between HIV-1-mediated Proc. Natl. Acad. Sci. USA 106, 12867–12872.
bystander apoptosis and fusion/hemifusion. J. Biol. Chem. 282, 16899–16906. Rehwinkel, J., and Reis e Sousa, C. (2010). RIGorous detection: exposing virus
Glushakova, S., Baibakov, B., Margolis, L.B., and Zimmerberg, J. (1995). through RNA sensing. Science 327, 284–286.
Infection of human tonsil histocultures: a model for HIV pathogenesis. Nat. Rimsky, L.T., Shugars, D.C., and Matthews, T.J. (1998). Determinants of
Med. 1, 1320–1322. human immunodeficiency virus type 1 resistance to gp41-derived inhibitory
Grivel, J.C., Biancotto, A., Ito, Y., Lima, R.G., and Margolis, L.B. (2003). peptides. J. Virol. 72, 986–993.
Bystander CD4+ T lymphocytes survive in HIV-infected human lymphoid
Santoni de Sio, F.R., and Trono, D. (2009). APOBEC3G-depleted resting CD4+
tissue. AIDS Res. Hum. Retroviruses 19, 211–216.
T cells remain refractory to HIV1 infection. PLoS ONE 4, e6571.
Hayden, M.S., Palacios, E.H., and Grant, R.M. (2003). Real-time quantitation of
Schindler, M., Münch, J., Kutsch, O., Li, H., Santiago, M.L., Bibollet-Ruche, F.,
HIV-1 p24 and SIV p27 using fluorescence-linked antigen quantification
Müller-Trutwin, M.C., Novembre, F.J., Peeters, M., Courgnaud, V., et al.
assays. AIDS 17, 629–631.
(2006). Nef-mediated suppression of T cell activation was lost in a lentiviral
Herbeuval, J.P., Grivel, J.C., Boasso, A., Hardy, A.W., Chougnet, C., Dolan, lineage that gave rise to HIV-1. Cell 125, 1055–1067.
M.J., Yagita, H., Lifson, J.D., and Shearer, G.M. (2005). CD4+ T-cell death
Schroder, K., and Tschopp, J. (2010). The inflammasomes. Cell 140, 821–832.
induced by infectious and noninfectious HIV-1: role of type 1 interferon-depen-
dent, TRAIL/DR5-mediated apoptosis. Blood 106, 3524–3531. Sherer, N.M., Lehmann, M.J., Jimenez-Soto, L.F., Horensavitz, C., Pypaert,
Holm, G.H., and Gabuzda, D. (2005). Distinct mechanisms of CD4+ and CD8+ M., and Mothes, W. (2007). Retroviruses can establish filopodial bridges for
T-cell activation and bystander apoptosis induced by human immunodefi- efficient cell-to-cell transmission. Nat. Cell Biol. 9, 310–315.
ciency virus type 1 virions. J. Virol. 79, 6299–6311. Sourisseau, M., Sol-Foulon, N., Porrot, F., Blanchet, F., and Schwartz, O.
Holm, G.H., Zhang, C., Gorry, P.R., Peden, K., Schols, D., De Clercq, E., and (2007). Inefficient human immunodeficiency virus replication in mobile lympho-
Gabuzda, D. (2004). Apoptosis of bystander T cells induced by human cytes. J. Virol. 81, 1000–1012.
immunodeficiency virus type 1 with increased envelope/receptor affinity and Stetson, D.B., Ko, J.S., Heidmann, T., and Medzhitov, R. (2008). Trex1
coreceptor binding site exposure. J. Virol. 78, 4541–4551. prevents cell-intrinsic initiation of autoimmunity. Cell 134, 587–598.
Hornung, V., Ablasser, A., Charrel-Dennis, M., Bauernfeind, F., Horvath, G., Stetson, D.B., and Medzhitov, R. (2006). Recognition of cytosolic DNA
Caffrey, D.R., Latz, E., and Fitzgerald, K.A. (2009). AIM2 recognizes cytosolic activates an IRF3-dependent innate immune response. Immunity 24, 93–103.
dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature
Swiggard, W.J., O’Doherty, U., McGain, D., Jeyakumar, D., and Malim, M.H.
458, 514–518, Epub 2009 Jan 2021.
(2004). Long HIV type 1 reverse transcripts can accumulate stably within
Jekle, A., Keppler, O.T., De Clercq, E., Schols, D., Weinstein, M., and resting CD4+ T cells while short ones are degraded. AIDS Res. Hum.
Goldsmith, M.A. (2003). In vivo evolution of human immunodeficiency virus Retroviruses 20, 285–295.
type 1 toward increased pathogenicity through CXCR4-mediated killing of
Thomas, C. (2009). Roadblocks in HIV research: five questions. Nat. Med. 15,
uninfected CD4 T cells. J. Virol. 77, 5846–5854.
855–859.
Kamata, M., Nagaoka, Y., and Chen, I.S. (2009). Reassessing the role of
APOBEC3G in human immunodeficiency virus type 1 infection of quiescent Unutmaz, D., KewalRamani, V.N., Marmon, S., and Littman, D.R. (1999).
CD4+ T-cells. PLoS Pathog 5, e1000342. Cytokine signals are sufficient for HIV-1 infection of resting human T lympho-
cytes. J. Exp. Med. 189, 1735–1746.
Kreisberg, J.F., Yonemoto, W., and Greene, W.C. (2006). Endogenous factors
enhance HIV infection of tissue naive CD4 T cells by stimulating high molecular Vlahakis, S.R., Algeciras-Schimnich, A., Bou, G., Heppelmann, C.J., Villasis-
mass APOBEC3G complex formation. J. Exp. Med. 203, 865–870. Keever, A., Collman, R.C., and Paya, C.V. (2001). Chemokine-receptor activa-
tion by env determines the mechanism of death in HIV-infected and uninfected
LaBonte, J.A., Patel, T., Hofmann, W., and Sodroski, J. (2000). Importance of
T lymphocytes. J. Clin. Invest. 107, 207–215.
membrane fusion mediated by human immunodeficiency virus envelope
glycoproteins for lysis of primary CD4-positive T cells. J. Virol. 74, 10690– Westendorp, M.O., Frank, R., Ochsenbauer, C., Stricker, K., Dhein, J.,
10698. Walczak, H., Debatin, K.M., and Krammer, P.H. (1995). Sensitization of
Larder, B.A., Darby, G., and Richman, D.D. (1989). HIV with reduced sensitivity T cells to CD95-mediated apoptosis by HIV-1 Tat and gp120. Nature 375,
to zidovudine (AZT) isolated during prolonged therapy. Science 243, 1731– 497–500.
1734. Wyma, D.J., Jiang, J., Shi, J., Zhou, J., Lineberger, J.E., Miller, M.D., and
Levy, D.N., Aldrovandi, G.M., Kutsch, O., and Shaw, G.M. (2004). Dynamics of Aiken, C. (2004). Coupling of human immunodeficiency virus type 1 fusion to
HIV-1 recombination in its natural target cells. Proc. Natl. Acad. Sci. USA 101, virion maturation: a novel role of the gp41 cytoplasmic tail. J. Virol. 78,
4204–4209. 3429–3435.
Li, X.Y., Guo, F., Zhang, L., Kleiman, L., and Cen, S. (2007). APOBEC3G Zack, J.A., Arrigo, S.J., Weitsman, S.R., Go, A.S., Haislip, A., and Chen, I.S.
inhibits DNA strand transfer during HIV-1 reverse transcription. J. Biol. (1990). HIV-1 entry into quiescent primary lymphocytes: molecular analysis
Chem. 282, 32065–32074. reveals a labile, latent viral structure. Cell 61, 213–222.
Manel, N., Hogstad, B., Wang, Y., Levy, D.E., Unutmaz, D., and Littman, D.R. Zhou, Y., Zhang, H., Siliciano, J.D., and Siliciano, R.F. (2005). Kinetics of
(2010). A cryptic sensor for HIV-1 activates antiviral innate immunity in human immunodeficiency virus type 1 decay following entry into resting CD4+
dendritic cells. Nature 467, 214–217. T cells. J. Virol. 79, 2199–2210.

Cell 143, 789–801, November 24, 2010 ª2010 Elsevier Inc. 801
Sirt3 Mediates Reduction of Oxidative
Damage and Prevention of Age-Related
Hearing Loss under Caloric Restriction
Shinichi Someya,1,3,5 Wei Yu,2,5 William C. Hallows,2 Jinze Xu,4 James M. Vann,1 Christiaan Leeuwenburgh,4
Masaru Tanokura,3 John M. Denu,2,* and Tomas A. Prolla1,*
1Departments of Genetics and Medical Genetics
2Department of Biomolecular Chemistry
University of Wisconsin, Madison, WI 53706, USA
3Department of Applied Biological Chemistry, University of Tokyo, Yayoi, Tokyo 113-8657, Japan
4Department of Aging and Geriatrics and The Institute on Aging, University of Florida, Gainesville, FL 32611, USA
5These authors contributed equally to this work

*Correspondence: jmdenu@wisc.edu (J.M.D.), taprolla@wisc.edu (T.A.P.)


DOI 10.1016/j.cell.2010.10.002

SUMMARY druch and Walford, 1988; Sohal and Weindruch, 1996; Someya
et al., 2007; Colman et al., 2009). Furthermore, CR reduces neu-
Caloric restriction (CR) extends the life span and rodegeneration in animal models of Parkinson’s disease (Matt-
health span of a variety of species and slows the son, 2000) as well as Alzheimer’s disease (Zhu et al., 1999).
progression of age-related hearing loss (AHL), The mitochondrial free radical theory of aging postulates that
a common age-related disorder associated with aging results from accumulated oxidative damage caused by
oxidative stress. Here, we report that CR reduces reactive oxygen species (ROS), originating from the mitochon-
drial respiratory chain (Balaban et al., 2005). Consistent with
oxidative DNA damage in multiple tissues and pre-
this hypothesis, mitochondria are a major source of ROS and
vents AHL in wild-type mice but fails to modify
of ROS-induced oxidative damage, and mitochondrial function
these phenotypes in mice lacking the mitochondrial declines during aging (Wallace, 2005). A large body of evidence
deacetylase Sirt3, a member of the sirtuin family. suggests that CR reduces the age-associated accumulation of
In response to CR, Sirt3 directly deacetylates and oxidatively damaged proteins, lipids, and DNA through reduction
activates mitochondrial isocitrate dehydrogenase 2 of oxidative damage to these macromolecules and/or enhanced
(Idh2), leading to increased NADPH levels and an antioxidant defenses to oxidative stress (Weindruch and Wal-
increased ratio of reduced-to-oxidized glutathione ford, 1988; Sohal and Weindruch, 1996; Masoro, 2000). Yet,
in mitochondria. In cultured cells, overexpression whether the anti-aging action of CR in mammals is a regulated
of Sirt3 and/or Idh2 increases NADPH levels and process and requires specific regulatory proteins such as sir-
protects from oxidative stress-induced cell death. tuins still remains unclear.
Sirtuins are NAD+-dependent protein deacetylases that regu-
Therefore, our findings identify Sirt3 as an essential
late life span in lower organisms and have emerged as broad
player in enhancing the mitochondrial glutathione
regulators of cellular fate and mammalian physiology (Donmez
antioxidant defense system during CR and suggest and Guarente, 2010; Finkel et al., 2009). A previous report has
that Sirt3-dependent mitochondrial adaptations shown that life span extension by CR in yeast requires Sir2,
may be a central mechanism of aging retardation in a member of the sirtuin family (Lin et al., 2000), linking sirtuins
mammals. and CR-mediated retardation of aging. In mammals, there are
seven sirtuins that display diverse cellular localization (Donmez
INTRODUCTION and Guarente, 2010; Finkel et al., 2009). Previous studies have
focused on the role of Sirt1 as the major sirtuin mediating the
It is well established that reducing food consumption by 25%– metabolic effects of CR in mammals (Chen et al., 2005; Bor-
60% without malnutrition consistently extends both the mean done et al., 2007; Chen et al., 2008). However, recent studies
and maximum life span of rodents (Weindruch and Walford, indicate that upregulation of Sirt1 in response to CR is not
1988; Koubova and Guarente, 2003). Caloric restriction (CR) is observed in all tissues examined (Cohen et al., 2004; Barger
also known to extend life span in yeast, worms, fruit flies, et al., 2008), and currently, no study has provided conclusive
spiders, birds, and monkeys and delays the progression of evidence that sirtuins play an essential role in CR-mediated
a variety of age-associated diseases such as cancer, diabetes, aging retardation in mammals. Sirt3 is a member of the
cataract, and age-related hearing loss (AHL) in mammals (Wein- mammalian sirtuin family that is localized to mitochondria and

802 Cell 143, 802–812, November 24, 2010 ª2010 Elsevier Inc.
regulates levels of ATP and the activity of complex I of the elec- phenotype (AHL) require a member of the sirtuin family in
tron transport chain (Ahn et al., 2008) and, as such, may play mammals.
a role in the metabolic reprogramming mediated by CR. A
recent study has shown that CR increases Sirt3 levels in liver RESULTS
mitochondria (Schwer et al., 2009). Fasting also increases
Sirt3 protein expression in liver mitochondria, and mice lacking Sirt3 Is Required for the CR-Mediated Prevention
Sirt3 display the hallmarks of fatty acid oxidation disorders, of Age-Related Cochlear Cell Death and Hearing Loss
indicating that Sirt3 modulates mitochondrial fatty acid oxida- First, to investigate whether Sirt3 plays a role in the CR preven-
tion in mammals (Hirschey et al., 2010). Furthermore, CR tion of AHL, we conducted a 10 month CR dietary study using
increases expression of Sirt3 in primary mouse cardiomyo- WT and Sirt3/ mice that have been backcrossed onto the
cytes, whereas overexpression of Sirt3 protects these cells C57BL/6J background. The C57BL/6J strain is considered an
from oxidative stress-induced cell death (Sundaresan et al., excellent model to study the anti-aging action of CR because
2008), suggesting a potential role of Sirt3 in the aging retarda- this mouse strain is the most widely used mouse model for the
tion associated with CR in mammals. study of aging and responds to CR with a robust extension of
AHL is a universal feature of mammalian aging and is the life span (Weindruch and Walford, 1988) and prevention of AHL
most common sensory disorder in the elderly (Someya and (Someya et al., 2007). We reduced the calorie intake of WT and
Prolla, 2010; Liu and Yan, 2007). AHL is characterized by an Sirt3/ mice to 75% (a 25% CR) of that fed to control diet
age-associated decline of hearing function associated with (CD) mice in early adulthood (2 months of age), and this dietary
loss of spiral ganglion neurons and sensory hair cells in the regimen was maintained until 12 months of age. The auditory
cochlea of the inner ear (Someya and Prolla, 2010; Liu and brainstem response (ABR), a common electrophysiological test
Yan, 2007). The progressive loss of neurons and hair cells in of hearing function, was used to monitor the progression of
the inner ear leads to the onset of AHL because these postmi- AHL in these mice (Someya et al., 2009). We first confirmed
totic cells do not regenerate in mammals. The onset of AHL that aging resulted in increased ABR hearing thresholds at the
begins in the high-frequency region and spreads toward the high (32 kHz), middle (16 kHz), and low (8 kHz) frequencies in
low-frequency region during aging (Keithley et al., 2004; Hunter 12-month-old WT mice (Figure 1A), indicating that these mice
and Willott, 1987). This is accompanied by the loss of neurons displayed hearing loss. As predicted, CR delayed the progres-
and hair cells beginning in the basal region and spreading sion of AHL at all tested frequencies in WT mice (Figure 1A).
toward the apex of the cochlea of the inner ear with age. Strikingly, CR did not delay the progression of AHL in Sirt3/
A previous study has shown that CR slows the progression of mice (Figure 1A), although CR had the same effect on body
AHL in CBA/J mice (Sweet et al., 1988), whereas we have weight reduction in both WT and Sirt3/ mice (Figures S2A
shown previously that CR prevents AHL in C57BL/6J mice, and S2B available online). Neural and hair cell degeneration
reduces cochlear degeneration, and induces Sirt3 in the are hallmarks of AHL (Keithley et al., 2004). In agreement with
cochlea (Someya et al., 2007). Both strains of mice have been the hearing test results, basal regions of the cochleae from
extensively used as a model of AHL, although the age of onset calorie-restricted WT mice displayed only minor loss of spiral
of AHL varies from 12–15 months of age in C57BL/6J mice to ganglion neurons (Figures 1J and 1K; see also Figures 1B, 1C,
18–22 months of age in CBA/J mice (Zheng et al., 1999). Exper- 1F and 1G) and hair cells (Figure S1E; see also Figures S1A
imental evidence suggests that oxidative stress plays a major and S1C), whereas CR failed to protect these cells in Sirt3/
role in AHL (Jiang et al., 2007; Someya et al., 2009) and that mice (Figures 1L and 1M; see also Figures 1D, 1E, 1H, and 1I;
CR protects cochlear cells through reduction of oxidative Figure S1F; see also Figures S1B and S1D). Collectively, these
damage and/or by enhancing cellular antioxidant defenses to results demonstrate that Sirt3 plays an essential role in the CR-
oxidative stress (Someya et al., 2007). Yet, the molecular mech- mediated prevention of age-related cochlear cell death and
anisms by which CR reduces oxidative cochlear cell damage hearing loss in mice.
remain unknown. Next, to investigate whether Sirt3 plays a role in the metabolic
In this report, we show that the mitochondrial deacetylase effects induced by CR, we conducted a 3 month CR dietary
Sirt3 is required for the CR-mediated prevention of AHL in study using WT and Sirt3/ mice starting at 2 months of age.
mice. We also show that Sirt3 is required for the reduction of Mice lacking the Sirt3 gene appeared phenotypically normal
oxidative damage in multiple tissues under CR conditions, as under basal and CR conditions: Sirt3/ mice were viable
evidenced by DNA damage levels. At the mechanistic level, and fertile, and no significant changes were observed in
Sirt3 directly deacetylates isocitrate dehydrogenase 2 (Idh2), body weight (Figures S2A and S2B), bone mineral density (Fig-
an enzyme that converts NADP+ to NADPH in mitochondria. ure S2C), body fat (Figure S2D), tissue weight (Figure S2E),
In response to CR, Sirt3 stimulates Idh2 activity in mitochon- serum glucose levels (Figure S3A), glucose tolerance (Fig-
dria, leading to increased levels of NADPH and an increased ure S3B), serum Igf-1 (Figure S3C), and cholesterol (Figure S3D)
ratio of reduced glutathione/oxidized glutathione, the major levels between control diet WT and Sirt3/ mice or calorie-
redox couple in the cell. In cultured cells, overexpression of restricted WT and Sirt3/ mice at 5 months of age. However,
Sirt3 and/or Idh2 increases NADPH levels and protects these though we found that WT mice displayed lower levels of serum
cells from oxidative stress. The data presented here provide insulin (Figure S3E) and triglycerides (Figure S3F) in response
the first conclusive evidence that CR-mediated reduction of to CR, no significant changes were observed in these serum
oxidative damage and prevention of a common age-related markers between control diet-fed and calorie-restricted Sirt3/

Cell 143, 802–812, November 24, 2010 ª2010 Elsevier Inc. 803
A WT Sirt3-/- Figure 1. CR Prevents AHL and Protects
Cochlear Neurons in WT Mice, but Not in
100 2mo CD 100 2mo CD
12mo CD Sirt3/ Mice
12mo CD
12mo CR 12mo CR (A) ABR hearing thresholds were measured at 32,
Threshold (dB SPL)

*
75 * 75 16, and 8 kHz from control diet and/or calorie-
* *
* restricted WT (left) and Sirt3/ (right) mice at
*
50 50 2 and 12 months of age (n = 9–12). *Significantly
different from 2-month-old WT or Sirt3/ mice
25 ** ** 25 (p < 0.05), **significantly different from 12-month-
**
old WT mice (p < 0.05). CD, control diet; CR,
0 0 calorie restricted diet.
8 16 32 8 16 32 (B–M) Neurons in the basal cochlear regions from
Frequency (kHz) WT mice in control diet at 2 (B and C) and 12
(F and G) months of age and calorie-restricted
WT WT Sirt3-/- Sirt3-/- diet at 12 months of age (J and K). Neurons from
control diet Sirt3/ mice at 2 (D and E) and 12
B C D E (H and I) months of age and calorie-restricted
Sirt3/ mice at 12 months of age (L and M)
2 mo CD

(n = 5). Arrows in the lower-magnification photos


indicate neuron regions. Scale bars, 100 mm (B,
F, J, D, H, and L) and 20 mm (C, G, J, E, I, and M).
Data are means ± SEM. See also Figure S1, Fig-
ure S2, and Figure S3.
F G H I
12 mo CD

mice (Figures 2C–2E). Together, these


results provide evidence that Sirt3 plays
an essential role in the CR-mediated
J K L M reduction of oxidative DNA damage in
multiple tissues.
12 mo CR

Sirt3 Enhances the Mitochondrial


Glutathione Antioxidant Defense
System in Response to CR
A previous study has shown that overex-
pression of Sirt3 increased mRNA
expression of the antioxidant genes
mice, suggesting a possible role of Sirt3 in metabolic adapta- manganese superoxide dismutase (MnSOD) and catalase (Cat)
tions to CR. in primary cardiomyocytes and that Sirt3/ primary cardiomyo-
cytes displayed higher levels of ROS compared to those of WT
Sirt3 Is Required for the CR-Mediated Reduction cells (Sundaresan et al., 2009), suggesting that Sirt3 may regu-
of Oxidative Damage in Multiple Tissues late the antioxidant systems. Glutathione acts as the major small
How does Sirt3 reduce cochlear cell degeneration and slow the molecule antioxidant in cells (Anderson, 1998; Halliwell and Gut-
progression of AHL in response to CR? It is well established that teridge, 2007; Marı́ et al., 2009; Rebrin et al., 2003), and NADPH-
CR reduces oxidative damage to DNA, proteins, and lipids in dependent glutathione reductase regenerates reduced gluta-
multiple tissues in mammals (Sohal and Weindruch, 1996; thione (GSH) from oxidized glutathione (GSSG) (Anderson,
Masoro, 2000; Hamilton et al., 2001). Hence, we hypothesized 1998; Marı́ et al., 2009). In healthy mitochondria from young
that Sirt3 may play a role in the CR-mediated reduction of oxida- mice, glutathione is found mostly in the reduced form, GSH
tive damage in the cochlea and other tissues. To test this hypoth- (Marı́ et al., 2009). During aging, oxidized glutathione accumu-
esis, we measured oxidative damage to DNA in the cochleae, lates, and hence an altered ratio of mitochondrial GSH to
brain (neocortex), and liver of control diet and calorie-restricted GSSG is thought to be a marker of both oxidative stress and
WT and Sirt3/ mice at 12 months of age. We found that CR aging (Rebrin et al., 2003; Schafer and Buettner, 2001; Marı́
reduced oxidative DNA damage in WT mice, as determined by et al., 2009). Thus, we hypothesized that Sirt3 may regulate the
measurements of 8-hydroxyguanosine and apurinic/aprimidinic mitochondrial glutathione antioxidant system under CR condi-
(AP) sites, but failed to reduce oxidative DNA damage in tissues tions. To test this hypothesis, we measured the ratio of
from Sirt3/ mice (Figures 2A and 2B). In agreement with the GSH:GSSG in the mitochondria of the inner ear, brain, and liver
oxidative damage results, CR increased spiral ganglion neuron of control diet and calorie-restricted WT and Sirt3/ mice at
survival (Figure 2C), outer hair cell survival (Figure 2D), and inner 5 months of age. Mitochondrial GSSG levels decreased during
hair cell survival (Figure 2E) in the basal regions of the cochleae of CR in the inner ear from WT mice, but not from Sirt3/ mice (Fig-
WT mice, whereas CR failed to protect these cells in Sirt3/ ure 3B; see also Figure 3C). We also found that the ratios of

804 Cell 143, 802–812, November 24, 2010 ª2010 Elsevier Inc.
A B A Inner Ear Brain Liver
Cochlea Brain Liver CD
120 120 120

8-oxodGuo/106 dGuo
AP Sites/105 bp DNA

(nmole/mg protein)
100 CD 100 CD 24 CD * CR
CR CR CR *

GSH:GSSG
80 80 80 80 * 80
60 60 16
* * 40 40 40
40 40
* 8
20 20
0 0 0
0 0 0 WT Sirt3-/- WT Sirt3-/- WT Sirt3-/-
WT Sirt3-/- WT Sirt3-/-
WT Sirt3-/-
Sirt3-/- WT
WT Sirt3 -/-
Sirt3-/- WT Sirt3 -/-

B Inner Ear Brain Liver


C Basal Region Middle Region Apical Region 0.6 CD 1.0 1.0

(nmole/mg protein)
CD CR
4000 CR 4000 4000 0.8 0.8
Neurons/mm2

* 0.4
*

GSSG
3000 * 3000 3000 0.5 0.5
2000 2000 2000 0.2
* 0.3 0.3
1000 1000 1000 0.0 0.0 0.0
0 0 0 WT
WT Sirt3-/-
Sirt3-/- WT
WT Sirt3-/-
Sirt3-/- WT Sirt3-/-
WT
WT Sirt3-/-
Sirt3-/- WT
WT Sirt3-/-
Sirt3-/- WT Sirt3-/-
C Inner Ear Brain Liver
D Basal Region Middle Region Apical Region 20 CD 20 40
(nmole/mg protein)
CD CR
100 100 100 * 15 15 30
CR
OH Cells (%)

75 * 75 75
GSH

10 10 20
50 50 50 5 5 10
25 25 25 0 0 0
0 0 0 WT Sirt3-/- WT Sirt3-/- WT Sirt3-/-
WT
WT Sirt3-/-
Sirt3-/- WT
WT Sirt3-/-
Sirt3-/- WT Sirt3-/-
Figure 3. Sirt3 Increases the Ratios Of GSH:GSSG in Mitochondria
E Basal Region Middle Region Apical Region during CR
100 CD 100 100 (A–C) Ratios of GSH:GSSG (A), GSSG (B), and GSH (C) were measured in the
CR *
* inner ear, brain (neocortex), and liver from control diet and calorie-restricted
IH Cells (%)

75 75 75
WT and Sirt3/ mice at 5 months of age (n = 4–5). *Significantly different
50 50 50 from 12- or 5-month-old WT mice (p < 0.05). Data are means ± SEM.
25 25 25
0 0 0 CR reduces oxidative damage in WT tissues, but not in the
WT Sirt3-/- WT Sirt3-/- WT Sirt3-/- Sirt3/ tissues. Thus, during CR, Sirt3 promotes a more reduc-
tive environment in mitochondria of multiple tissues, thereby
Figure 2. CR Reduces Oxidative DNA Damage and Increases Cell enhancing the glutathione antioxidant defense system.
Survival in the Cochleae from WT Mice, but Not from Sirt3/ Mice
(A) Oxidative damage to DNA (apurinic/apyrimidinic sites) was measured in the
Sirt3 Stimulates Idh2 Activity and Increases NADPH
cochlea and neocortex from control diet and calorie-restricted WT and Sirt3/
mice at 12 months of age (n = 4–5). AP sites, apurinic/apyrimidinic sites.
Levels in Mitochondria in Response to CR
*Significantly different from 12-month-old WT mice (p < 0.05). Enzymes of mitochondrial antioxidant pathways require NADPH
(B) Oxidative damage to DNA (8-oxodGuo) was measured in the liver from to perform their reductive functions. NADP+-dependent Idh2
control diet and calorie-restricted WT and Sirt3/ mice at 12 months of age from mitochondria converts NADP+ to NADPH, thereby pro-
(n = 4–5). moting regeneration of GSH by supplying NADPH to glutathione
(C) Neuron survival (neuron density) of basal, middle, and apical cochlear reductase (Jo et al., 2001). A previous in vitro study suggested
regions was measured from control diet and calorie-restricted WT and Sirt3/
that Idh2 might be a target of Sirt3, as incubation of Sirt3 with iso-
mice at 12 months of age (n = 4–5).
(D) OH (outer hair) cell survival (%) of basal, middle, and apical cochlear citrate dehydrogenase led to an apparent increase in dehydro-
regions was measured from control diet and calorie-restricted WT and Sirt3/ genase activity (Schlicker et al., 2008). Thus, we hypothesized
mice at 12 months of age (n = 4–5). that, in response to CR, the mitochondrial deacetylase Sirt3
(E) IH (inner hair) cell survival (%) of basal, middle, and apical cochlear regions might directly deacetylate and activate Idh2, thereby regulating
was measured from control diet and calorie-restricted WT and Sirt3/ mice at the levels of NADPH and, consequently, the glutathione antioxi-
12 months of age (n = 4–5).
dant defense system.
Data are means ± SEM. See also Figures 1B–1M.
To provide initial support for the hypothesis that Sirt3 regulates
Idh2 activity through deacetylation, we measured the acetylation
GSH:GSSG in mitochondria increased during CR in all of the levels of Idh2 in the liver mitochondria of WT and Sirt3/ mice
tested WT tissues (Figure 3A); however, CR failed to increase fed control and CR diets. In WT tissues, acetylation of Idh2
the ratios of GSH:GSSG in Sirt3/ tissues (Figure 3A). These was substantial in the control diet fed tissues, but CR induced
results are consistent with the histological, cochlear cell count- an 8-fold decrease in acetylation (Figures 4A and 4B). Robust
ing, and oxidative DNA damage results that demonstrated that acetylation of Idh2 was observed in Sirt3/ mice from both

Cell 143, 802–812, November 24, 2010 ª2010 Elsevier Inc. 805
A WT Sirt3-/- from the liver, inner ear, and brain of control diet and calorie-
CD CR CD CR restricted WT and Sirt3/ mice. We found that Idh2 activity
WB: -IDH2
significantly increased during CR in all of the WT tissues (Fig-
INPUT ure 4D); however, CR failed to increase Idh2 activity in the
WB: -Sirt3 Sirt3/ tissues (Figure 4D). If CR can induce a Sirt3-dependent
WB: -IDH2
increase in Idh2 activity, we anticipated increased levels of
IP: -IDH2 NADPH, providing the primary source of reducing equivalents
WB: -AcK for the glutathione antioxidant system (Jo et al., 2001; Schafer
and Buettner, 2001). To test this hypothesis, we measured
B C NADPH levels in mitochondria of WT and Sirt3/ mice. We
Acetylation Level (%)

200 CD WT
Relative Sirt3 Protein

CR * 4.0 found that levels of NADPH increased during CR in all tissues


Relative IDH2

150 * tested from WT mice (Figure 4E); however, no significant


3.0
changes in NADPH levels were observed between control diet
Level

100 2.0
50
and CR Sirt3/ tissues. Collectively, these results provide
1.0
* evidence that, during CR, Sirt3 induces the deacetylation and
0 0.0
activation of Idh2, leading to increased levels of NADPH in
WT
WT Sirt3-/-
Sirt3-/- CD CR
CD CR mitochondria of multiple tissues. We note that we observed
D Liver Inner Ear Brain a reduction in Idh2 activity in liver from Sirt3/ mice fed the
0.6 CD 0.6 CD 0.8 CD control diet and that this correlates with a slightly increased
(μM/s/μg protein)

CR * CR * CR
IDH2 Activity

0.6 level of acetylated Idh2 as compared to WT mice (Figure 4B).


0.4 0.4
* However, we did not observe reduced Idh2 activity or reduced
0.4
0.2 * 0.2 NADPH levels in the inner ear or brain of Sirt3/ mice. We
0.2
postulate that, under basal conditions (control diet fed), addi-
0.0 0.0 0.0 tional factors regulate mitochondrial Idh2 activity and NADPH
WT
WT Sirt3-/-
Sirt3-/-
WT
WT
Sirt3-/-
Sirt3-/- WT Sirt3-/- levels.
E Liver Inner Ear Brain To provide direct evidence that Sirt3 deacetylates Idh2,
1.0 1.0 CD 1.0 a number of biochemical experiments were performed.
*
NADPH/total NADP

CR
0.8 0.8 0.8 Although most enzyme:substrate reactions are necessarily
* * transient interactions to promote rapid turnover, coimmunopre-
0.5 0.5 0.5
cipitation (co-IP) experiments can sometimes trap these inter-
0.3 0.3 0.3 actions. Co-IP experiments were performed in human kidney
0.0 0.0 0.0 cells (HEK293) cotransfected with Sirt3 and Idh2. We found
WT Sirt3 -/- WT Sirt3 -/- WT Sirt3-/- that precipitated Idh2-FLAG was able to co-IP Sirt3-HA (Fig-
ure 5A), whereas precipitated Sirt3-FLAG was able to co-IP
Figure 4. Sirt3 Increases Idh2 Activity and NADPH Levels in Mito- Idh2-MYC (Figure 5B), suggesting that a physical interaction
chondria by Decreasing the Acetylation State of Idh2 during CR can occur between Sirt3 and Idh2 in human cells. However,
(A) (Top) Western blot analysis of Sirt3 and Idh2 levels in the liver from 5-month- co-IP experiments do not prove a direct functional interaction.
old WT or Sirt3/ fed either control or calorie-restricted diet. (Bottom) Endog- To provide support for a functional interaction between Sirt3
enous acetylated Idh2 was isolated by immunoprecipitation with anti-Idh2 and acetylated Idh2, deacetylation assays were carried out in
antibody followed by western blotting with anti-acetyl-lysine antibody (n = 3).
HEK293 cells (Figure 5C) and in vitro using purified components
(B and C) Quantification of the amounts of total Idh2 acetylation (B) and Sirt3
protein (C) from (A). Western blot was normalized with Idh2 levels or Sirt3 levels
(Figure 5D). Utilizing HEK293 cells, Idh2 was cotransfected with
quantified and analyzed by Image software (n = 3). or without Sirt3, isolated by immunoprecipitation with anti-MYC
(D) Idh2 activities were measured in the liver, inner ear (cochlea), and brain antibody followed by western blotting with anti-acetyl-lysine
(neocortex) from control diet and calorie-restricted WT and Sirt3/ mice at antibody. Coexpression with Sirt3 induced the deacetylation
5 months of age (n = 3–5). of Idh2 to background levels (Figure 5C). For the in vitro anal-
(E) Ratios of NADPH:total NADP (NADP+ + NADPH) were measured in the liver, ysis, acetylated Idh2 was prepared (see Figure S4 and Experi-
inner ear, and brain (neocortex) from control diet and caloric restricted WT and
mental Procedures) and utilized as a substrate for purified
Sirt3/ mice at 5 months of age (n = 3–5). *Significantly different from control
diet fed WT mice (p < 0.05). recombinant Sirt3 or Sirt5. Acetylation status was assessed
Data are means ± SEM. by western blotting with anti-acetyl-lysine antibody (Figure 5D),
and the resulting change in Idh2 activity was measured sepa-
rately (Figure 5E). We found that Sirt3, but not Sirt5, deacety-
control and CR diet-fed conditions, indicating that Sirt3 is lated IDH2 in an NAD+-dependent fashion (Figure 5E). The
required for the CR-induced deacetylation of Idh2 (Figures 4A corresponding Idh2 activity measurements indicated that de-
and 4B). As predicted, CR induced Sirt3 protein levels that acetylation by Sirt3, but not Sirt5, stimulated Idh2 activity
were approximately three times higher than those observed by 100% (Figure 5E). Together, these data provide strong
with control diet tissues in WT mice (Figure 4C). biochemical evidence that Sirt3 deacetylates and stimulates
To establish whether Idh2 activity is stimulated by Sirt3 under Idh2 activity and increases NADPH levels in mitochondria in
CR conditions, we measured Idh2 activity in the mitochondria response to CR.

806 Cell 143, 802–812, November 24, 2010 ª2010 Elsevier Inc.
A B Figure 5. Sirt3 Directly Deacetylates Idh2
Sirt3-HA + + + IDH2-MYC + + + and Stimulates Activity
IDH2-FLAG - + - Sirt3-FLAG - + - (A and B) Sirt3 interacts with Idh2. Idh2 or Sirt3
IP α-IgG α-FLAG IP α-IgG α-FLAG were immunoprecipitated from HEK293 cell
lysates with IgG antibody or FLAG beads. Precip-
INPUT WB: -HA INPUT WB: -MYC itated Idh2-FLAG was detected by anti-FLAG anti-
body, and co-IP Sirt3-HA was detected by anti-HA
WB: -FLAG WB: -FLAG
as indicated (A). Precipitated Sirt3-FLAG was
WB: -HA WB: -MYC detected by anti-FLAG antibody, and co-IP Idh2-
MYC was detected by anti-MYC as indicated (B)
(n = 3).
C D IDH2-FLAG + + + + + +
IDH2-MYC + + (C) Sirt3 deacetylates Idh2 in HEK293 cells. Idh2
NAD+ - + - + - + was cotransfected with or without Sirt3, isolated
Sirt3-FLAG - +
SIRTUIN - - Sirt3 Sirt3 Sirt5 Sirt5 by immunoprecipitation with anti-MYC antibody
WB: -MYC followed by western blotting with anti-acetyl-
IP: α-MYC WB: -FLAG
lysine antibody (n = 3).
IP: α-FLAG
WB: -AcK (D) Sirt3, but not Sirt5, deacetylates Idh2 in vitro.
WB: -AcK
INPUT WB: -FLAG
Acetylated Idh2 was prepared as outlined in the
COOMASSIE Experimental Procedures and was incubated
BLUE
with purified recombinant Sirt3 or Sirt5 with or
E without NAD+ at 37 C for 1 hr. Acetylation status
Relative IDH2 Activity (%)

250
200 * was assessed by western blotting with anti-
150 acetyl-lysine antibody (n = 3). An anti-FLAG
100 western shows that equivalent Idh2 protein levels
50 were used, and Coomassie staining shows puri-
0 fied Sirt3 and Sirt5.
IDH2+NAD

IDH2+Sirt3+NAD

IDH2+Sirt5+NAD
IDH2+Sirt3

IDH2+Sirt5
IDH2

(E) In vitro deacetylation of Idh2 by Sirt3, but not


Sirt5, stimulates Idh2 activity. Acetylated Idh2 in
buffer (Tris [pH 7.5], with or without 1 mM NAD,
and 1 mM DTT) was incubated with purified
50 nM Sirt3 or Sirt5 (Hallows et al., 2006) at
37 C for 1 hr, followed by Idh2 activity assay
(n = 3). *Significantly different from Idh2 alone
(p < 0.05).
Data are means ± SEM. See also Figure S4.

Overexpression of Sirt3 and/or Idh2 Increases NADPH stress by stimulating Idh2 activity and increasing NADPH levels
Levels and Protects Cells from Oxidative under stress conditions.
Stress-Induced Cell Death
Our physiological, histological, and biochemical results indicate DISCUSSION
that Sirt3 mediates reduction of oxidative damage by deacetyla-
tion and stimulating the activity of Idh2, which increases NADPH Sirt3 Reduces Oxidative Damage and Enhances
levels for antioxidant systems in mitochondria during CR. To the Glutathione Antioxidant Defense System under
provide support for this mechanism, we investigated whether CR Conditions
Sirt3 and Idh2 are sufficient to alter the NADPH levels in cultured A widely accepted hypothesis of how aging leads to age-related
cells. HEK293 cells stably transfected with vector, Sirt3, Idh2, or hearing loss is through the accumulation of oxidative damage in
Sirt3 with Idh2 were generated, and their NADPH levels were the inner ear (Someya and Prolla, 2010; Liu and Yan, 2007). In
measured. NADPH levels were significantly increased when support of this hypothesis, oxidative protein damage increases
either Idh2 or Sirt3 or both proteins were stably overexpressed in the cochlea of CBA/J mice (Jiang et al., 2007), and oxidative
in HEK293 cells (Figures 6A and 6B). Importantly, overexpres- DNA damage increases in the cochlea of C57BL/6J mice during
sion of both Sirt3 and Idh2 yielded a greater increase in NADPH aging (Someya et al., 2009). Age-related hair cell loss is also
levels than either Sirt3 or Idh2 overexpressed alone (Figure 6A). enhanced in mice lacking the antioxidant enzyme superoxide
Finally, to investigate whether overexpression of Sirt3, Idh2, or dismutase 1 (McFadden et al., 1999), whereas the same mutant
Sirt3 with Idh2 can protect cells from oxidative stress, the four animals show enhanced susceptibility to noise-induced hearing
HEK293 cell lines were treated with oxidants H2O2 (hydrogen loss (Ohlemiller et al., 1999). We have shown recently that over-
peroxide) (Figure 6C) or menadione (Figure 6D), and cell viability expression of mitochondrially targeted catalase delays the onset
was measured. Overexpression of Sirt3 or Idh2 was sufficient to of AHL in C57BL/6J mice, reduces hair cell loss, and reduces
protect cells from oxidative stress induced by both oxidants oxidative DNA damage in the inner ear (Someya et al., 2009).
(Figures 6C and 6D). Again, overexpression of both Sirt3 and Of interest, overexpression of catalase in the mitochondria leads
Idh2 led to higher cell viability than either Sirt3 or Idh2 overex- to extension of life span in C57BL/6J mice, but overexpression
pressed alone (Figures 6C and 6D). These results provide strong of catalase in the peroxisome or nucleus does not (Schriner
biochemical evidence that Sirt3 mediates reduction of oxidative et al., 2005). Under normal conditions, catalase decomposes

Cell 143, 802–812, November 24, 2010 ª2010 Elsevier Inc. 807
A B Figure 6. Overexpression of Sirt3 and/or
pBabe-IDH2-FLAG - - + + Idh2 Is Sufficient to Increase NADPH Levels
[NADPH] (nmol/mg protein)

180 **
* pCDNA3-Sirt3-FLAG - + - + and Protects HEK293 Cells from Oxidative
Stress
* INPUT
120 (A and B) (A) NADPH concentrations were sig-
* IDH2-FLAG nificantly increased when either Idh2 or Sirt3 or
WB: α-FLAG both were stably overexpressed in HEK293 cells.
60 Sirt3-FLAG
Measurements with errors are shown for the four
different stable cell populations from each type
0 of transfection (vector alone, Sirt3, Idh2, and
Sirt3 with Idh2) (n = 3). *Significantly different
Sirt3+IDH2
IDH2
VEC

Sirt3

from vector alone (p < 0.05); **Significantly dif-


ferent from Idh2 or Sirt3 (p < 0.05). (B) Western
blotting confirms Idh2 and Sirt3 stable expression.
(C and D) Sirt3 and/or Idh2 overexpression is suffi-
C D cient to protect HEK293 cells from the exogenous
120 120 oxidants hydrogen peroxide (H2O2) (C) and mena-
** ** dione (D). The four different stable cells were tran-
* *
Cell Viability (%)
Cell Viability (%)

90 *
90 * siently exposed to either 1 mM H2O2 or 25 mM
* menadione (n = 16).
60 * 60 Data are means ± SEM.

30 30

0 0
the glutathione antioxidant defense sys-
Sirt3+IDH2
Sirt3+IDH2

IDH2
IDH2
VEC
VEC

VEC
VEC
Sirt3

Sirt3

tem may play a central role in reduction


of oxidative stress in multiple tissues
1 mM 25 μM under CR conditions, leading to aging
H2O2 - + + + + Menadione - + + + + retardation. We also note that other mito-
chondrial effects of Sirt3, such as regula-
tion of fatty acid oxidation (Hirschey et al.,
2010) and modulation of complex I
hydrogen peroxide in the peroxisome, whereas in mitochondria, activity (Ahn, et al., 2008), are likely to contribute to the metabolic
hydrogen peroxide is decomposed into water by glutathione adaptations in response to CR.
peroxidase or peroxiredoxin (Finkel and Holbrook, 2000; Marı́
et al., 2009). Hence, these results suggest that mitochondrial Idh2 Regulates the Redox State of Mitochondria under
ROS play a critical role in cochlear aging, AHL, and aging in CR Conditions
general. A large body of evidence indicates that the antioxidant defense
We have demonstrated that Sirt3 mediates the CR reduction systems do not keep pace with the age-related increase in
of oxidative DNA damage in multiple tissues and that these ROS production, and thus the balance between antioxidant
effects are likely to arise through an enhanced mitochondrial defenses and ROS production shifts progressively toward
glutathione antioxidant defense system. As discussed earlier, a more pro-oxidant state during aging (Sohal and Weindruch,
the GSH:GSSG ratio is thought to be a marker of oxidative stress 1996; Rebrin and Sohal, 2008). This balance is determined in
(Rebrin and Sohal, 2008). Experimental evidence indicates that part by the ratios of interconvertible forms of redox couples,
aging results in a decrease in the ratio of GSH:GSSG in the mito- such as GSH/GSSG, NADPH/NADP+, NADH/NAD+, thioredox-
chondria of brain, liver, kidney, eye, heart, and testis from aged inred/thioredoxinoxid, and glutaredoxinred/glutaredoxinoxid. The
C57BL/6J mice due to elevated levels of GSSG, whereas CR GSH/GSSH couple is thought to be the primary cellular determi-
decreases the ratio of GSH:GSSG in the mitochondria of these nant of the cellular redox state because its abundance is three to
tissues by lowering GSSG levels (Rebrin et al., 2003, 2007). four orders of magnitude higher than the other redox couples
Our findings demonstrate that CR increases these ratios of (Rebrin and Sohal, 2008). NADPH is the reducing equivalent
GSH:GSSG in the mitochondria of brain, liver, and inner ear required for the regeneration of GSH and the GSH-mediated
from WT mice but fails to increase the ratios in the same tissues antioxidant defense system, which includes glutathione peroxi-
from Sirt3/ mice. Consistent with these results, CR reduced dases, glutathione transferases, and glutathione reductase,
oxidative DNA damage in tissues from WT mice but failed to playing a critical role in oxidative stress resistance (Halliwell
reduce such damage in tissues from Sirt3/ mice. CR also and Gutteridge, 2007). GSH is synthesized in the cytosol and
increased spiral ganglion neuron and hair cell survival in the transported into the mitochondria through protein channels in
WT cochlea, but not in Sirt3/ mice. Tissues that are composed the outer mitochondrial membrane (Halliwell and Gutteridge,
of postmitotic cells such as the brain and the inner ear are partic- 2007; Anderson, 1998). Although GSH can cross the outer mito-
ularly vulnerable to oxidative damage because of their high chondrial membrane through these channels, GSSG cannot be
energy requirements and inability to undergo regeneration. exported into the cytosol (Olafsdottir and Reed, 1988). Thus,
Therefore, we speculate that the Sirt3-mediated modulation of GSSG is reduced to GSH by mitochondrial NADPH-dependent

808 Cell 143, 802–812, November 24, 2010 ª2010 Elsevier Inc.
glutathione reductase, preventing accumulation of GSSG in the
mitochondrial matrix (Schafer and Buettner, 2001; Marı́ et al.,
2009). We have demonstrated that Sirt3 directly deacetylates
and activates Idh2 under CR conditions. In response to CR,
deacetylated Idh2 displays increased catalytic activity, which is
correlated with increased NADPH levels in the mitochondria of
multiple tissues from WT mice, but not from Sirt3/ mice.
Hence, we speculate that Idh2 may be a major player in regu-
lating the redox state of mitochondria under CR conditions given
its role in mitochondrial NADPH production. A previous study
has shown that Idh2 is induced in response to ROS in mouse
fibroblasts, whereas decreased levels of Idh2 lead to higher
ROS and accumulation of oxidative damage to DNA and lipids
(Jo et al., 2001). Our in vitro findings demonstrate that overex-
pression of Sirt3 and/or Idh2 increases NADPH levels and
protects cells from oxidative stress-induced cell death. Thus,
these observations underlie a critical role for Idh2 in the genera-
tion of NADPH in mitochondria under conditions of CR, providing
reducing capacity for the glutathione antioxidant system and
increasing oxidative stress resistance.

A Role for Sirt3 in CR-Mediated Prevention of AHL


The mouse is considered a good model for the study of human
AHL because the mouse cochlea is anatomically similar to that Figure 7. A Model for the CR-Mediated Prevention of AHL in
of humans (Steel et al., 1996; Steel and Bock, 1983). Most in- Mammals
In response to CR, SIRT3 activates IDH2, thereby increasing NADPH levels in
bred mouse strains display some degree of AHL, and the age
mitochondria. This in turn leads to an increased ratio of GSH:GSSG and
of onset of AHL is known to vary from 3 months in DBA/2J
decreased levels of ROS, thereby resulting in protection from oxidative stress
mice to more than 20 months in CBA/CaJ mice (Zheng et al., and prevention of AHL in mammals.
1999). The C57BL/6J mouse strain, which is the most widely
used mouse model for the study of aging, displays the classic cular disease, diabetes, high blood pressure, hypertension,
pattern of AHL by 12–15 months of age (Hunter and Willott, and certain cancers (Paeratakul et al., 2002; Poirier et al.,
1987; Keithley et al., 2004). We have previously shown that 2006). Obesity is also associated with an increased risk of
AHL in C57BL/6J mice occurs through Bak-mediated apoptosis mortality (Poirier et al., 2006; Lee et al., 1993). Of interest, CR
and that it can be prevented by the intake of small molecule anti- failed to reduce oxidative damage in multiple tissues and slow
oxidants (Someya et al., 2009). We note that C57BL/6J and many the progression of AHL in CR Sirt3/ mice, despite the fact
other mouse strains carry a specific mutation (Cdh23753A) in the that these mice were lean (Figures S2A and S2B). Thus, these
Cdh23 gene, which encodes a component of the hair cell tip results suggest that weight loss may not be sufficient for the
link, and this mutation is known to promote early onset of AHL anti-aging action of CR. Instead, we postulate that critical meta-
in these animals (Noben-Trauth et al., 2003). Of interest, the bolic effectors such as Sirt3 mediate the positive effects of CR.
Cdh23753A allele may increase the susceptibility to oxidative
stress in hair cells because a Sod1 mutation greatly enhances
Conclusions
AHL in mice carrying Cdh23753A, but not in mice wild-type for In summary, we propose that, in response to CR, Sirt3 activates
Cdh23 (Johnson, et al., 2010). However, oxidative damage Idh2, thereby increasing NADPH levels in mitochondria. This in
increases with age in the cochlea of both C57BL/6J mice and
turn leads to increased ratios of GSH:GSSG in mitochondria
the CBA/J mouse strain that does not carry the Cdh23753A allele, and decreased levels of ROS, resulting in protection of inner ear
indicating that oxidative stress plays a role in AHL independent cells and prevention of AHL in mammals (Figure 7). Because we
of Cdh23 (Someya et al., 2009; Jiang et al., 2007; Zheng et al.,
observed similar effects of CR in the mitochondrial GSH/GSSG
1999). In both strains, the loss of hair cells and spiral ganglion ratios in multiple tissues, we postulate that this may be a major
neurons begins in the base of the cochlea and spreads toward mechanism of aging retardation by CR. We also postulate that
the apex with age (Keithley et al., 2004; Hunter and Willott,
pharmaceutical interventions that induce Sirt3 activity in multiple
1987). Importantly, CR slows the progression of AHL in both
tissues will mimic CR by increasing oxidative stress resistance
C57BL/6J and CBA/J strains (Someya et al., 2007; Sweet and preventing the mitochondrial decay associated with aging.
et al., 1988). Therefore, the protective effects of Sirt3 in AHL
are likely to be of general relevance to AHL.
EXPERIMENTAL PROCEDURES
It is thought that some of the effects of CR in aging retardation
require significant reduction of body weight through reducing Animals
food consumption. In agreement with this hypothesis, obesity Male and female Sirt3+/ mice were purchased from the Mutant Mouse
promotes a variety of age-related diseases, such as cardiovas- Resource Centers (MMRRC) at the University of North Carolina-Chapel Hill

Cell 143, 802–812, November 24, 2010 ª2010 Elsevier Inc. 809
(Chapel Hill, NC). In brief, these mice were created by generating embryonic Idh2 Activity
stem (ES) cells (Omni bank number OST341297) bearing a retroviral promoter Activities of Idh2 were measured by the Kornberg method (Kornberg, 1955). In
trap that functionally inactivates one allele of the Sirt3 gene (MGI, 2010). brief, 20 ml of the mitochondrial lysate sample was added in each well of a
Male and female C57BL/6J mice were purchased from Jackson Laboratory 96-well plate, and then 180 ml of a reaction mixture (33 mM KH2PO4dK2HPO4,
(Bar Harbor, ME). Sirt3+/ mice have been backcrossed for four generations 3.3 mM MgCl2, 167 mM NADP+, and 167 mM (+)-potassium Ds-threo-isocitrate
onto the C57BL/6J background. All animal studies were conducted at the monobasic) was added in each well. The absorbance was immediately read at
AAALAC-approved Animal Facility in the Genetics and Biotechnology Center 340 nm every 10 s for 1 min in a microplate reader (Bio-Rad, Hercules, CA). All
of the University of Wisconsin-Madison. Experiments were performed in samples were run in duplicate. The reaction rates were calculated, and the
accordance with protocols approved by the University of Wisconsin-Madison Idh2 activity in the sample was defined as the production of one mmole of
Institutional Animal Care and Use Committee (Madison, WI). NADPH per sec.

Dietary Study In Vitro Deacetylation Assay


Details on the methods used to house and feed mice have been described Idh2-FLAG was transfected into HEK293 cells, which were then treated with
previously (Pugh et al., 1999). Mice are housed individually. Control diet (CD) 5 mM nicotinamide for 16 hr. Nicotinamide is a widely used sirtuin inhibitor.
groups were fed 86.4 kcal/week of the precision pellet diet AIN-93M (BioServ, Nicotinamide treatment leads to increased acetylation of Idh2, with a corre-
Frenchtown, NJ), and caloric-restricted (CR) groups were fed 64.8 kcal/week sponding decrease in enzymatic activity (Figure S4). Idh2 from cell lysates
(a 25% CR) of the precision pellet diet AIN-93M 40%DR (BioServ, Frenchtown, was immunoprecipitated with anti-FLAG beads at 4 C for 2 hr, and then
NJ). The schedule of feeding for control diet was 7 g on Mondays and Wednes- Idh2-FLAG on beads was utilized in 200 ul deacetylation buffer (Tris
days and 10 g on Fridays, whereas the schedule of feeding for calorie- [pH 7.5], with or without 1 mM NAD, and 1 mM DTT) and incubated with puri-
restricted diets was 5 g on Mondays and Wednesdays and 8 g on Fridays. fied 50 nM Sirt3 or Sirt5 (Hallows et al., 2006) at 37 C for 1 hr. Aliquots were
This dietary regimen was maintained from 2 months of age until 5 months of removed for Idh2 activity assay and western blotting with anti-FLAG antibody
age for a 3 month CR study and from 2 months of age until 12 months of or anti-acetyl-lysine antibody.
age for a 10 month CR study.
Statistical Analysis
ABR Hearing Test All Statistical analyses were carried out by one-way ANOVA with post-Tukey
At 12 months of age, ABRs were measured with a tone burst stimulus at 8, 16, multiple comparison tests using the Prism 4.0 statistical analysis program
and 32 kHz using an ABR recording system (Intelligent Hearing System, Miami, (GraphPad, San Diego, CA). All tests were two-sided with statistical signifi-
FL) as previously described (Someya et al., 2009). Mice were anesthetized cance set at p < 0.05.
with a mixture of xylazine hydrochloride (10 mg/kg, i.m.) (Phoenix Urology of
St. Joseph, St. Joseph, MO) and ketamine hydrochloride (40 mg/kg, i.m.)
SUPPLEMENTAL INFORMATION
(Phoenix Urology of St. Joseph).

Supplemental Information includes Extended Experimental Procedures, four


Measurement of DNA Oxidation Levels
figures, and one table and can be found at doi:10.1016/j.cell.2010.10.002.
At 12 months of age, cochlea and neocortex were collected, and DNA was
extracted with ethanol precipitation. DNA concentrations for each sample
were adjusted to 0.1 mg/ml, and numbers of apurinic/apyrimidinic (AP) sites ACKNOWLEDGMENTS
were determined using the DNA Damage Quantification Kit (Dojindo, Rockville,
MD) and performed according to the manufacturer’s instructions and as previ- We thank S. Kinoshita for histological processing. This research was sup-
ously described (Kubo et al., 1992; Meira, et al., 2009; McNeill and Wilson, ported by NIH grants AG021905 (T.A.P.) and GM065386 (J.M.D.), the National
2007). Liver was also collected from the same mice, and 8-hydroxyguanosine Projects on Protein Structural and Functional Analyses from the Ministry of
levels (8-oxo-7,8-20 -deoxyguanosine/106 deoxyguanosine) in the DNA were Education, Culture, Sports, Science, and Technologies of Japan, and Marine
determined using a HPLC-ECD method as previously described (Hofer Bio Foundation.
et al., 2006).
Received: July 19, 2010
Measurement of Total GSH and GSSG Revised: September 3, 2010
Just after mitochondrial lysate preparation, 100 ml of the lysate was mixed with Accepted: September 30, 2010
100 ml of 10% metaphosphoric acid, incubated for 30 min at 4 C, and centri- Published online: November 18, 2010
fuged at 14,000 3 g for 10 min at 4 C. The supernatant was used for the
measurements of mitochondrial glutathione contents. Total glutathione REFERENCES
(GSH + GSSG) and GSSG levels were determined by the method of Rahman
et al. (2006). All samples were run in duplicate. The rates of 2-nitro-5-thioben- Ahn, B.H., Kim, H.S., Song, S., Lee, I.H., Liu, J., Vassilopoulos, A., Deng, C.X.,
zoic acid formation were calculated, and the total glutathione (tGSH) and and Finkel, T. (2008). A role for the mitochondrial deacetylase Sirt3 in regu-
GSSG concentrations in the samples were determined by using linear regres- lating energy homeostasis. Proc. Natl. Acad. Sci. USA 105, 14447–14452.
sion to calculate the values obtained from the standard curve. The GSH Anderson, M.E. (1998). Glutathione: an overview of biosynthesis and modula-
concentration was determined by subtracting the GSSG concentration from tion. Chem. Biol. Interact. 111-112, 1–14.
the tGSH concentration. Balaban, R.S., Nemoto, S., and Finkel, T. (2005). Mitochondria, oxidants, and
aging. Cell 120, 483–495.
Idh2 Acetylation Analysis
Barger, J.L., Kayo, T., Vann, J.M., Arias, E.B., Wang, J., Hacker, T.A., Wang,
Antibodies used for western blotting included anti-Idh2 antibody (Santa Cruz,
Y., Raederstorff, D., Morrow, J.D., Leeuwenburgh, C., et al. (2008). A low
Santa Cruz, CA), anti-Sirt3 antibody (gift of Dr. Eric Verdin, UCSF), protein A/G
dose of dietary resveratrol partially mimics caloric restriction and retards aging
plus agarose (Santa Cruz, Santa Cruz, CA), and pan-acetylated lysine (gener-
parameters in mice. PLoS ONE 3, e2264.
ated following the procedure of Zhao, et al. [2010], GeneTel Laboratories LLC,
Madison, WI). For immunoprecipitation, liver mitochondria lysates were incu- Bordone, L., Cohen, D., Robinson, A., Motta, M.C., van Veen, E., Czopik, A.,
bated with anti-Idh2 antibody overnight at 4 C. Then protein A/G plus agarose Steele, A.D., Crowe, H., Marmor, S., Luo, J., et al. (2007). SIRT1 transgenic
were added and incubated for 3 hr. After resins were washed, samples were mice show phenotypes resembling calorie restriction. Aging Cell 6, 759–767.
boiled with SDS loading buffer and subjected to western blotting (Smith Chen, D., Steele, A.D., Lindquist, S., and Guarente, L. (2005). Increase in
et al., 2009). activity during calorie restriction requires Sirt1. Science 310, 1641.

810 Cell 143, 802–812, November 24, 2010 ª2010 Elsevier Inc.
Chen, D., Bruno, J., Easlon, E., Lin, S.J., Cheng, H.L., Alt, F.W., and Guarente, Marı́, M., Morales, A., Colell, A., Garcı́a-Ruiz, C., and Fernández-Checa, J.C.
L. (2008). Tissue-specific regulation of SIRT1 by calorie restriction. Genes Dev. (2009). Mitochondrial glutathione, a key survival antioxidant. Antioxid. Redox
22, 1753–1757. Signal. 11, 2685–2700.
Cohen, H.Y., Miller, C., Bitterman, K.J., Wall, N.R., Hekking, B., Kessler, B., Masoro, E.J. (2000). Caloric restriction and aging: an update. Exp. Gerontol.
Howitz, K.T., Gorospe, M., de Cabo, R., and Sinclair, D.A. (2004). Calorie 35, 299–305.
restriction promotes mammalian cell survival by inducing the SIRT1 deacety-
Mattson, M.P. (2000). Apoptosis in neurodegenerative disorders. Nat. Rev.
lase. Science 305, 390–392.
Mol. Cell Biol. 1, 120–129.
Colman, R.J., Anderson, R.M., Johnson, S.C., Kastman, E.K., Kosmatka, K.J.,
McFadden, S.L., Ding, D., Reaume, A.G., Flood, D.G., and Salvi, R.J. (1999).
Beasley, T.M., Allison, D.B., Cruzen, C., Simmons, H.A., Kemnitz, J.W., and
Age-related cochlear hair cell loss is enhanced in mice lacking copper/zinc
Weindruch, R. (2009). Caloric restriction delays disease onset and mortality
superoxide dismutase. Neurobiol. Aging 20, 1–8.
in rhesus monkeys. Science 325, 201–204.
McNeill, D.R., and Wilson, D.M.I.I.I., III. (2007). A dominant-negative form of
Donmez, G., and Guarente, L. (2010). Aging and disease: connections to
the major human abasic endonuclease enhances cellular sensitivity to labora-
sirtuins. Aging Cell 9, 285–290.
tory and clinical DNA-damaging agents. Mol. Cancer Res. 5, 61–70.
Finkel, T., and Holbrook, N.J. (2000). Oxidants, oxidative stress and the
biology of ageing. Nature 408, 239–247. Meira, L.B., Moroski-Erkul, C.A., Green, S.L., Calvo, J.A., Bronson, R.T., Shah,
D., and Samson, L.D. (2009). Aag-initiated base excision repair drives alkyl-
Finkel, T., Deng, C.X., and Mostoslavsky, R. (2009). Recent progress in the
ation-induced retinal degeneration in mice. Proc. Natl. Acad. Sci. USA 106,
biology and physiology of sirtuins. Nature 460, 587–591.
888–893.
Halliwell, B., and Gutteridge, J.M.C. (2007). Free Radicals in Biology and Medi-
MGI. Sirt3Gt(OST341297)Lex. (2010). Available at http://www.informatics.jax.org/
cine (New York, NY: Oxford University Press).
searches/accession_report.cgi?id=MGI:3529767.
Hallows, W.C., Lee, S., and Denu, J.M. (2006). Sirtuins deacetylate and acti-
Noben-Trauth, K., Zheng, Q.Y., and Johnson, K.R. (2003). Association of cad-
vate mammalian acetyl-CoA synthetases. Proc. Natl. Acad. Sci. USA 103,
herin 23 with polygenic inheritance and genetic modification of sensorineural
10230–10235.
hearing loss. Nat. Genet. 35, 21–23.
Hamilton, M.L., Van Remmen, H., Drake, J.A., Yang, H., Guo, Z.M., Kewitt, K.,
Ohlemiller, K.K., McFadden, S.L., Ding, D.L., Flood, D.G., Reaume, A.G., Hoff-
Walter, C.A., and Richardson, A. (2001). Does oxidative damage to DNA
man, E.K., Scott, R.W., Wright, J.S., Putcha, G.V., and Salvi, R.J. (1999). Tar-
increase with age? Proc. Natl. Acad. Sci. USA 98, 10469–10474.
geted deletion of the cytosolic Cu/Zn-superoxide dismutase gene (Sod1)
Hirschey, M.D., Shimazu, T., Goetzman, E., Jing, E., Schwer, B., Lombard,
increases susceptibility to noise-induced hearing loss. Audiol. Neurootol. 4,
D.B., Grueter, C.A., Harris, C., Biddinger, S., Ilkayeva, O.R., et al. (2010).
237–246.
SIRT3 regulates mitochondrial fatty-acid oxidation by reversible enzyme
deacetylation. Nature 464, 121–125. Olafsdottir, K., and Reed, D.J. (1988). Retention of oxidized glutathione by iso-
lated rat liver mitochondria during hydroperoxide treatment. Biochim. Biophys.
Hofer, T., Seo, A.Y., Prudencio, M., and Leeuwenburgh, C. (2006). A method to
Acta 964, 377–382.
determine RNA and DNA oxidation simultaneously by HPLC-ECD: greater
RNA than DNA oxidation in rat liver after doxorubicin administration. Biol. Paeratakul, S., Lovejoy, J.C., Ryan, D.H., and Bray, G.A. (2002). The relation of
Chem. 387, 103–111. gender, race and socioeconomic status to obesity and obesity comorbidities
in a sample of US adults. Int. J. Obes. Relat. Metab. Disord. 26, 1205–1210.
Hunter, K.P., and Willott, J.F. (1987). Aging and the auditory brainstem
response in mice with severe or minimal presbycusis. Hear. Res. 30, 207–218. Poirier, P., Giles, T.D., Bray, G.A., Hong, Y., Stern, J.S., Pi-Sunyer, F.X., and
Eckel, R.H. (2006). Obesity and cardiovascular disease: pathophysiology,
Jiang, H., Talaska, A.E., Schacht, J., and Sha, S.H. (2007). Oxidative imbal-
evaluation, and effect of weight loss. Arterioscler. Thromb. Vasc. Biol. 26,
ance in the aging inner ear. Neurobiol. Aging 28, 1605–1612.
968–976.
Jo, S.H., Son, M.K., Koh, H.J., Lee, S.M., Song, I.H., Kim, Y.O., Lee, Y.S.,
Jeong, K.S., Kim, W.B., Park, J.W., et al. (2001). Control of mitochondrial redox Pugh, T.D., Klopp, R.G., and Weindruch, R. (1999). Controlling caloric
balance and cellular defense against oxidative damage by mitochondrial consumption: protocols for rodents and rhesus monkeys. Neurobiol. Aging
NADP+-dependent isocitrate dehydrogenase. J. Biol. Chem. 276, 16168– 20, 157–165.
16176. Rahman, I., Kode, A., and Biswas, S.K. (2006). Assay for quantitative determi-
Johnson, K.R., Yu, H., Ding, D., Jiang, H., Gagnon, L.H., and Salvi, R.J. (2010). nation of glutathione and glutathione disulfide levels using enzymatic recycling
Separate and combined effects of Sod1 and Cdh23 mutations on age-related method. Nat. Protoc. 1, 3159–3165.
hearing loss and cochlear pathology in C57BL/6J mice. Hear. Res. 268, 85–92. Rebrin, I., and Sohal, R.S. (2008). Pro-oxidant shift in glutathione redox state
Keithley, E.M., Canto, C., Zheng, Q.Y., Fischel-Ghodsian, N., and Johnson, during aging. Adv. Drug Deliv. Rev. 60, 1545–1552.
K.R. (2004). Age-related hearing loss and the ahl locus in mice. Hear. Res. Rebrin, I., Kamzalov, S., and Sohal, R.S. (2003). Effects of age and caloric
188, 21–28. restriction on glutathione redox state in mice. Free Radic. Biol. Med. 35,
Kornberg, A. (1955). Isocitric dehydrogenase of yeast (TPN). Methods Enzy- 626–635.
mol. 1, 705–707. Rebrin, I., Forster, M.J., and Sohal, R.S. (2007). Effects of age and caloric
Koubova, J., and Guarente, L. (2003). How does calorie restriction work? intake on glutathione redox state in different brain regions of C57BL/6 and
Genes Dev. 17, 313–321. DBA/2 mice. Brain Res. 1127, 10–18.
Kubo, K., Ide, H., Wallace, S.S., and Kow, Y.W. (1992). A novel, sensitive, and Schafer, F.Q., and Buettner, G.R. (2001). Redox environment of the cell as
specific assay for abasic sites, the most commonly produced DNA lesion. viewed through the redox state of the glutathione disulfide/glutathione couple.
Biochemistry 31, 3703–3708. Free Radic. Biol. Med. 30, 1191–1212.
Lee, I.M., Manson, J.E., Hennekens, C.H., and Paffenbarger, R.S., Jr. (1993). Schlicker, C., Gertz, M., Papatheodorou, P., Kachholz, B., Becker, C.F., and
Body weight and mortality. A 27-year follow-up of middle-aged men. JAMA Steegborn, C. (2008). Substrates and regulation mechanisms for the human
270, 2823–2828. mitochondrial sirtuins Sirt3 and Sirt5. J. Mol. Biol. 382, 790–801.
Lin, S.J., Defossez, P.A., and Guarente, L. (2000). Requirement of NAD and Schriner, S.E., Linford, N.J., Martin, G.M., Treuting, P., Ogburn, C.E., Emond,
SIR2 for life-span extension by calorie restriction in Saccharomyces cerevi- M., Coskun, P.E., Ladiges, W., Wolf, N., Van Remmen, H., et al. (2005). Exten-
siae. Science 289, 2126–2128. sion of murine life span by overexpression of catalase targeted to mitochon-
Liu, X.Z., and Yan, D. (2007). Ageing and hearing loss. J. Pathol. 211, 188–197. dria. Science 308, 1909–1911.

Cell 143, 802–812, November 24, 2010 ª2010 Elsevier Inc. 811
Schwer, B., Eckersdorff, M., Li, Y., Silva, J.C., Fermin, D., Kurtev, M.V., Gial- Sundaresan, N.R., Samant, S.A., Pillai, V.B., Rajamohan, S.B., and Gupta,
lourakis, C., Comb, M.J., Alt, F.W., and Lombard, D.B. (2009). Calorie restric- M.P. (2008). SIRT3 is a stress-responsive deacetylase in cardiomyocytes
tion alters mitochondrial protein acetylation. Aging Cell 8, 604–606. that protects cells from stress-mediated cell death by deacetylation of Ku70.
Mol. Cell. Biol. 28, 6384–6401.
Smith, B.C., Hallows, W.C., and Denu, J.M. (2009). A continuous microplate
assay for sirtuins and nicotinamide-producing enzymes. Anal. Biochem. 394, Sundaresan, N.R., Gupta, M., Kim, G., Rajamohan, S.B., Isbatan, A., and
101–109. Gupta, M.P. (2009). Sirt3 blocks the cardiac hypertrophic response by aug-
menting Foxo3a-dependent antioxidant defense mechanisms in mice.
Sohal, R.S., and Weindruch, R. (1996). Oxidative stress, caloric restriction, and
J. Clin. Invest. 119, 2758–2771.
aging. Science 273, 59–63.
Sweet, R.J., Price, J.M., and Henry, K.R. (1988). Dietary restriction and pres-
Someya, S., Yamasoba, T., Weindruch, R., Prolla, T.A., and Tanokura, M. byacusis: periods of restriction and auditory threshold losses in the CBA/J
(2007). Caloric restriction suppresses apoptotic cell death in the mammalian mouse. Audiology 27, 305–312.
cochlea and leads to prevention of presbycusis. Neurobiol. Aging 28, 1613–
Wallace, D.C. (2005). A mitochondrial paradigm of metabolic and degenerative
1622.
diseases, aging, and cancer: a dawn for evolutionary medicine. Annu. Rev.
Someya, S., Xu, J., Kondo, K., Ding, D., Salvi, R.J., Yamasoba, T., Rabinovitch, Genet. 39, 359–407.
P.S., Weindruch, R., Leeuwenburgh, C., Tanokura, M., and Prolla, T.A. (2009).
Weindruch, R., and Walford, R.L. (1988). The Retardation of Aging and Disease
Age-related hearing loss in C57BL/6J mice is mediated by Bak-dependent
by Dietary Restriction (Springfield, IL: Charles C Thomas Publishing, LTD).
mitochondrial apoptosis. Proc. Natl. Acad. Sci. USA 106, 19432–19437.
Zhao, S., Xu, W., Jiang, W., Yu, W., Lin, Y., Zhang, T., Yao, J., Zhou, L., Zeng,
Someya, S., and Prolla, T.A. (2010). Mitochondrial oxidative damage and Y., Li, H., et al. (2010). Regulation of cellular metabolism by protein lysine acet-
apoptosis in age-related hearing loss. Mech. Ageing Dev. 131, 480–486. ylation. Science 327, 1000–1004.
Steel, K.P., and Bock, G.R. (1983). Hereditary inner-ear abnormalities in Zheng, Q.Y., Johnson, K.R., and Erway, L.C. (1999). Assessment of hearing in
animals. Relationships with human abnormalities. Arch. Otolaryngol. 109, 80 inbred strains of mice by ABR threshold analyses. Hear. Res. 130, 94–107.
22–29.
Zhu, H., Guo, Q., and Mattson, M.P. (1999). Dietary restriction protects hippo-
Steel, K.P., Moorjani, P., and Bock, G.R. (1996). Mixed conductive and senso- campal neurons against the death-promoting action of a presenilin-1 mutation.
rineural hearing loss in LP/J mice. Hear. Res. 28, 227–236. Brain Res. 842, 224–229.

812 Cell 143, 802–812, November 24, 2010 ª2010 Elsevier Inc.
FOXO/4E-BP Signaling in Drosophila
Muscles Regulates Organism-wide
Proteostasis during Aging
Fabio Demontis1,* and Norbert Perrimon1,2,*
1Department of Genetics
2Howard Hughes Medical Institute
Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
*Correspondence: fdemontis@genetics.med.harvard.edu (F.D.), perrimon@receptor.med.harvard.edu (N.P.)
DOI 10.1016/j.cell.2010.10.007

SUMMARY body extends life span, indicating a key role of this tissue in
the regulation of longevity (Giannakou et al., 2004; Hwangbo
The progressive loss of muscle strength during aging et al., 2004). In addition, because most tissues undergo progres-
is a common degenerative event of unclear pathogen- sive deterioration during aging (Garigan et al., 2002), it is thought
esis. Although muscle functional decline precedes that organismal life span may be linked to tissue senescence.
age-related changes in other tissues, its contribution However, our understanding of the mechanisms regulating
to systemic aging is unknown. Here, we show that tissue aging and their interconnection to life span is limited. For
example, analysis in Drosophila has revealed that the prevention
muscle aging is characterized in Drosophila by the
of age-dependent changes in cardiac performance does not
progressive accumulation of protein aggregates
alter life span (Wessells et al., 2004), raising the possibility that
that associate with impaired muscle function. The functional decline in distinct tissues may have different
transcription factor FOXO and its target 4E-BP re- outcomes on the systemic regulation of aging.
move damaged proteins at least in part via the The Insulin/IGF-1 signaling pathway has been implicated in the
autophagy/lysosome system, whereas foxo mutants control of aging across evolution via its downstream signaling
have dysfunctional proteostasis. Both FOXO and component FOXO (DAF-16 in C. elegans), a member of the
4E-BP delay muscle functional decay and extend life fork-head box O transcription factor family (Salih and Brunet,
span. Moreover, FOXO/4E-BP signaling in muscles 2008). FOXO regulates the expression of a series of target genes
decreases feeding behavior and the release of insulin involved in metabolism, cell growth, cell proliferation, stress
from producing cells, which in turn delays the age- resistance, and differentiation via direct binding to target gene
promoter regions (Salih and Brunet, 2008). Mutations in foxo/
related accumulation of protein aggregates in other
daf-16 reduce life span and stress resistance in both C. elegans
tissues. These findings reveal an organism-wide and flies, indicating a key role in organism aging (Junger et al.,
regulation of proteostasis in response to muscle 2003; Salih and Brunet, 2008). In addition to regulating life
aging and a key role of FOXO/4E-BP signaling in the span, FOXO has been reported to prevent the pathogenesis of
coordination of organismal and tissue aging. some age-related diseases. For example, FOXO reduces the
toxicity associated with aggregation-prone human mutant
INTRODUCTION Alzheimer’s and Huntington’s disease proteins (proteotoxicity)
in C. elegans and mice, suggesting that regulating protein
Aging of multicellular organisms involves distinct pathogenic homeostasis (proteostasis) during aging may have a direct effect
events that include higher mortality, the progressive loss of on the pathogenesis of human neurodegenerative diseases
organ function, and susceptibility to degenerative diseases, (Cohen et al., 2006; Hsu et al., 2003; Morley et al., 2002).
some of which arise from protein misfolding and aggregation. However, little is known on the protective mechanisms induced
Recent genetic studies in the mouse, the nematode Caenorhab- in response to FOXO signaling and whether they vary in different
ditis elegans, and the fruitfly Drosophila melanogaster have aging tissues and disease contexts.
expanded our understanding of the evolutionarily conserved Among the plethora of age-related pathological conditions,
signaling pathways regulating aging, with the identification of the gradual decay in muscle strength is one of the first hallmarks
several mutants that have prolonged or shortened life spans of aging in many organisms, including Drosophila, C. elegans,
(Kenyon, 2005). Manipulation of longevity-regulating pathways mice and, importantly, humans (Augustin and Partridge, 2009;
in certain tissues is sufficient to extend life expectancy, indi- Herndon et al., 2002; Nair, 2005; Zheng et al., 2005). However,
cating that some tissues have a predominant role in life span despite its medical relevance, the mechanisms underlying
extension (Libina et al., 2003; Wang et al., 2005; Wolkow et al., muscle aging are incompletely understood. Functional changes
2000). For example, foxo overexpression in the Drosophila fat in skeletal muscles temporally precede the manifestation of

Cell 143, 813–825, November 24, 2010 ª2010 Elsevier Inc. 813
Figure 1. FOXO Signaling in Skeletal Muscles Preserves Proteostasis during Aging
(A–D) Electron micrographs of immunogold-labeled Drosophila skeletal muscles of wild-type flies at one (A and B) and 5 weeks of age (C and D). Protein
aggregates (PA) are detected in the cytoplasm in proximity to mitochondria (Mt) and myofibrils (Myof) in old (C and D) but not young flies (A and B). Numerous
gold particles (indicative of anti-ubiquitin immunoreactivity) localize to filamentous structures at 5 weeks of age (C and D), while only a few are present in muscles
from young flies. Scale bars are 1 mm (A and C) and 500 nm (B and D).

814 Cell 143, 813–825, November 24, 2010 ª2010 Elsevier Inc.
aging in other tissues (Herndon et al., 2002), and reduced muscle number of gold particles (Figure 1E). To test the hypothesis
strength is associated with an increased risk in developing that muscle function during aging may decrease due to defects
Alzheimer’s and Parkinson’s diseases (Boyle et al., 2009; Chen in protein homeostasis, we better characterized the age-related
et al., 2005). However, although aging-related changes in deposition of protein aggregates by immunofluorescence. In
skeletal muscles have been proposed to affect physiological agreement with the IEM analysis (Figures 1A–1E), we observed
processes in distal organs (Nair, 2005), whether or not muscle that aging skeletal muscles progressively accumulate aggre-
senescence modulates the pathogenesis of degenerative events gates of polyubiquitinated proteins (ranging up to several mm)
in other tissues is unknown. that colocalize with p62/Ref(2)P, an inclusion body component
The fruit fly Drosophila is an excellent model to study muscle (Figures 1F and1I). The cumulative area of protein aggregates
aging. The progressive decline in muscle strength and function increases during aging (Figure 1L), suggesting that the progres-
observed in humans is recapitulated in this system (Rhodenizer sive protein damage, together with a decrease in the turnover of
et al., 2008), which is amenable to extensive genetic manipula- muscle proteins, may result in the age-related decline of muscle
tion. By using this model organism, we have searched for the strength.
molecular mechanisms responsible for muscle aging and To better characterize how protein quality control is linked with
found that decreased protein quality control plays a role in the aging in muscles, we analyzed the deposition of protein
pathogenesis of age-related muscle weakness. Interestingly, aggregates in syngenic flies with foxo overexpression. Foxo
increased activity of the transcription factor FOXO and its target overexpression results in its activation (Giannakou et al., 2004;
Thor/4E-BP are sufficient to delay this process and preserve Hwangbo et al., 2004) and was achieved specifically in muscles
muscle function at least in part by promoting the basal activity via the UAS-Gal4 system using the Mhc-Gal4 driver (see Fig-
of the autophagy/lysosome system, an intracellular protein ure S1 available online). Increased FOXO activity in muscles
degradation pathway that removes damaged protein aggregates did not affect developmental growth and differentiation (as esti-
(Rubinsztein, 2006). mated by body weight and sarcomere assembly) (Figure S2), and
Moreover, we report that FOXO/4E-BP signaling in muscles resulted in the delayed accumulation of aggregates containing
extends life span and regulates proteostasis organism-wide by polyubiquitinated proteins and Ref(2)P during aging (Figures 1G
regulating feeding behavior, release of insulin from producing and 1J, compare with control muscles in Figures 1F and 1I).
cells, and 4E-BP induction in nonmuscle tissues. Thus, we Next, we tested whether foxo null animals display accelerated
propose a model by which FOXO/4E-BP signaling in muscles muscle aging, and found an increased accumulation of protein
preserves systemic proteostasis by mimicking some of the aggregates (Figures 1H and1K), indicating that FOXO is both
protective effects of decreased nutrient intake. necessary and sufficient to modulate muscle proteostasis
(Figure 1L).
RESULTS To further corroborate these findings, we overexpressed
either the wild-type or the constitutive-active foxo transgenes
Loss of Proteostasis during Muscle Aging using the Dmef2-Gal4 muscle driver in combination with the
Is Prevented by FOXO temperature-sensitive tubulin-Gal80ts transgene to achieve
To detect cellular processes that are responsible for decreased adult-onset foxo overexpression in muscles (Figure S3). Trans-
muscle strength in aging flies, we monitored cellular changes in gene overexpression significantly preserved muscle proteosta-
indirect flight muscles of wild-type flies by immunogold-electron sis in both cases, while the controls displayed an increased
microscopy (IEM). In older flies, we detected filamentous cyto- accumulation of protein aggregates (Figure S3). All together,
plasmic structures that were instead absent in muscles from these results indicate that protein homeostasis depends on
young flies (Figures 1A–1D). Filamentous materials present in FOXO activity during muscle aging.
these structures stained with an anti-ubiquitin antibody (Fig-
ure 1D), a marker for proteins that are polyubiquitinated, sug- 4E-BP Controls Proteostasis in Response
gesting that the cytoplasmic structures are aggregates of to Pten/FOXO Activity
damaged proteins. Aggregates were variable in size and were To dissect the stimuli that encroach on FOXO to control proteo-
detected in both resin-embedded sections (Figure 1) and cryo- stasis, we tested whether Pten overexpression phenocopies
sections (data not shown) of thoracic muscles of the old but FOXO activation. Consistent with its role in activating FOXO,
not the young flies, in parallel with an increase in the overall we found that Pten decreased the accumulation of protein

(E) The number of gold particles, indicative of ubiquitin immunoreactivity, significantly increases in old age (standard error of the mean [SEM] is indicated with n;
**p < 0.01).
(F–L) Immunostaining of indirect flight muscles from flies with (UAS-foxo/+;Mhc-Gal4/+) or without (Mhc-Gal4/+) foxo overexpression at 1 week (F and G) and
5 weeks of age (I and J), and foxo homozygous null (MhcGal4, foxo21/25) flies (H and K). Polyubiquitin (red) and p62/Ref(2)P (green) immunoreactivities reveal
an increased deposition of aggregates containing polyubiquitinated proteins during aging in muscles of control flies (F and I), and, to a lesser extent, in muscles
overexpressing foxo (G and J). Conversely, muscles from foxo null animals display an accelerated deposition of protein aggregates (H and K) in comparison with
controls (F and I). Note the significant increase in the cumulative area of protein aggregates (indicative of both aggregate size and number) in (K) versus (I), and in (I)
versus (J), indicating that the control of protein homeostasis is linked to FOXO activity in muscles (quantification in [L]) (SEM is indicated with n; *p < 0.05, **p <
0.01). Representative polyubiquitin and Ref(2)P immunoreactivities are shown in insets. Phalloidin staining (blue) outlines F-actin, which is a component of muscle
myofibrils. Scale bar is 20 mm (F–K).
See also Figure S1, Figure S2, and Figure S3.

Cell 143, 813–825, November 24, 2010 ª2010 Elsevier Inc. 815
Figure 2. 4E-BP Preserves Proteostasis in Response to Pten/FOXO Signaling
(A–F) Immunostaining of muscles overexpressing Pten and constitutive active (CA) 4E-BP. In both cases, a decrease in the accumulation of polyubiquitin protein
aggregates is observed at 5 weeks of age in comparison with age-matched controls, suggesting that these interventions can preserve proteostasis in aging
muscles. Scale bar is 20 mm. Hsp70 overexpression has instead limited effects (Figure S4, Table S1, Table S2).
(G) A reduction in the cumulative area of protein aggregates is observed upon increased activity of either Pten or 4E-BP in comparison with controls (SEM is
indicated with n; **p < 0.01, ***p < 0.001).
(H) Relative quantification of Thor/4E-BP mRNA levels from thoraces of syngenic flies at 1 and 5 weeks of age. A significant increase in 4E-BP expression is
detected in response to fasting and Pten and FOXO activity (**p < 0.01, ***p < 0.001; SEM is indicated with n = 4).

aggregates during aging (Figures 2B and 2E; see controls in expression of Hsp70 and its cofactors, as estimated with Lucif-
Figures 2A and 2D). erase transcriptional reporters based on the proximal promoter
Next, we examined the responses induced by Pten/FOXO region of target genes (Figure S4 and Table S2). On this basis,
signaling. First, we examined whether FOXO activity delays we tested whether Hsp70 overexpression preserves proteosta-
protein damage by inducing chaperones that are key for protein sis during aging but found little changes in the age-related accu-
quality control (Tower, 2009). In response to FOXO activity in mulation of protein aggregates (Figure S4). Thus, we conclude
muscles, we detected an increase in the mRNA levels of that additional FOXO-dependent responses are involved.
Hsp70 and its cofactors involved in protein folding (Hip, Hop, Among the FOXO-target genes, Thor/4E-BP has a key role
Hsp40, and Hsp90) but not in protein degradation (Chip and in delaying aging by regulating protein translation (Zid et al.,
Chap) (Figure S4 and Table S1). FOXO regulates directly the 2009; Tain et al., 2009). However, the cellular mechanisms that

816 Cell 143, 813–825, November 24, 2010 ª2010 Elsevier Inc.
are regulated by 4E-BP are largely unknown. To test whether the basal expression of several Atg genes at both young and
4E-BP controls proteostasis during muscle aging, we overex- old age, suggesting that their increased expression contributes
pressed a constitutive active form of 4E-BP in muscles and to the beneficial effects of FOXO on proteostasis. To test this
observed limited accumulation of protein aggregates during hypothesis, we knocked down Atg7 levels in foxo-overexpress-
aging (Figures 2C and 2F) compared with controls (Figures 2A ing flies and analyzed the deposition of polyuiquitinated protein
and 2D). All together, increased activity of Pten or 4E-BP sig- aggregates. Interestingly, RNAi treatment brought about a
nificantly decreases the cumulative area of protein aggregates 50% decrease in Atg7 mRNA levels and resulted in a partial
(Figure 2G). increase in the buildup of insoluble ubiquitinated proteins at
In addition, a significant increase in 4E-BP mRNA levels is 8 weeks, compared with age-matched, mock-treated flies
induced in muscles upon Pten, foxo overexpression, and fasting (white RNAi) and 1-week-old flies (Figure 3P).
(Figure 2H). All together, these findings suggest that 4E-BP is key All together, these findings suggest that FOXO/4E-BP
to control proteostasis in response to Pten/FOXO signaling. signaling prevents the buildup in protein damage, at least in
part by promoting the basal activity of the autophagy/lysosome
FOXO/4E-BP Signaling Regulates Proteostasis via system.
the Autophagy/Lysosome System
While FOXO/4E-BP signaling mounts a stress resistance Prevention of Muscle Aging by FOXO and 4E-BP
response that may decrease the extent of protein damage due Extends Life Span
to various stressors (Salih and Brunet, 2008; Tain et al., 2009), To evaluate whether preserving proteostasis can prevent
we wondered whether it regulates the removal of damaged functional alterations in aging muscles, we assessed muscle
proteins via macroautophagy. In this process, entire regions of strength with negative geotaxis and flight assays (see Experi-
the cytoplasm are sequestered in a double membrane vesicle mental Procedures). As shown in Figures 4A and 4B, muscle
(autophagosome) that subsequently fuses with a lysosome, functionality gradually decreases in aging flies, resulting in
where the autophagic cargo is degraded (Rubinsztein, 2006). impaired climbing and flight ability. Notably, foxo (Figure 4A)
Although the primary role of autophagy is to mount an adaptive and 4E-BP activity (Figure 4B) significantly preserve muscle
response to nutrient deprivation, its basal activity is required strength during aging. Thus, FOXO and 4E-BP prevent both
for normal protein turnover (Hara et al., 2006). In agreement the cellular degenerative events and the functional decay of
with this notion, suppression of basal autophagy leads to the aging muscles.
accumulation of polyubiquitin protein aggregates in a number Epidemiological studies in humans have associated muscle
of contexts (Korolchuk et al., 2009; Rubinsztein, 2006). senescence with increased mortality (Nair, 2005), implying that
To test whether autophagy is regulated in response to FOXO muscle aging may have organism-wide consequences beyond
signaling in muscles, we used a GFP-tagged version of the muscle function. To ask whether the prevention of muscle aging
autophagosome marker Atg5 (Rusten et al., 2004). While the affects the organism life span, we manipulated the activity of
number of Atg5-GFP punctae decreases during aging in control components of the Akt pathway in muscles and scored for their
muscles (Figures 3A and 3B), it is in part maintained in response effects on viability. As shown in Figures 4C and 4D, either Pten,
to foxo overexpression (Figures 3C and 3D, and quantification in foxo, or 4E-BP CA overexpression in muscles is sufficient to
Figure 3E). In addition, given the interconnection between significantly extend longevity by increasing the median and
the lysosome system and autophagy, we monitored a GFP- maximum life span. 4E-BP increased life span also in foxo
tagged version of the lysosome marker Lamp1 (lysosome-asso- heterozygous null animals (Figure 4D), while Hsp70 overexpres-
ciated membrane protein 1) and detected an overall increase in sion on the other hand showed little effects (Figure S5). All
the number of GFP punctae in response to overexpression of the together, these findings indicate that the extent of muscle aging
autophagy inducer kinase Atg1, foxo, and 4E-BP CA in muscles is interconnected with the life span of the organism.
at both 1 and 5 weeks of age (Figures 3G–3I and 3K–3M in
comparison with controls in Figures 3F and 3J and quantification FOXO/4E-BP Signaling in Muscles Influences
in Figure 3N). Feeding Behavior and the Release of Insulin
Closer inspection revealed that the abundance of Lamp1-GFP from Producing Cells
vesicles inversely correlates with the progressive deposition of Considering that both fasting and FOXO induce 4E-BP expres-
polyubiquitin protein aggregates, suggesting that FOXO/4E-BP sion (Figure 2H), we wondered whether the systemic effect of
signaling regulates proteostasis at least in part via the FOXO signaling on life span extension can result, at least in
autophagy/lysosome system. To further test this hypothesis, part, from reduced food intake.
we analyzed the age-related changes in autophagy gene expres- To test this hypothesis, we examined whether feeding
sion, which have been previously used as a correlative measure- behavior would be decreased in adults with FOXO and 4E-BP
ment of autophagic activity (Gorski et al., 2003; Simonsen et al., activation in muscles. We first monitored the amount of
2008). Interestingly, the expression of several autophagy genes liquid food ingested using the CAFÉ assay (capillary feeding)
involved in autophagosome induction (Atg1), nucleation (Atg6), (Ja et al., 2007). Interestingly, feeding was decreased in
and elongation (Atg5, Atg7, and Atg8) progressively declines response to FOXO/4E-BP signaling in muscles (Figure 5A). To
during aging in muscles (Figure 3O), suggesting that gene substantiate this finding, we measured the ingestion of blue-
expression changes likely contribute to the accumulation of colored food (Xu et al., 2008) and detected significant differ-
damaged proteins. Conversely, foxo overexpression increased ences in food intake with this assay (Figure 5B), confirming

Cell 143, 813–825, November 24, 2010 ª2010 Elsevier Inc. 817
818 Cell 143, 813–825, November 24, 2010 ª2010 Elsevier Inc.
Figure 4. FOXO/4E-BP Signaling Preserves Muscle Function and Extends Life Span
(A) Muscle function gradually decreases during aging as indicated by an increase in the percentage of flies with climbing and flight defects. However, foxo
preserves their function in comparison with controls (flight ability: n[flies] = 10 (week 1 and 5) and 30 (week 8) with n[batch] = 3 (week 1 and 5) and 2 (week 8);
standard deviation (SD) is indicated and *p < 0.05. Climbing ability: (n[Mhc-Gal4/+] = 1264, n[Mhc-Gal4/UAS-foxo] = 966, with n indicating the number of flies
at day 1; p < 0.001).
(B) Similar to FOXO, 4E-BP activity also results in decreased age-related flight and climbing deficits in comparison with controls (flight ability: n[flies] R 10 (week 1
and 5) and 25 (week 8) with n[batch] R 3 (week 1 and 5) and 2 (week 8); SD is indicated and *p < 0.05. Climbing ability: (n[Mhc-Gal4/+] = 204, n[Mhc-Gal4/UAS-4E-
BP CA] = 403, p < 0.001).
(C) Survival of flies during aging. Foxo overexpression in muscles significantly extends the median and maximum life span (median and maximum life span:
Mhc-Gal4/+ = 61 and 82 days (n = 1264); UAS-foxo tr.#1/+;Mhc-Gal4/+ = 73 and 100 days (n = 1184); Mhc-Gal4/UAS-foxo tr.#2 = 76 and 94 days
(n = 966); p < 0.001).
(D) Life span of flies with increased Pten and 4E-BP activity in muscles is extended in comparison with matched controls (median and maximum life span of
4E-BP: Mhc-Gal4/+ = 63 and 78 days (n = 204); Mhc-Gal4/UAS-4E-BP CA = 71 and 84 days (n = 403); Pten: Mhc-Gal4/+ = 55 and 76 days (n = 162);
Mhc-Gal4/UAS-Pten = 66 and 88 days (n = 130); p < 0.001). Similar increase in life span is brought about by 4E-BP CA overexpression in foxo21 heterozygous
null flies.
See also Figure S5 and Figure S7.

that feeding behavior is affected. Next, to assess whether the behavior of flies overexpressing foxo and 4E-BP CA in
decreased feeding behavior arises from developmental defects, muscles most likely is not caused by developmental defects.
we measured the body weight of adult flies, which is a sensitive To assess the metabolic status, we monitored the glucose
indicator of developmental feeding (Demontis and Perrimon, concentration (glycemia) in the hemolymph. Similar to wild-
2009), but found no significant differences (Figure 5C). Thus, type flies starved for 24 hr, we detected a significant decrease

Figure 3. FOXO and 4E-BP Regulate Proteostasis at Least in Part via the Autophagy/Lysosome System
(A–E) Immunostaining of muscles expressing the marker of autophagosomes Atg5-GFP reveals a significant increase in their number (E) and maintenance at
1 and 5 weeks of age upon foxo overexpression (C and D) in comparison with controls (A and B). In (E), SEM is indicated with n; *p < 0.05 and **p < 0.01.
(F–N) Immunostaining of muscles expressing the lysosomal marker Lamp1-GFP and overexpressing either Atg1, foxo, or 4E-BP CA. Note an increase in the
number of lysosomes (N) at both 1 (G-I) and 5 weeks of age (K–M), which inversely correlates with polyubiquitin immunoreactivity in comparison with control
muscles (F and J). Scale bar is 10 mm (A–D and F-–M). In (N), SEM is indicated with n; *p < 0.05 and ***p < 0.001.
(O) Relative mRNA levels of autophagy genes from thoraces of 1- and 5-week-old flies decrease during normal muscle aging, while their expression increases and
persists in response to FOXO. SEM is indicated with n = 4; *p < 0.05, **p < 0.01 and ***p < 0.001.
(P) RNAi treatment against Atg7 results in a 50% knockdown of its mRNA levels in muscles and partially impairs FOXO-mediated proteostasis, as indicated by
the increased detection of ubiquitin-conjugated proteins in Triton X-100 insoluble fractions at 8 weeks (old, red) in comparison with mock-treated (white RNAi) and
young flies (1 week old, black). Normalized values based on a-tubulin levels are indicated.

Cell 143, 813–825, November 24, 2010 ª2010 Elsevier Inc. 819
Figure 5. FOXO Signaling in Muscles Partially
Mimics Systemic Metabolic Changes Associated
with Fasting by Modulating Feeding Behavior
(A–C) Flies in which FOXO/4E-BP activity has been altered
specifically in muscles consume less food than matched
controls. Food consumption was determined via capillary
feeding CAFÉ assay over 2 hr periods (A), and by moni-
toring the ingestion of blue colored food in 24 hr (B). Error
bars represent SEM with n[measurements] = 44, 46, 52,
37, 103, and 61 in (A) and n = 2 in (B), with *p < 0.05,
**p < 0.01, ***p < 0.001. Decreased feeding does not result
from developmental defects, as indicated by similar body
weights of flies analyzed (C) (error bars represent SD with
n R 3).
(D) Relative glucose levels (glycemia) in the hemolymph of
flies overexpressing either foxo or 4E-BP CA in muscles,
and matched controls. Manipulation of FOXO/4E-BP
signaling in muscles brings about a reduction of glycemia
similar in part to that of wild-type flies starved for 24 hr, as
estimated with the glucose hexokinase assay (SEM is
indicated with n = 5, and **p < 0.01, ***p < 0.001).
(E–H) Immunostaining of Dilp-producing median neurose-
cretory cells in the brain of starved wild-type flies, flies
overexpressing foxo in muscles, and controls. Increase
in the immunoreactivity of the insulin-like peptide Dilp2
(green) is detected in producing cells in response to either
starvation (F) or foxo overexpression in muscles (H), in
comparison respectively with fed wild-type flies (E) and
controls with no foxo overexpression in muscles (G).
Smaller changes in Dilp5 levels are observed. Phalloidin
staining (blue) detects F-actin (scale bar is 20 mm; images
in [E]–[H] have the same magnification).
(I) Quantification of the intensity of staining indicates
that differences in Dilp2 fluorescence are significant
(SD is indicated with n[measurements] = 35, 69, 37, and
96 from n[brains] = 2, 4, 3, and 4; *p < 0.05).
(J–L) Quantification and immunostaining of adipose tissue
(peripheral fat body of the abdomen) from 2 week old flies.
(J) Note a significant increase in nuclear b-galactosidase
immunoreactivity (red) in the adipose tissue from flies
with a nuclear 4E-BP-lacZ reporter and foxo overexpres-
sion in muscles (L) in comparison with controls (K). F-actin
(green) and DAPI staining (indicative of nuclei, blue) are
shown. Scale bar is 20 mm. In (J), SEM is indicated with
n = 20 and ***p < 0.001.

feeding behavior, mimic at least in part the


physiological changes that are associated with
fasting.
To gain mechanistic insights into the systemic
regulation of aging by FOXO/4E-BP signaling
in muscles, we next monitored the release of
insulin-like peptides (Dilps) from the Dilp-
producing median neurosecretory cells in the
brain, which have been previously shown to
mediate the response of life span to nutrition
in Drosophila (Broughton et al., 2010). We
detected a significant accumulation of the
insulin-like peptide Dilp2 (and to a lesser extent,
of glycemia in flies with FOXO and 4E-BP activation in muscles Dilp5) in starved wild-type flies in comparison with fed flies
(Figure 5D). All together, these findings suggest that FOXO and (Figures 5E and 5F). Increased immunoreactivity indicates
4E-BP act as a metabolic brake in muscles that, by influencing decreased release of Dilps and has been previously shown to

820 Cell 143, 813–825, November 24, 2010 ª2010 Elsevier Inc.
occur in response to starvation (Geminard et al., 2009). Next, we activity in muscles also confers systemic protection from the
tested whether similar changes would occur upon FOXO age-related decline in proteostasis. To test whether this effect
signaling in muscles and found a partial accumulation of Dilps is muscle-specific, we overexpressed foxo in the adipose tissue
(Figures 5G–5I). (abdominal fat body) with the S106GS-Gal4 driver, and analyzed
Assuming that decreased Dilps secretion may result in the deposition of polyubiquitinated proteins in Triton X-100
systemic FOXO activation, we monitored its activity using a insoluble fractions from thoraces. Under these conditions, we
nuclear 4E-BP-lacZ transcriptional reporter. By immunostaining seemingly detected no differences (Figure S6), suggesting that,
adipose tissues with anti-b-galactosidase antibodies, we although other tissues may be involved, muscles may play a
detected higher 4E-BP expression upon foxo activation in key role in this regulation. Altogether, these observations sug-
muscles in comparison with controls (Figures 5J–5L). Thus, gest that FOXO and 4E-BP activity in muscles mitigates the
FOXO signaling in muscles appears to systemically activate loss of proteostasis nonautonomously by influencing feeding
4E-BP expression in other tissues by regulating food intake behavior, insulin release from producing cells, and 4E-BP activity
and insulin release. in other tissues.

FOXO/4E-BP Signaling in Muscles Regulates DISCUSSION


Proteostasis in Other Aging Tissues
Our demonstration that FOXO/4E-BP signaling in muscles By using a number of behavioral, genetic, and molecular assays,
extends life span in Drosophila and induces a systemic fasting- we have described a mechanism in the pathogenesis of muscle
like response, along with the observation that muscles undergo aging that is based on the loss of protein homeostasis (proteo-
age-related structural and functional changes precociously in stasis) and the resulting decrease in muscle strength (Figure 7).
comparison with other tissues (Herndon et al., 2002; Zheng Increased activity of Pten and the transcription factor FOXO is
et al., 2005), raises the possibility that muscle senescence may sufficient to delay this process, while foxo null animals experi-
influence the progression of age-related degenerative events in ence accelerated loss of proteostasis during muscle aging.
the entire organism. Pten and FOXO induce multiple protective responses, including
To test this hypothesis, we examined whether, in addition to the expression of folding chaperones and the regulator of protein
life span extension, FOXO signaling in muscles can affect protein translation 4E-BP that has a pivotal role in preserving proteosta-
homeostasis in other tissues. As in the case of muscles (Figure 1 sis. FOXO and 4E-BP preserve muscle function, at least in part
and Figure 2), we found that Ref(2)P/polyubiquitin aggregates by sustaining the basal activity of the autophagy/lysosome
progressively accumulate in aging retinas (Figures 6A and 6D), system, which removes aggregates of damaged proteins.
brains (Figures 6B and 6E), and adipose tissue (Figures 6C and However, additional mechanisms may be involved. For example,
6F) (peripheral fat body of the abdomen). However, foxo overex- the proteasome system may degrade damaged proteins and
pression in muscle resulted in decreased accumulation of thus avoid their accumulation in aggregates (Rubinsztein,
protein aggregates in other aging tissues (Figures 6D–6F; quan- 2006). Thus, perturbation in proteasome assembly and subunit
tification in Figure 6G). Similar changes were observed in composition may contribute to muscle aging in response to
response to 4E-BP activity in muscles in comparison with FOXO activity. In addition, whereas overexpression of a single
syngenic controls (Figure 6H). Importantly, this regulation is chaperone had limited effects, interventions to effectively limit
muscle nonautonomous, as Mhc-Gal4 drives transgene expres- the extent of protein damage are likely to delay the decay in
sion only in muscles (and not in the retina, brain or adipose proteostasis by decreasing the workload for the proteasome
tissue) (Figure S1). To further test the finding that FOXO/4E-BP and autophagy systems (Tower, 2009).
signaling in muscles delays the systemic impairment of proteo- By comparing the accumulation of polyubiquitinated proteins
stasis in other tissues (Figures 6A–6H), we analyzed by western in aggregates of aging muscles, retinas, brains, and adipose
blot the ubiquitin levels of Triton X-100 insoluble fractions, which tissue, we have found that reduced protein homeostasis is a
included protein aggregates, from either thoraces (which mainly general feature of tissue aging that is particularly prominent in
consist of foxo-overexpressing muscles) or heads and abdo- muscles (Figure 1, Figure 6, and Figure S6). The observation
mens (which are enriched in nonmuscle tissues and muscles that muscle aging is characterized by loss of proteostasis further
with little foxo overexpression) (Figure S1), at 1 and 8 weeks of suggests some similarity between muscle aging and neurode-
age. In agreement with the increased deposition of protein generative diseases, many of which are characterized by the
aggregates observed during aging by immunofluorescence accumulation of protein aggregates (Rubinsztein, 2006).
(Figure 1, Figure 2, and Figures 6A–6F), ubiquitin levels were Mechanical, thermal, and oxidative stressors occur during
dramatically increased in the Triton X-100 insoluble fractions muscle contraction (Arndt et al., 2010), and therefore muscle
from control thoraces, and head and abdominal extracts at proteins may be particularly susceptible to damage in compar-
8 weeks of age, in comparison with 1 week of age (Figure 6I). ison with other tissues. While our findings refer to the loss of
However, ubiquitin levels were only partially increased in old proteostasis in the context of normal aging, it is likely that a better
foxo-overexpressing flies in both thoracic and head and abdom- understanding of this process will help cure muscle pathologies
inal extracts. No substantial differences were instead detected in associated with aging, as some of the underlying mechanisms of
the Triton X-100 soluble fractions (data not shown). Similar etiology may be shared. For example, most cases of inclusion
results were obtained by 4E-BP CA but not Hsp70 overexpres- body myositis (IBM) arise over the age of 50 years, defining
sion in muscles (Figure 6I; Figure S5), indicating that 4E-BP aging as a major risk factor for the pathogenesis of this disease.

Cell 143, 813–825, November 24, 2010 ª2010 Elsevier Inc. 821
Figure 6. Systemic Proteostasis Is Remotely Controlled by FOXO/4E-BP Signaling in Muscles
(A–F) Aggregates of polyubiquitinated proteins accumulate during aging in the retina (A and D), brain (B and E), and the adipose tissue (C and F) of control flies
(Mhc-Gal4/+), but to a lesser extent in tissues from flies overexpressing foxo in muscles (UAS-foxo/+;Mhc-Gal4/+), as indicated by polyubiquitin (red) and p62/Ref
(2)P (green) stainings. Phalloidin staining (blue) outlines F-actin. Note that Mhc-Gal4 does not drive transgene expression in these tissues (Figure S1). Scale bar is
10 mm.
(G and H) The age-related increase in the cumulative area of protein aggregates is significantly less prominent in tissues from flies overexpressing foxo (G) or
4E-BP CA (H) in muscles in comparison with controls (SEM is indicated with n; *p < 0.05. **p < 0.01, and ***p < 0.001).
(I) Ubiquitin levels (indicative of protein aggregates) are detected in Triton X-100 insoluble fractions from thoraces, and head and abdominal tissues from flies
overexpressing foxo in muscles or control flies at 1 (young, black) and 8 (old, red) weeks of age. Ubiquitin levels are increased in old flies in comparison with young

822 Cell 143, 813–825, November 24, 2010 ª2010 Elsevier Inc.
Figure 7. FOXO/4E-BP Signaling in Muscles
Controls Proteostasis and Systemic Aging
Muscle aging is characterized by protein damage
and accumulation of cytoplasmic aggregates.
Loss of protein homeostasis (proteostasis) associ-
ates with the progressive decrease in muscle
strength and can affect the life span of the
organism. Pten/FOXO signaling induces multiple
targets including several folding chaperones
and the regulator of protein translation 4E-BP.
FOXO/4E-BP activity regulates muscle proteosta-
sis at least in part via the autophagy/lysosome
pathway of protein degradation, preserves muscle
function, and extends life span. In addition, FOXO/
4E-BP signaling in muscles decreases feeding
behavior that, similar to fasting, results in reduced
insulin release from producing cells. This in turn
promotes FOXO and 4E-BP activity in other
tissues, preserving proteostasis organism-wide
and mitigating systemic aging.

against age-dependent changes in pro-


teostasis while also blunting develop-
mental muscle growth in flies (Demontis
and Perrimon, 2009), as observed in
mammals (Kamei et al., 2004). Thus,
Interestingly, muscle weakness in patients with IBM is character- deleterious effects of FOXO activation as observed in mamma-
ized by the accumulation of protein aggregates (Needham and lian muscles may result from the inhibition of the growth of novel
Mastaglia, 2008), which we have now described as occurring myofibers in postnatal development and adulthood, a process
in the context of regular muscle aging in Drosophila. Thus, which is thought to be limited to development in Drosophila
FOXO may interfere with the pathogenesis of muscle degenera- (Grefte et al., 2007).
tive diseases in addition to muscle aging. Studies in animal An interesting observation of our study is that interventions
disease models of IBM will be needed to test this hypothesis. that decrease muscle aging also extend the life span of the
There is an apparent contradiction between our findings and organism. In particular, our work raises the prospect that the
the data describing the FOXO-dependent induction of muscle extent of muscle aging may be a key determinant of systemic
atrophy in mice (Bodine et al., 2001; Sandri et al., 2004), a serious aging (Figure 7). Reduced muscle proteostasis may be detri-
form of muscle degeneration that results in decreased muscle mental per se for life expectancy, presumably due to the involve-
strength (Augustin and Partridge, 2009). The observation that ment of muscles in a number of key physiological functions.
different degrees of FOXO activation can promote stress resis- Consistent with this view, overexpression in muscles of aggrega-
tance, or rather cell death (Salih and Brunet, 2008), could explain tion-prone human Huntington’s disease proteins is sufficient to
why FOXO activity can be protective or rather detrimental during decrease life span (Figure S7). Moreover, FOXO signaling in
muscle aging. In particular, while physiologic FOXO activation muscles regulates proteostasis in other tissues, via the inhibition
can preserve protein homeostasis and muscle function, its of feeding behavior and the decreased release of insulin from
excessive activation may lead to decreased muscle function producing cells, which in turn promote 4E-BP activity systemi-
due to hyperactivation of the protein turnover pathways. Consis- cally. Thus, we propose that FOXO/4E-BP signaling in muscles
tent with this view, the autophagy pathway has also been regulates life span and remotely controls aging events in other
involved in both muscle atrophy (Mammucari et al., 2007; tissues by bringing about some of the protection associated
Zhao et al., 2007) and in the preservation of muscle sarcomere with decreased food intake.
organization (Arndt et al., 2010; Masiero et al., 2009), high- In mammals, muscles produce a number of cytokines involved
lighting the importance of fine-tuning the degree of activation in the control of systemic metabolism (Nair, 2005; Pedersen and
of stress resistance pathways to maintain muscle homeostasis. Febbraio, 2008). For example, interleukin-6 (IL-6) is produced by
In addition, the output of FOXO activity may radically differ in muscles and has been proposed to control glucose homeostasis
growing versus preexisting myofibers. In particular, our present and feeding behavior through peripheral and brain mechanisms
study indicates that FOXO protects preexisting myofibers (Febbraio and Pedersen, 2002; Plata-Salaman, 1998). Thus,

flies in extracts from both muscles (thoraces) and nonmuscle tissues (heads and abdomens). However, flies overexpressing foxo in muscles have reduced
deposition of protein aggregates at 8 weeks of age in both muscles and nonmuscle tissues. Similar results are obtained in response to increased 4E-BP activity
in muscles (I), but not Hsp70 (Figure S5). Quantification of ubiquitin-conjugated proteins normalized to a-tubulin or histone H3 levels is indicated.
See also Figures S1, Figure S5, and Figure S6.

Cell 143, 813–825, November 24, 2010 ª2010 Elsevier Inc. 823
a muscle-based network of systemic aging as observed in flies dissected flies were homogenized in ice-cold PBS with 1% Triton X-100 and
may occur in humans. protease inhibitors, and the resulting unsoluble pellet resuspended in RIPA
buffer with 5% SDS and 8M urea. See Extended Experimental Procedures
This study supports the common belief that preserving muscle
for a complete protocol.
function is beneficial for overall aging (Boyle et al., 2009; Chen
et al., 2005), and the notion that muscles are central tissues
SUPPLEMENTAL INFORMATION
to coordinate organism-wide processes, including aging and
metabolic homeostasis (Nair, 2005). Moreover, the observation Supplemental Information includes Extended Experimental Procedures,
that FOXO signaling in muscles influences aging events in other seven figures, and two tables and can be found with this article online at
tissues suggests that the systemic regulation of aging relies on doi:10.1016/j.cell.2010.10.007.
tissue-to-tissue communication (Russell and Kahn, 2007), which
may provide the basis for interventions to extend healthy life ACKNOWLEDGMENTS
span.
We are grateful to Andreas Brech, Didier Contamine, Ernst Hafen, Pierre
Leopold, Susan Lindquist, Ioannis Nezis, Amita Sehgal, Marc Tatar, Robert
EXPERIMENTAL PROCEDURES
Tjian, John Tower, the DRSC/TRiP, and members of the Perrimon lab for fly
stocks, reagents, and advice. We thank Maria Ericsson for assistance with
Drosophila Strains and Life Span Analysis
electron microscopy, Christians Villalta for embryo injection, and Chris Bakal,
Details on fly strains can be found in Extended Experimental Procedures.
Rami Rahal, and Jonathan Zirin for critically reading the manuscript. This work
For longevity measurement, male flies were collected within 24 hr from
was supported by the NIH (1P01CA120964-01A1) and a Pilot Project Grant
eclosion and reared at standard density (20 flies per vial) on cornmeal/soy
from the Paul F. Glenn Labs for the Molecular Biology of Aging. F.D. is an
flour/yeast fly food at 25 C. Dead flies were counted every other day and
Ellison Medical Foundation/AFAR postdoctoral fellow. N.P. is an investigator
food changed. For each genotype, at least two independent cohorts of flies,
of the Howard Hughes Medical Institute.
raised at different times from independent crosses, were analyzed. For starva-
tion treatments, flies were kept in normal vials with 1.5% agar as a water
source for the period of time indicated. For all experiments, Mhc-Gal4 females Received: February 3, 2010
were mated with male transgenic and syngenic control flies, and the resulting Revised: June 24, 2010
male offspring analyzed in parallel by comparing transgene expressing Accepted: October 1, 2010
flies with matched controls flies having the same genetic background. For Published: November 24, 2010
transgene expression with the Gal4-UAS system, flies were reared at 25 C.
REFERENCES
Behavioral and Metabolic Assays
Flight ability was scored according to Park et al. (2006), and negative geotaxis Arndt, V., Dick, N., Tawo, R., Dreiseidler, M., Wenzel, D., Hesse, M., Furst,
assays were performed as previously described (Rhodenizer et al., 2008). In D.O., Saftig, P., Saint, R., Fleischmann, B.K., et al. (2010). Chaperone-assisted
brief, flies were gently tapped to the bottom of a plastic vial, and the number selective autophagy is essential for muscle maintenance. Curr. Biol. 20,
of flies that could climb to the top of the vial after 20 s was scored. Quantifica- 143–148.
tion of the glucose concentration in the hemolymph, and capillary (CAFÉ) and Augustin, H., and Partridge, L. (2009). Invertebrate models of age-related
blue-colored food feeding assays were done as previously described muscle degeneration. Biochim. Biophys. Acta 1790, 1084–1094.
(Geminard et al., 2009; Xu et al., 2008) and are described in detail in Extended Bodine, S.C., Stitt, T.N., Gonzalez, M., Kline, W.O., Stover, G.L., Bauerlein, R.,
Experimental Procedures. Zlotchenko, E., Scrimgeour, A., Lawrence, J.C., Glass, D.J., and Yancopoulos,
G.D. (2001). Akt/mTOR pathway is a crucial regulator of skeletal muscle hyper-
Immunostaining, Confocal and Electron Microscopy, trophy and can prevent muscle atrophy in vivo. Nat. Cell Biol. 3, 1014–1019.
and Image Analysis
Boyle, P.A., Buchman, A.S., Wilson, R.S., Leurgans, S.E., and Bennett, D.A.
For whole-mount immunostaining of the fly tissues, indirect flight muscles, and
(2009). Association of muscle strength with the risk of Alzheimer disease
peripheral fat body of the abdomen, retinas, and brains were dissected from
and the rate of cognitive decline in community-dwelling older persons. Arch.
male flies and fixed for 30–40 min in PBS with 4% paraformaldehyde and
Neurol. 66, 1339–1344.
0.2% Triton X-100. After washing, samples were incubated overnight with
appropriate primary and secondary antibodies. Image analysis was done Broughton, S.J., Slack, C., Alic, N., Metaxakis, A., Bass, T.M., Driege, Y., and
with ImageJ and Photoshop. Immuno-gold electron microscopy was done Partridge, L. (2010). DILP-producing median neurosecretory cells in the
similar to Nezis et al., (2008). See Extended Experimental Procedures for Drosophila brain mediate the response of lifespan to nutrition. Aging Cell 9,
further information and a list of the antibodies used. 336–346.
Chen, H., Zhang, S.M., Schwarzschild, M.A., Hernan, M.A., and Ascherio, A.
Quantitative Real-Time RT-PCR (2005). Physical activity and the risk of Parkinson disease. Neurology 64,
qRT-PCR was done as previously described (Demontis and Perrimon, 2009). 664–669.
Total RNA was prepared from fly thoraces and qRT-PCR was performed Cohen, E., Bieschke, J., Perciavalle, R.M., Kelly, J.W., and Dillin, A. (2006).
with the QuantiTect SYBR Green PCR kit (QIAGEN). Alpha-Tubulin 84B was Opposing activities protect against age-onset proteotoxicity. Science 313,
used as normalization reference. Relative quantification of mRNA levels was 1604–1610.
calculated using the comparative CT method.
Demontis, F., and Perrimon, N. (2009). Integration of Insulin receptor/Foxo
signaling and dMyc activity during muscle growth regulates body size in
Statistical Analysis
Drosophila. Development 136, 983–993.
Statistical analysis was performed with Excel (Microsoft) and p values were
calculated with Student’s t tests and log-rank tests. Febbraio, M.A., and Pedersen, B.K. (2002). Muscle-derived interleukin-6:
mechanisms for activation and possible biological roles. FASEB J. 16, 1335–
Western Blot and Biochemical Analysis of Detergent-Insoluble 1347.
Fractions Garigan, D., Hsu, A.L., Fraser, A.G., Kamath, R.S., Ahringer, J., and Kenyon, C.
Western blot and biochemical analysis of detergent-insoluble fractions were (2002). Genetic analysis of tissue aging in Caenorhabditis elegans: a role for
done substantially as previously described (Nezis et al., 2008). In brief, heat-shock factor and bacterial proliferation. Genetics 161, 1101–1112.

824 Cell 143, 813–825, November 24, 2010 ª2010 Elsevier Inc.
Geminard, C., Rulifson, E.J., and Leopold, P. (2009). Remote control of insulin Nezis, I.P., Simonsen, A., Sagona, A.P., Finley, K., Gaumer, S., Contamine, D.,
secretion by fat cells in Drosophila. Cell Metab. 10, 199–207. Rusten, T.E., Stenmark, H., and Brech, A. (2008). Ref(2)P, the Drosophila
Giannakou, M.E., Goss, M., Junger, M.A., Hafen, E., Leevers, S.J., and melanogaster homologue of mammalian p62, is required for the formation of
Partridge, L. (2004). Long-lived Drosophila with overexpressed dFOXO in adult protein aggregates in adult brain. J. Cell Biol. 180, 1065–1071.
fat body. Science 305, 361. Park, J., Lee, S.B., Lee, S., Kim, Y., Song, S., Kim, S., Bae, E., Kim, J., Shong,
Gorski, S.M., Chittaranjan, S., Pleasance, E.D., Freeman, J.D., Anderson, C.L., M., Kim, J.M., and Chung, J. (2006). Mitochondrial dysfunction in Drosophila
Varhol, R.J., Coughlin, S.M., Zuyderduyn, S.D., Jones, S.J., and Marra, M.A. PINK1 mutants is complemented by parkin. Nature 441, 1157–1161.
(2003). A SAGE approach to discovery of genes involved in autophagic cell Pedersen, B.K., and Febbraio, M.A. (2008). Muscle as an endocrine organ:
death. Curr. Biol. 13, 358–363. focus on muscle-derived interleukin-6. Physiol. Rev. 88, 1379–1406.
Grefte, S., Kuijpers-Jagtman, A.M., Torensma, R., and Von den Hoff, J.W. Plata-Salaman, C.R. (1998). Cytokines and feeding. News Physiol. Sci. 13,
(2007). Skeletal muscle development and regeneration. Stem Cells Dev. 16, 298–304.
857–868. Rhodenizer, D., Martin, I., Bhandari, P., Pletcher, S.D., and Grotewiel, M.
Hara, T., Nakamura, K., Matsui, M., Yamamoto, A., Nakahara, Y., Suzuki- (2008). Genetic and environmental factors impact age-related impairment of
Migishima, R., Yokoyama, M., Mishima, K., Saito, I., Okano, H., and negative geotaxis in Drosophila by altering age-dependent climbing speed.
Mizushima, N. (2006). Suppression of basal autophagy in neural cells causes Exp. Gerontol. 43, 739–748.
neurodegenerative disease in mice. Nature 441, 885–889. Rubinsztein, D.C. (2006). The roles of intracellular protein-degradation
Herndon, L.A., Schmeissner, P.J., Dudaronek, J.M., Brown, P.A., Listner, pathways in neurodegeneration. Nature 443, 780–786.
K.M., Sakano, Y., Paupard, M.C., Hall, D.H., and Driscoll, M. (2002). Stochastic Russell, S.J., and Kahn, C.R. (2007). Endocrine regulation of ageing. Nat. Rev.
and genetic factors influence tissue-specific decline in ageing C. elegans. Mol. Cell Biol. 8, 681–691.
Nature 419, 808–814. Rusten, T.E., Lindmo, K., Juhasz, G., Sass, M., Seglen, P.O., Brech, A., and
Hsu, A.L., Murphy, C.T., and Kenyon, C. (2003). Regulation of aging and age- Stenmark, H. (2004). Programmed autophagy in the Drosophila fat body is
related disease by DAF-16 and heat-shock factor. Science 300, 1142–1145. induced by ecdysone through regulation of the PI3K pathway. Dev. Cell 7,
179–192.
Hwangbo, D.S., Gershman, B., Tu, M.P., Palmer, M., and Tatar, M. (2004).
Drosophila dFOXO controls lifespan and regulates insulin signalling in brain Salih, D.A., and Brunet, A. (2008). FoxO transcription factors in the mainte-
and fat body. Nature 429, 562–566. nance of cellular homeostasis during aging. Curr. Opin. Cell Biol. 20, 126–136.

Ja, W.W., Carvalho, G.B., Mak, E.M., de la Rosa, N.N., Fang, A.Y., Liong, J.C., Sandri, M., Sandri, C., Gilbert, A., Skurk, C., Calabria, E., Picard, A., Walsh, K.,
Brummel, T., and Benzer, S. (2007). Prandiology of Drosophila and the CAFE Schiaffino, S., Lecker, S.H., and Goldberg, A.L. (2004). Foxo transcription
assay. Proc. Natl. Acad. Sci. USA 104, 8253–8256. factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal
muscle atrophy. Cell 117, 399–412.
Junger, M.A., Rintelen, F., Stocker, H., Wasserman, J.D., Vegh, M.,
Simonsen, A., Cumming, R.C., Brech, A., Isakson, P., Schubert, D.R., and
Radimerski, T., Greenberg, M.E., and Hafen, E. (2003). The Drosophila
Finley, K.D. (2008). Promoting basal levels of autophagy in the nervous system
forkhead transcription factor FOXO mediates the reduction in cell number
enhances longevity and oxidant resistance in adult Drosophila. Autophagy 4,
associated with reduced insulin signaling. J. Biol. 2, 20.
176–184.
Kamei, Y., Miura, S., Suzuki, M., Kai, Y., Mizukami, J., Taniguchi, T., Mochida,
Tain, L.S., Mortiboys, H., Tao, R.N., Ziviani, E., Bandmann, O., and Whitworth,
K., Hata, T., Matsuda, J., Aburatani, H., et al. (2004). Skeletal muscle FOXO1
A.J. (2009). Rapamycin activation of 4E-BP prevents parkinsonian dopami-
(FKHR) transgenic mice have less skeletal muscle mass, down-regulated
nergic neuron loss. Nat. Neurosci. 12, 1129–1135.
Type I (slow twitch/red muscle) fiber genes, and impaired glycemic control.
J. Biol. Chem. 279, 41114–41123. Tower, J. (2009). Hsps and aging. Trends Endocrinol. Metab. 20, 216–222.

Kenyon, C. (2005). The plasticity of aging: insights from long-lived mutants. Wang, M.C., Bohmann, D., and Jasper, H. (2005). JNK extends life span and
Cell 120, 449–460. limits growth by antagonizing cellular and organism-wide responses to insulin
signaling. Cell 121, 115–125.
Korolchuk, V.I., Mansilla, A., Menzies, F.M., and Rubinsztein, D.C. (2009).
Wessells, R.J., Fitzgerald, E., Cypser, J.R., Tatar, M., and Bodmer, R. (2004).
Autophagy inhibition compromises degradation of ubiquitin-proteasome
Insulin regulation of heart function in aging fruit flies. Nat. Genet. 36, 1275–
pathway substrates. Mol. Cell 33, 517–527.
1281.
Libina, N., Berman, J.R., and Kenyon, C. (2003). Tissue-specific activities of
Wolkow, C.A., Kimura, K.D., Lee, M.S., and Ruvkun, G. (2000). Regulation of
C. elegans DAF-16 in the regulation of lifespan. Cell 115, 489–502.
C. elegans life-span by insulinlike signaling in the nervous system. Science
Mammucari, C., Milan, G., Romanello, V., Masiero, E., Rudolf, R., Del Piccolo, 290, 147–150.
P., Burden, S.J., Di Lisi, R., Sandri, C., Zhao, J., et al. (2007). FoxO3 controls
Xu, K., Zheng, X., and Sehgal, A. (2008). Regulation of feeding and metabolism
autophagy in skeletal muscle in vivo. Cell Metab. 6, 458–471.
by neuronal and peripheral clocks in Drosophila. Cell Metab. 8, 289–300.
Masiero, E., Agatea, L., Mammucari, C., Blaauw, B., Loro, E., Komatsu, M.,
Zhao, J., Brault, J.J., Schild, A., Cao, P., Sandri, M., Schiaffino, S., Lecker,
Metzger, D., Reggiani, C., Schiaffino, S., and Sandri, M. (2009). Autophagy
S.H., and Goldberg, A.L. (2007). FoxO3 coordinately activates protein
is required to maintain muscle mass. Cell Metab. 10, 507–515.
degradation by the autophagic/lysosomal and proteasomal pathways in
Morley, J.F., Brignull, H.R., Weyers, J.J., and Morimoto, R.I. (2002). The atrophying muscle cells. Cell Metab. 6, 472–483.
threshold for polyglutamine-expansion protein aggregation and cellular Zheng, J., Edelman, S.W., Tharmarajah, G., Walker, D.W., Pletcher, S.D., and
toxicity is dynamic and influenced by aging in Caenorhabditis elegans. Proc. Seroude, L. (2005). Differential patterns of apoptosis in response to aging in
Natl. Acad. Sci. USA 99, 10417–10422. Drosophila. Proc. Natl. Acad. Sci. USA 102, 12083–12088.
Nair, K.S. (2005). Aging muscle. Am. J. Clin. Nutr. 81, 953–963. Zid, B.M., Rogers, A.N., Katewa, S.D., Vargas, M.A., Kolipinski, M.C., Lu, T.A.,
Needham, M., and Mastaglia, F.L. (2008). Sporadic inclusion body myositis: Benzer, S., and Kapahi, P. (2009). 4E-BP extends lifespan upon dietary restric-
a continuing puzzle. Neuromuscul. Disord. 18, 6–16. tion by enhancing mitochondrial activity in Drosophilia. Cell 139, 149–160.

Cell 143, 813–825, November 24, 2010 ª2010 Elsevier Inc. 825
Reelin and Stk25 Have Opposing
Roles in Neuronal Polarization
and Dendritic Golgi Deployment
Tohru Matsuki,1 Russell T. Matthews,1 Jonathan A. Cooper,3 Marcel P. van der Brug,2,4 Mark R. Cookson,2
John A. Hardy,2,5 Eric C. Olson,1 and Brian W. Howell1,*
1Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
2Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
3Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
4Present address: Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
5Present address: Department of Molecular Neuroscience and Reta Lila Weston Laboratories, University College, Queens Square House,

London WC1 3BG, UK


*Correspondence: howellb@upstate.edu
DOI 10.1016/j.cell.2010.10.029

SUMMARY the adjoined centrosome correlates with the site of axon emer-
gence, which becomes the future basal side of a mature pyra-
The Reelin ligand regulates a Dab1-dependent sig- midal neuron (de Anda et al., 2005, 2010; Zmuda and Rivas,
naling pathway required for brain lamination and 1998). Later, the Golgi apparatus is positioned on the apical
normal dendritogenesis, but the specific mecha- side of pyramidal neurons, proximal to the major apical dendritic
nisms underlying these actions remain unclear. We tree and opposite to the axon and minor basal dendrites (Horton
find that Stk25, a modifier of Reelin-Dab1 signaling, et al., 2005). Dispersion of the Golgi apparatus away from
the apical pole leads to a loss of dendrite asymmetry in these
regulates Golgi morphology and neuronal polariza-
cells, with equal-sized apical and basal dendrites (Horton et al.,
tion as part of an LKB1-Stk25-Golgi matrix protein
2005). Furthermore, specialized Golgi outposts, which populate
130 (GM130) signaling pathway. Overexpression of dendrites, promote the elaboration of dendritic branches (Ye
Stk25 induces Golgi condensation and multiple et al., 2007). However, it remains to be determined how Golgi
axons, both of which are rescued by Reelin treat- positioning within neurons is regulated.
ment. Reelin stimulation of cultured neurons induces Mutations in the genes encoding the Reelin-Dab1 signaling
the extension of the Golgi into dendrites, which is pathway lead to profound defects in neuronal positioning and
suppressed by Stk25 overexpression. In vivo, Reelin dendritogenesis during brain development (Niu et al., 2004;
and Dab1 are required for the normal extension of the Rice et al., 2001). The lamination of the cerebral cortex, hippo-
Golgi apparatus into the apical dendrites of hippo- campus, and cerebellum is disorganized and appears approxi-
campal and neocortical pyramidal neurons. This mately inverted compared to normal. Reelin is a secreted ligand
that is produced in discreet layers in the developing brain
demonstrates that the balance between Reelin-
(D’Arcangelo et al., 1995; Ogawa et al., 1995). Genetic and
Dab1 signaling and LKB1-Stk25-GM130 regulates
biochemical studies have shown that it regulates a signal trans-
Golgi dispersion, axon specification, and dendrite duction pathway requiring the ApoE receptors ApoER2 and
growth and provides insights into the importance of VLDLR (D’Arcangelo et al., 1999; Hiesberger et al., 1999;
the Golgi apparatus for cell polarization. Trommsdorff et al., 1999), the cytoplasmic adaptor protein
Dab1 (Howell et al., 2000), and Src family kinases (Arnaud
INTRODUCTION et al., 2003; Bock and Herz, 2003). Disparate functions have
been proposed for Reelin-Dab1 signaling, though a clear biolog-
The development of the exquisite morphology of neurons is ical response to clarify its role in brain development is lacking
a carefully orchestrated process that optimizes the ability of indi- (Chai et al., 2009; Cooper, 2008; Förster et al., 2010; Sanada
vidual neurons to receive signals, integrate them, and transmit et al., 2004).
the output to target cells. Neuronal polarization, first observed The severity of dab1-dependent phenotypes depends on the
as the rapid growth of a process that will ultimately become an genetic background (Brich et al., 2003). We have recently identi-
axon, followed by the asymmetrical development of dendrites fied stk25 as a modifier of dab1 mutant phenotypes (unpublished
are key steps in morphological and functional maturation data). Here we characterize the role of Stk25 (also YSK1, Sok1) in
(Arimura and Kaibuchi, 2005). Interestingly, the Golgi apparatus nervous system development. Previous work has implicated
has been implicated in these different aspects of neuronal Stk25 in regulating Golgi morphology through the Golgi matrix
polarity. In the nascent neuron, the position of the Golgi and protein GM130 (Preisinger et al., 2004), which we confirm here.

826 Cell 143, 826–836, November 24, 2010 ª2010 Elsevier Inc.
GM130 regulates the fusion of ER-to-Golgi vesicles with the the axon-less phenotype in Stk25 shRNA-expressing cells was
Golgi cisternae and the fusion of Golgi cisternae into elongated the specific result of reducing Stk25 expression and that Stk25
ribbons (Barr and Short, 2003; Puthenveedu et al., 2006). Deple- kinase activity is not required for axon production.
tion or mitotic phosphorylation of GM130 leads to Golgi frag- To investigate whether Stk25 affected axon initiation or main-
mentation and reduced efficiency of biosynthetic processing tenance, we examined stage III hippocampal neurons (Figures
(Lowe et al., 1998; Marra et al., 2007; Puthenveedu et al., 2006). 1D and 1E). We found that 56% ± 5% of Stk25 knockdown
The protein kinase LKB1 and its associated factors STRAD neurons lacked an axon compared to only 7% ± 8% of control
and MO25 are known to be important for neuronal polarization, samples (Figure 1G). The longest neurite in Stk25 knockdown
axon specification, and dendrite growth (Asada et al., 2007; neurons was also significantly shorter than the incipient axon in
Barnes et al., 2007; Shelly et al., 2007). In this study, we find control cultures. Moreover, overexpression of Stk25 induced
that Stk25 is part of an LKB1 cell polarization pathway. Stk25, multiple axons. Expression of either the wild-type or kinase-inac-
LKB1, and GM130 are shown to regulate Golgi morphology tive Stk25*-RFP fusion proteins, or an Stk25-green fluorescent
and axon initiation. In addition, we show that Stk25 and Reelin- protein (GFP) fusion that has previously been shown to be bio-
Dab1 signaling have antagonistic effects on neuronal polariza- logically active (Preisinger et al., 2004), induced multiple SMI-
tion and the morphology and subcellular distribution of the positive axons in approximately 45%–50% of neurons as
Golgi. As the position of the Golgi plays roles in cell polarization, compared to 15% ± 3% in GFP-alone expressing controls
process extension, and cell migration (Fidalgo et al., 2010; (Figures 1C and 1F, lanes 5, 6, 8, and 9). Stk25 overexpression
Horton et al., 2005; Yadav et al., 2009; Ye et al., 2007), this did not increase axon length (Figure 1F). Taken together, the
evidence is fundamental for understanding the molecular control results show that Stk25 regulates axon initiation but not axon
of neuronal morphogenesis and provides new insights into the growth in cultured neurons.
biological role of Reelin-Dab1 signaling.
Reelin-Dab1 Signaling Suppresses Multiple Axon
RESULTS Production
Stk25 is expressed at relatively high levels in Reelin-Dab1
Stk25 Regulates Neuronal Polarity responsive cells in the developing cortical plate (Figure S1E)
Stk25 has previously been shown to regulate the polarized and in the adult hippocampus and cerebellar Purkinje cells (Fig-
migration of epithelial cells. As other Ste20-like kinases have ure S1F). Because we identified stk25 in a screen for modifiers of
roles in neuronal polarization (Jacobs et al., 2007; Preisinger dab1 mutant phenotypes (unpublished data), we examined
et al., 2004), we sought to assess a role for Stk25 in neuronal whether Reelin-Dab1 signaling might have an undiscovered
polarization by using hippocampal neuronal cultures (Dotti and role in axon initiation. Hippocampal neurons were cultured
Banker, 1987). These neurons have a stereotypic morphology from dab1/ mutant embryos and infected with GFP-express-
and program of differentiation and respond to Reelin-Dab1 ing lentiviruses to survey their morphology. Surprisingly, approx-
signaling (Matsuki et al., 2008). Soon after plating, they extend imately 30% of the dab1/ mutant neurons produced multiple
short uniform processes that have the potential to develop axons as compared to approximately 15% of the wild-type
into either axons or dendrites (Arimura and Kaibuchi, 2007). By neurons (Figure 1H). To determine whether the multiple axon
stage III, 48 to 72 hr later, one of the processes can be identified phenotype in dab1/ mutant neurons was sensitive to Stk25
as an axon whereas the other processes differentiate into expression level, we examined the effect of knocking down
dendrites. Stk25. Significantly fewer dab1/ mutant neurons infected
We reduced Stk25 levels by infection with a lentivirus carrying with the Stk25 shRNA-expressing lentivirus produced multiple
GFP and Stk25 shRNA and identified axons 6 days later using axons than the GFP-expressing control sample (Figure 1H). In
SMI-312, a pan-axonal neurofilament marker. Depletion of addition, a significant number of the Stk25 shRNA-expressing
Stk25 inhibited axon specification. At least 30% of the Stk25 neurons completely lacked axons. This shows that Reelin-
shRNA lentivirus-infected, GFP-positive neurons lacked an Dab1 signaling regulates axon initiation and that the multiple
axon (Figures 1B and 1F, lane 2), whereas axons were detected axon phenotype in dab1/ mutant mice is dependent upon
in all neurons infected with either empty vector (EV) or control Stk25 expression.
shRNA vectors (Figures 1A and 1F, lanes 1 and 3 and insets). Congruent with this result, growth of neurons in the presence
The longest process in Stk25 shRNA-expressing cells was also of Reelin suppressed the multiple axon phenotype caused
much shorter than the long axons of control cells (Figures 1A, by Stk25 overexpression (Figure 1I). This treatment did not,
1B, and 1F, lane 2), consistent with a failure to induce an axon. however, lead to the loss of axon production, which would be ex-
To assess whether axon absence was specifically caused by pected if Stk25 function was abolished. None of these treat-
reduced Stk25 expression, we tested for rescue by Stk25 over- ments affected axon length. Therefore, Reelin-Dab1 signaling
expression. Both kinase-active and kinase-inactive versions of appears to counteract the effects of high Stk25 expression
an shRNA-resistant Stk25 (Stk25*) were expressed as red fluo- without completely blocking its function in axon induction.
rescent protein (RFP) fusion proteins in cultures that were also
infected with the GFP-expressing, Stk25 shRNA virus (Figures Stk25 Regulates Axon Formation and Dendrite
S1A–S1D available online). Both kinase-active and kinase-inac- Asymmetry In Vivo
tive Stk25*-RFP rescued the axon-less phenotype caused by To investigate whether Stk25 regulates neuronal differentiation
Stk25 knockdown (Figure 1F, lanes 7–9). This suggests that in vivo, we electroporated the Stk25 shRNA-expressing vector

Cell 143, 826–836, November 24, 2010 ª2010 Elsevier Inc. 827
Figure 1. Stk25 Expression Regulates Axon
Differentiation in Culture
(A) Primary hippocampal neurons (E17.5) infected
with the GFP-expressing EV-control virus had
typical pyramidal neuron morphologies, including
a long SMI-positive axon (inset a) and shorter
dendrites.
(B) Neurons infected with the Stk25 shRNA virus
had shorter processes and frequently lacked
long (>250 mm) SMI-positive processes that met
the criteria for axons (inset b). An SMI-positive
process (arrowhead) from a noninfected neuron
runs parallel to the GFP-positive process (arrow).
(C) Cells overexpressing Stk25 wild-type (WT)-
GFP had multiple SMI-positive axons (insets c, c0 ).
(D) At stage III (2DIV), EV-control infected neurons
had one dominant SMI-positive axon.
(E) In contrast, Stk25 shRNA-expressing neurons
often lacked SMI-positive, axon-like processes.
(F) The number of neurons with 0, 1, 2, or more
axons and the length of the longest processes
were determined for neurons infected with
the indicated viruses. For rescue experiments,
neurons were coinfected with the Stk25 shRNA
(GFP-positive) and either RFP, Stk25* WT-RFP,
or Stk25* K49R-RFP expressing viruses (lanes
7–9, Figure S1).
(G) At stage III (2DIV), many Stk25 shRNA-
expressing neurons lacked axons as compared
to a small percentage of EV-control infected
neurons.
(H) The number of neurons with multiple axons
was increased in dab1/ (lane 2) compared to
wild-type neurons (lane 1, duplicated from F),
and this was reduced by Stk25 shRNA expression
(lanes 3).
(I) Primary hippocampal neurons that were in-
fected with either GFP- or Stk25 WT-GFP-ex-
pressing viruses were split into three groups and
grown in either neurobasal (NB), control-condi-
tioned (CCM), or Reelin-conditioned (RCM) media
for 6 days.
Statistical significance (*,**,***p < 0.0001,
Student’s t test, compared between the sample
pairs: (F) 1:2; 4:5,6,7; 7:8,9; (G) 1:2; (H) 1:2, 2:3;
n > 60; (I) 5:6; n indicated in bars). Bars: (C)
50 mm; (a) 10 mm; (c0 ) 5 mm; and (E) 20 mm. See
also Figure S1.

significantly longer (Figures 2A, 2B, and


2E). In addition, approximately 40% of
the strongly GFP-positive, Stk25 shRNA-
expressing neurons lacked identifiable
axon initial segments, detected using
anti-phospho-IkBa antibodies, suggest-
ing that axons were either absent or failed
to mature normally (Figures 2D and 2F;
into the hippocampi of fetal mice. The brains of these mice were Movie S1). By comparison, all of the GFP-positive, EV-control
analyzed for GFP expression and neuronal polarization of Ctip2- electroporated neurons examined had axon initial segments
positive, pyramidal neurons in the CA1 region of the hippo- (Figures 2C and 2F; Movie S1). This suggests that Stk25 regulates
campus at postnatal day 7 (P7). Stk25 shRNA did not interfere axon specification and dendrite growth in hippocampal pyra-
with the positioning of neurons, but their apical dendrites were midal neurons in vivo.

828 Cell 143, 826–836, November 24, 2010 ª2010 Elsevier Inc.
Figure 2. Stk25 Regulates Neuronal Polarity during Brain Development
(A) EV-control vector (GFP-positive, green) electroporated at E16.5 in utero was expressed in Ctip2-positive (red), hippocampal-pyramidal neurons at P7.
(B) Stk25 shRNA-expressing neurons (GFP-positive) were appropriately positioned in the CA1 layer, and their apical dendrites extended further than EV-control.
(C) GFP-expressing, EV-control transfected CA1 neurons had the typical pyramidal shape and phospho-IkBa- (red), GFP-positive (green) axon initial segments
(Sanchez-Ponce et al., 2008) (Movie S1).
(D) In contrast, a high percentage of strongly GFP-positive, Stk25 shRNA-expressing neurons were often misshapen and lacked axon initial segments (Movie S1).
(E) Quantification of apical dendrite length in EV-control and Stk25 shRNA hippocampi.
(F) Quantification of the number of GFP-, Ctip2-positive pyramidal neurons that had axon initial segments (n indicated in bar.)
(G) In EV-control neurons, the Golgi apparatus (trace of GRASP65 signal) is concentrated on the apical side of the neuron (Movie S2).
(H) In Stk25 shRNA-expressing neurons, the Golgi apparatus is broadly distributed throughout the neuron (Movie S2).
(I) Scheme used to determine Golgi distribution in (J).
(J) The Golgi distribution in apical, lateral (combined), or basal quadrants was quantified.
(K) The diameters of the largest apical and basal processes were determined (*p < 0.0005, Student’s t test, n R 12, neurons from three animals).
Bars: (B) 200 mm; (D and H) 10 mm. Error bars indicate standard error of the mean (SEM) in all figures.

In addition to having longer apical dendrites, the basal Stk25 Interacts with STRADa and Acts on the LKB1
dendrites of Stk25 shRNA-expressing neurons were also atyp- Signaling Pathway
ical. Normal pyramidal neurons have long, thick apical dendrites The functions of Stk25 resemble those reported for LKB1-
and much thinner and shorter basal dendrites (Horton et al., STRAD signaling (Barnes et al., 2007; Kishi et al., 2005; Shelly
2005; Figures 2G and 2K; Movie S2). The apical dendrites of et al., 2007). This pathway has a prominent role in cell polarity
Stk25 shRNA-expressing neurons had normal thickness, but control across numerous cell types from Caenorhabditis elegans
the basal dendrites were thicker than normal (Figures 2H and to man. LKB1 is partially regulated by binding STRAD, which
2K; Movie S2). We were not able to measure the length of the both shuttles it from the nucleus to the cytoplasm and stabilizes
basal dendrites. Therefore, there is evidence for growth of both it. We therefore investigated whether Stk25 associates with the
apical and basal dendrites, and this reduced the distinction LKB1-STRAD signaling complex. By immunoprecipitating
between apical and basal dendrites in terms of thickness. This tagged fusion proteins coexpressed in HEK293T cells, we found
suggests that Stk25 is needed for normal axon production and that both wild-type and kinase-inactive HA-Stk25 coimmuno-
dendrite asymmetry in vivo. precipitated with myc-STRADa (Figure S2A). Identifying Stk25

Cell 143, 826–836, November 24, 2010 ª2010 Elsevier Inc. 829
Figure 3. Stk25-RFP Overexpression Rescues the Neuronal Polarization Defect Caused by LKB1 but Not by GM130 Knockdown
(A) Expression of LKB1 shRNA (GFP-positive, green) in hippocampal neurons led to an increase in the number of neurons that lack an axon at 6DIV in cells also
expressing RFP (red). (a) Longest process lacks SMI immunoreactivity.
(B) In contrast, overexpressing Stk25* WT-RFP in LKB1 knockdown neurons rescued axon production. (b) Long, axon-like process is SMI positive.
(C) GM130 knockdown (GFP-positive) also caused a reduction in axon production in RFP-positive cells. (c) No SMI imunoreactivity was detected in processes of
the GFP-, RFP-positive neuron.
(D) Stk25* WT-RFP expression did not rescue axonogenesis in GM130 knockdown neurons. (d) Longest process is SMI negative.
(E) Axon number and the length of the longest processes were quantified for the indicated treatment groups. (Lane 1 was duplicated from Figure 1F lane 1.)
(*p < 0.005 compared to lane 1, **p = 0.01 compared to lane 2, Student’s t test.)
Bars: (D) 50 mm; (d) 5 mm. See also Figure S2.

as a direct or indirect STRAD-binding protein suggests a poten- these experiments show that the Stk25 protein, not its kinase
tial role for Stk25 on the LKB1 pathway. activity, is required for LKB1-STRAD-regulated epithelial cell
To investigate whether Stk25 is important for LKB1 function, polarization.
we took two approaches. We examined whether (1) Stk25 is We then confirmed that LKB1 knockdown leads to a loss of
required for LKB1-STRAD-regulated epithelial cell polarization axon initiation in cultured hippocampal neurons (Figure 3A;
and (2) Stk25 overexpression rescues the LKB1 knockdown Barnes et al., 2007; Shelly et al., 2007). We tested whether
phenotype in neurons. Stk25 can rescue or bypass the LKB1 requirement by overex-
We first tested whether reduced Stk25 expression would pressing Stk25* wild-type (WT)-RFP in LKB1 shRNA-expressing
inhibit the LKB1-STRAD-dependent polarization of W4 intestinal neurons (Figure 3B). Ninety-two percent of LKB1 knockdown
epithelial cells. These cells have been engineered to constitu- neurons that expressed Stk25* WT-RFP produced at least one
tively express LKB1 and express STRAD in response to doxycy- axon compared to only 48% of RFP-, LKB1 shRNA-coexpress-
line, which leads to their polarization (Baas et al., 2004). Most W4 ing neurons (Figure 3E). These results are consistent with a role
cells infected with EV and control shRNA lentiviruses became of Stk25 on the LKB1 pathway to regulate axon induction.
polarized within 24 hr of doxycycline treatment (Figures S2C
and S2E). In contrast, only 20% of cells infected by the human- GM130 Interacts with Stk25 and Regulates Axon
ized (h) Stk25 shRNA lentivirus were polarized by doxycycline Induction
treatment (Figures S2C and S2E). The Golgi matrix protein GM130, which has critical roles in regu-
Furthermore, expression of either wild-type or kinase-inactive lating Golgi dynamics, was identified in a yeast two-hybrid
Stk25*-RFP rescued STRAD-induced polarization in Stk25 screen as an Stk25 binding partner (Preisinger et al., 2004). We
shRNA-expressing W4 epithelial cells (Figure S2F). Collectively, confirmed this interaction by coimmunoprecipitating tagged

830 Cell 143, 826–836, November 24, 2010 ª2010 Elsevier Inc.
Figure 4. Golgi Apparatus Morphology Is
Regulated by Stk25, LKB1, and GM130
Expression and Reelin Signaling
(A) Stage III neurons that were infected with the
EV-control virus had typical cis-Golgi ribbons
(GRASP65, Movie S3). In contrast, the cis-Golgi
in Stk25 shRNA-, LKB1 shRNA-, or GM130
shRNA-expressing neurons was fragmented
(Movie S3). GFP signal was omitted for clarity.
(B) Significantly more Stk25 knockdown neurons
had fragmented Golgi complexes compared to
the EV-control and the control shRNA (n, as indi-
cated). LKB1 and GM130 knockdown also
caused significant Golgi fragmentation as com-
pared to EV-control infected neurons. Stk25*-
RFP expression rescued Golgi fragmentation in
LKB1 shRNA but not GM130 shRNA-expressing
neurons.
(C) Neurons overexpressing either Stk25 WT-GFP
or Stk25 K49R-GFP had condensed cis-Golgi
(GRASP65 signal) compared to EV-controls when
grown in either neurobasal or control-CM. Growth
in Reelin-CM partially rescued the Golgi appear-
ance in Stk25-overexpressing cells. GM130 and
GRASP65 colocalized under all conditions (not
shown).
(D) Golgi volume (upper panel) and the length of
the longest Golgi ribbon (lower panel) were deter-
mined (*p < 0.0001, Student’s t test, n indicated in
bars).
Bars: 5 mm. See also Figure S3.

Stk25, GM130, and LKB1 Regulate


Golgi Distribution
Previously it was shown that GM130
regulates Golgi morphology in HeLa cells
(Puthenveedu et al., 2006). Given that
Stk25, LKB1, and GM130 regulate axon
fusions of GM130 and Stk25 (Figure S2B). Interestingly, kinase- initiation, and the position of the Golgi apparatus early in differen-
inactive Stk25 consistently immunoprecipitated with GM130 tiation normally coincides with axonal localization (de Anda et al.,
more efficiently than wild-type, suggesting that Stk25-depen- 2005, 2010), we examined whether Stk25, LKB1, and GM130
dent phosphorylation may destabilize the complex. regulate Golgi morphology (Figure 4). Individually knocking
Stk25 colocalizes with GM130 at the Golgi apparatus of HeLa down Stk25, LKB1, and GM130 in stage III primary hippocampal
cells (Preisinger et al., 2004). To determine whether Stk25 local- neurons resulted in dispersion of Golgi elements in a high
izes to the Golgi complex in neurons, we raised an antibody to percentage of cells, in contrast to the typical elongated
a region of Stk25 that is divergent from the close relatives morphology observed in the EV-control neurons (Figures 4A
Mst3 and Mst4 (Extended Experimental Procedures). Endoge- and 4B; Movie S3).
nous Stk25 expression overlapped with the GM130-positive Interestingly, the Golgi fragmentation caused by LKB1 knock-
cis-Golgi in neurons at stage III, coincident with axon specifica- down was rescued by Stk25*-RFP overexpression (Figure 4B),
tion (Figure S2D). suggesting that Stk25 overexpression can compensate for
To asses whether GM130 plays a role in neuronal differentia- reductions in LKB1 signaling. In contrast, Golgi fragmentation
tion, we examined GM130 shRNA-expressing neurons for in GM130 shRNA-expressing cells was not rescued by Stk25
defects in polarity. Similar to Stk25 and LKB1 knockdown overexpression (Figure 4B). Overexpression of either Stk25
neurons, knockdown of GM130 reduced axon number at 6DIV WT-GFP or Stk25 K49R-GFP led to the condensation of the
(Figure 3C). GM130 knockdown also caused a significant reduc- Golgi into a smaller volume (Figure 4C, neurobasal). Therefore,
tion in axon initiation in stage III (2DIV) neurons (data not shown). increasing or decreasing Stk25 expression from endogenous
Stk25*-RFP overexpression in GM130-deficient cells did not levels has different consequences for Golgi morphology, in addi-
rescue axon number at 6DIV (Figure 3D), which suggests that tion to having the opposite effects on axon production. These
GM130 is required for neuronal polarization downstream of results suggest an LKB1-Stk25-GM130 pathway for Golgi regu-
Stk25. lation in cultured neurons.

Cell 143, 826–836, November 24, 2010 ª2010 Elsevier Inc. 831
Importantly, Stk25 knockdown in hippocampal pyramidal
neurons also caused Golgi fragmentation in vivo, as determined
by use of in utero electroporation. Normally, the Golgi is strictly
localized to the apical side of the soma and forms outposts in
the apical dendrite (Horton et al., 2005; Figures 2G and 2J; Movie
S2). However, in Stk25 shRNA-expressing, Ctip2-positive
neurons, the Golgi apparatus was often broadly distributed
throughout the soma (Figures 2H and 2J; Movie S2).
In summary, these results indicate that Stk25, LKB1, and
GM130 are required for normal Golgi morphology in neurons at
a time when axons are first appearing. Furthermore, the frag-
mented Golgi phenotype correlated with the loss of axon
production in neurons, and both phenotypes were rescued by
Stk25 overexpression in LKB1 knockdown cells.

Reelin Signaling Regulates Golgi Morphology


As Stk25 and Reelin have opposing effects on axon initiation
(Figure 1H) and Stk25 affects Golgi morphology (Figures 4A
and 4B), we investigated the role of Reelin in regulating Golgi
morphology.
First we examined the appearance of the Golgi apparatus in
hippocampal and neocortical pyramidal neurons of reelin/
and dab1/ mutant mice. In the pyramidal layer of the wild-
type CA1 zone and in developing neocortical layers, the Golgi
apparati were linearly organized and extended tens of microns
into the apical processes (Figure 5D; Figures S4D and S4G,
insets). The Golgi of the reelin/ and dab1/ mutants often
appear convoluted near the nucleus rather than extended into Figure 5. The Golgi Apparatus Extends into an Apical Process in
a dendrite (Figures 5E and 5F; Figures S4E and S4F, insets). Neonatal Hippocampus in a reelin- and dab1-Dependent Manner
The distance from the Ctip2-positive nucleus to the tip of (A) Ctip2-positive CA1 neurons are organized into a tight lamella in wild-type
the Golgi ribbon was significantly decreased in reelin/ and brain.
dab1/ mutants as compared to wild-type (Figure 5G and Fig- (B) Homozygous disruption of reelin or (C) dab1 causes dispersion of these
neurons.
ure S4G), indicating that the reelin and dab1 genes either directly
(D) Confocal imaging through the CA1 region of the wild-type hippocampus
or indirectly regulate Golgi extension into the apical process of
revealed that the Golgi apparatus (white or green, inset) extends radially into
pyramidal neurons. the presumptive apical dendrite of Ctip2-positive neurons (red, inset).
As reelin and dab1 also regulate the proper layering of hippo- (E) In equivalent reelin/ or (F) dab1/ mutant sections, the Golgi is more
campal pyramidal neurons (Caviness and Sidman, 1973; Goffi- often convoluted proximal to the nucleus (inset). Insets were selected from
net, 1984; Rice et al., 2001) (Figures 5B and 5C), the effects of regions where isolated cells could be distinguished.
reelin and dab1 on Golgi deployment may be indirect. Therefore, (G) The Golgi phenotype was quantified by measuring the distance from the
nucleus to the furthest tip of the Golgi ribbon. (*p < 0.0001, Student’s t test,
we tested whether Reelin-Dab1 signaling acutely induces
n indicated in bar from three animals per group.)
changes in Golgi morphology or localization by treating hippo- Bar: 200 mm in (C), 20 mm in (F), and 2 mm in inset. See also Figure S4.
campal neuron cultures with Reelin for 30 min. Hippocampal
pyramidal neurons were infected with a low titer GFP-expressing
lentivirus to help visualize individual neurons. The Golgi was expressing viruses after 72 hr in culture and treated analogously
largely localized close to the nucleus in control-conditioned to experiments described above. Expression of either Stk25
media (CM) and neurobasal-treated Ctip2-positive pyramidal WT-GFP and Stk25 K49R-GFP reduced but did not eliminate
neurons (Figures 6A and 6C). However, in approximately the Golgi extension in response to Reelin (Figures 6B and 6C).
80% ± 5% of Reelin-CM-treated neurons, the Golgi apparati Under these conditions, linear Golgi ribbons were observed
extended into the largest dendritic process (Figures 6A and extending into the dendrites, but on average this was approxi-
6C). The distance between the nucleus and the most distal mately 50% the distance observed in the Reelin-treated, GFP-
portion of the Golgi ribbon from randomly selected Ctip2-posi- expressing cells (Figure 6B). Furthermore, Reelin signaling
tive neurons was significantly larger in the Reelin-CM-treated suppressed Golgi compaction induced by Stk25 overexpression
samples compared to the control-CM- and neurobasal-treated (Figures 4C and 4D). In cultures that were grown in Reelin-CM for
samples (Figure 6B). The Golgi apparatus is therefore rapidly 2 days (Figure 4), we did not observe Golgi deployment into
deployed into dendrites in response to Reelin stimulation. dendrites. This is not surprising as components of the Reelin-
We next evaluated whether the Golgi response to Reelin was Dab1 pathway begin to be degraded within a few hours. In
sensitive to elevated Stk25 expression levels. Hippocampal 60-day-old animals, Golgi extension into dendrites was also
neurons were infected with Stk25 WT-GFP or Stk25 K49R-GFP reduced (data not shown). Therefore, Golgi deployment appears

832 Cell 143, 826–836, November 24, 2010 ª2010 Elsevier Inc.
Figure 6. Reelin Stimulation Leads to Rapid Golgi Extension into Dendrites
Primary hippocampal neurons were infected with GFP-expressing viruses after 3DIV and stimulated 3 days later.
(A) The Golgi apparati in Reelin-CM-treated neurons extended tens of microns into dendrites, compared to little or no extension into dendrites of control-CM or
neurobasal-treated neurons.
(B) The distance between the nucleus and the tip of the Golgi was measured for GFP-, Ctip2-positive neurons. Expression of Stk25 WT-GFP and Stk25 K49R-GFP
caused a significant reduction in Reelin-induced Golgi extension.
(C) The Golgi of most GFP-, Ctip2-positive Reelin-CM-treated neurons extended at least 10 mm from the nucleus into or toward a dendrite. Significantly fewer
Golgi were observed in the processes of control-treated samples or Reelin-CM-treated samples that also overexpressed Stk25.
Yellow arrows indicate furthest tip of Golgi ribbon from nucleus. (*p < 0.0001, **p = 0.0002, ***p < 0.05, Student’s t test, between Reelin-CM- and control-treated
samples and between GFP- and Stk25-expressing samples treated with Reelin-CM.) Bars: 10 mm.

to be a transient, developmental phenomenon. Thus, similar to Figure 4, and Figure 6). Together this implicates the LKB1
the manifestation of the multiple axon phenotype caused by pathway, GM130, Stk25, and Reelin-Dab1 signaling in Golgi
Stk25 overexpression or loss of dab1 gene function, the degree regulation during neuronal polarization.
of Golgi extension seems to be determined by a competition
between Reelin-Dab1 signaling and Stk25 levels. Involvement of the Golgi Apparatus in Neuronal
Polarization
DISCUSSION The Golgi apparatus and centrosomes reorient as neurons
migrate into the cortical plate (de Anda et al., 2010; Nichols and
In this study, we find that Reelin-Dab1 signaling acts in an Olson, 2010). At the time of axon initiation, the centrosome is
opposing manner to LKB1, GM130, and Stk25 to regulate the near the basal pole (rear) of the cell. It then moves to the opposite
polarization of axons, dendrites, and Golgi apparati of hippo- pole (front) and is important for extending an apical process that
campal neurons, as shown in Figure 7. Knocking down these is used for radial migration (de Anda et al., 2010). The apical
three proteins led to Golgi fragmentation and inhibited axon process subsequently transforms into the apical dendritic tree,
initiation (Figure 1, Figure 3, and Figure 4). In contrast, Stk25 with the Golgi and centrosomes at its base (Barnes et al., 2008;
overexpression caused Golgi condensation and the formation Horton et al., 2005). The same events presumably occur during
of multiple axons (Figure 1 and Figure 4). It also rescued axon migration of hippocampal pyramidal neurons in vivo. When
production and Golgi fragmentation caused by LKB1 knockdown hippocampal neurons are cultured, the centrosome position
but did not rescue either phenotype caused by reduced GM130 determines which neurite becomes an axon (de Anda et al.,
expression (Figure 3 and Figure 4), suggesting that Stk25 func- 2005). Later, the apical localization of the Golgi apparatus pro-
tions as an intermediary between LKB1 and GM130. Stk25 motes the asymmetric growth of the apical compared to the basal
directly or indirectly binds to the LKB1-STRAD complex and dendrites (Horton et al., 2005). Consistent with this, Stk25 knock-
GM130 and may play a scaffolding role to link LKB1 signaling down led to Golgi disorganization, inhibited axon induction, and
to GM130 and Golgi regulation (Figure S2). Reelin-Dab1 signaling lessened the asymmetry between the long, thick apical dendrite
antagonizes the effects of Stk25 overexpression on Golgi and short, slender basal dendrites (Figures 2F, 2H, 2J, and 2K).
morphology and neuronal polarization as well as inducing polar- The Golgi may influence axon initiation through nucleating
ized deployment of the Golgi into the apical dendrite (Figure 1, microtubules, regulating secretory trafficking, or interacting

Cell 143, 826–836, November 24, 2010 ª2010 Elsevier Inc. 833
et al., 2009; Figure S2). However, although Mst4 kinase activity
is required during this process, the kinase activity of Stk25 is
not needed to induce polarized brush border formation, regulate
Golgi morphogenesis, or polarize hippocampal neurons (Fig-
ure 1F and Figures 4C and 4D). This suggests a kinase-indepen-
dent scaffolding function for Stk25 (Figure 7), which is reminis-
cent of the pseudokinase STRAD (Lizcano et al., 2004). GM130
appears to be necessary for Stk25 effects on Golgi and neuronal
Figure 7. Model of Stk25 as a Scaffolding Protein Acting Competi- polarization; however, it may not be sufficient. By linking LKB1
tively with Reelin-Dab1 Signaling signaling to GM130, Stk25 may directly regulate GM130 or indi-
LKB1 is known to act in complex with STRAD to regulate cellular polarity rectly modulate the activity of other Golgi proteins.
(Alessi et al., 2006). Reelin, the receptors ApoER2 and VLDLR, and Dab1
also form a signaling complex (Hiesberger et al., 1999; Trommsdorff et al.,
1998). STK25 coimmunoprecipitates with STRAD and GM130 (Figure 2S). Reelin-Dab1 Signaling Regulates Neuronal Polarization
Overexpression of LKB1 and STRAD is known to induce the formation of and Golgi Deployment
multiple axons (Barnes et al., 2007; Shelly et al., 2007). Independent of its Our work also shows that Reelin-Dab1 signaling, acting in oppo-
kinase activity, STK25 does so also and induces Golgi condensation (Figure 1F sition to LKB1-Stk25-GM130, affects Golgi morphology and
and Figure 4A). Knocking down LKB1, Stk25, or GM130 causes Golgi frag-
axon formation. The absence of Reelin or Dab1 inhibited Golgi
mentation/dispersion and lost axon production, the opposite to Golgi conden-
sation and multiple axon formation (Figure 1, Figure 3, and Figure 4) (Barnes deployment into the apical dendrite in vivo (Figure 5 and
et al., 2007; Shelly et al., 2007). The overexpression phenotypes are sup- Figure S4), and long-term growth in Reelin opposed Golgi
pressed by Reelin stimulation. Dab1/ neurons (Reelin signaling deficient) condensation induced by Stk25 overexpression in vitro (Fig-
have multiple axons and shorter dendrites (Figure 1F) (Niu et al., 2004). Reelin ure 4). Similarly, Dab1 absence induced supernumerary axons
stimulation induces Golgi deployment and dendrite growth, phenotypes sup- in vitro (Figure 1H), the opposite effect to depleting Stk25.
pressed by Stk25 expression/overexpression (Figure 2 and Figure 6).
However, Reelin-Dab1 and LKB1-Stk25-GM130 do not fit into
a simple epistatic relationship. For example, Stk25 depletion
reduces axon number even when Dab1 is absent, suggesting
with the centrosome (Efimov et al., 2007; Pfenninger, 2009;
that Stk25 does not require Dab1 to regulate axon number (Fig-
Rosso et al., 2004; Sütterlin and Colanzi, 2010). It seems less
ure 1). This indicates that LKB1-Stk25-GM130 and Reelin-Dab1
likely that the Golgi is required to supply materials to sustain
act on the Golgi and axon initiation through different pathways,
axon growth, as none of our manipulations affected axon length,
and the balance between the two pathways determines the
only axon number. Therefore, the Golgi probably has a signaling
outcome. In this respect, Golgi distribution is a quantitative trait,
or microtubule nucleation role in axon specification. Indeed,
not all or none, and may be influenced by other factors. Indeed,
microtubule stabilization has been shown to enhance axon
extended Golgi were observed in a subset of neurons in reelin/
formation (Witte et al., 2008), and inhibiting post-Golgi trafficking
and dab1/ mutant brains (Figure 5 and Figure S4). One possi-
disrupts axo-dendritic polarization (Bisbal et al., 2008; Yin et al.,
bility is that Reelin-Dab1 and LKB1-Stk25-GM130 regulate
2008). In dendrites, however, the Golgi may have a role in
different aspects of Golgi morphology through different mecha-
supplying materials for dendrite growth, as we detected effects
nisms. For example, Reelin-Dab1 may regulate ER-Golgi vesicle
on dendrite thickness and length (Figures 2E and 2K). Deploy-
movement, and LKB1-Stk25-GM130 may affect vesicle fusion.
ment of the Golgi into the apical dendrite may initiate the forma-
In sum, we have characterized Stk25, a modifier of the Reelin-
tion of dendritic Golgi outposts, which have been shown to
Dab1 pathway, and shown that it acts on the LKB1-STRAD
promote dendrite growth and branching (Horton et al., 2005;
pathway to regulate Golgi morphology and neuronal polariza-
Ye et al., 2007).
tion. Stk25 may play a scaffolding role to link LKB1-STRAD to
We found that Stk25 functions in Golgi morphology and axon
Golgi regulation through binding GM130, as the kinase activity
specification as part of an LKB1 pathway (Figure 3 and Figure 4).
was shown to be dispensable for neuronal polarization and Golgi
LKB1, the mammalian Par-4 homolog, is an evolutionarily
morphogenesis. We find that Reelin-Dab1 signaling regulates
conserved cell polarity protein that is known to regulate axo-
Golgi morphology and deployment into dendrites in a competi-
dendritic polarity in neurons (Barnes et al., 2008). LKB1 is acti-
tive manner with Stk25. Golgi position has been shown to
vated upon binding STRAD and MO25 (Alessi et al., 2006).
enhance local secretory trafficking (Horton et al., 2005; Ye
STRAD stabilized LKB1 in processes prior to axon production
et al., 2007); thus, this competition may regulate membrane
and in the nascent axon, suggesting a role in axon specification
and protein cargo flow into proximal dendrites. Our findings
(Shelly et al., 2007). As a master kinase, LKB1 activates several
provide new insights into the regulation of morphogenic changes
downstream kinases that regulate various aspects of cell
in neurons that drive neuronal polarization and brain lamination.
polarity. These include the Sad A and Sad B kinases, which
are required for neuronal polarization (Barnes et al., 2007; Kishi
et al., 2005). Mst4, another downstream kinase, is closely related EXPERIMENTAL PROCEDURES

to Stk25. Like Stk25, it binds to GM130 and is enriched in the


Expression Vectors
Golgi apparatus (Preisinger et al., 2004). Both Mst4 and Stk25 The lentiviral vectors used in this study were based on pLentiLox 3.7 (pLL3.7)
are required downstream of LKB1-STRAD induction for polar- vectors (Rubinson et al., 2003) with the following substitutions: (1) for shRNA
ized brush border formation in epithelial cells (ten Klooster experiments, instead of the CMV promoter, the CMV enhancer/chicken b-actin

834 Cell 143, 826–836, November 24, 2010 ª2010 Elsevier Inc.
promoter (Niwa et al., 1991) directs GFP expression; (2) for fusion protein ACKNOWLEDGMENTS
experiments, instead of the U6 promoter the CMV enhancer/chicken b-actin
promoter directs expression. The shRNA constructs include Stk25 shRNA AG We would like to thank Zainab Mansaray and Kristin Giamanco for experi-
GAGCTCCTGAAGCACAAAT and control shRNA AGTAGCTCCTAAAGCACA mental assistance, Michael Zuber for comments on the manuscript, Hans
CAT. The lentivirus production was as previously described (Matsuki et al., Clevers for cell lines, Louis Cantley and Jun-ichi Miyazaki for DNA vectors,
2008). The knockdown viruses were confirmed to reduce expression of either Arvydas Matiukas and Melissa Pepling for assistance with confocal micros-
Stk25, LKB1, or GM130 (Figure S1 and Figure S3). The Stk25 K49R mutant has copy, and Bonnie Lee Howell for editing. This work was supported by funds
previously been reported to be kinase inactive, which we confirmed (Preisinger from the NINDS intramural program and SUNY Upstate Medical University
et al., 2004 and data not shown). to B.W.H.; NIH grants NS066071 to E.C.O., NS069660 to R.T.M., and
CA41072 to J.A.C.; and NIA intramural funds for M.R.C.
Animals
All animals were used in accordance with protocols approved by the Animal Received: May 3, 2010
Care and Use Committees of SUNY Upstate Medical University, National Insti- Revised: August 27, 2010
tutes of Neurological Disorders and Stroke, and the Fred Hutchinson Cancer Accepted: October 20, 2010
Research Center, following NIH guidelines. Time pregnant mice (C57BL/6 Published: November 24, 2010
for in vitro experiments and Swiss Webster for in utero electroporations) and
rats (Sprague Dawley) were purchased from Charles River Laboratories and REFERENCES
Taconic. The dab1/ (Howell et al., 1997) and reelin/ (Jackson Labs)
mice were on the C57BL/6 strain. Alessi, D.R., Sakamoto, K., and Bayascas, J.R. (2006). LKB1-dependent
signaling pathways. Annu. Rev. Biochem. 75, 137–163.
Immunocytochemistry
Arimura, N., and Kaibuchi, K. (2005). Key regulators in neuronal polarity.
Immunocytochemistry was done according to published methods (Matsuki
Neuron 48, 881–884.
et al., 2008) and is detailed in the Extended Experimental Procedures along
with a list of the antibodies used. To measure Golgi volumes and length of Arimura, N., and Kaibuchi, K. (2007). Neuronal polarity: from extracellular
the longest Golgi ribbon, we immunostained the neurons with anti-GRASP65, signals to intracellular mechanisms. Nat. Rev. Neurosci. 8, 194–205.
anti-GFP, and anti-Ctip2, which recognizes a CA1 and layer V pyramidal Arnaud, L., Ballif, B.A., Förster, E., and Cooper, J.A. (2003). Fyn tyrosine kinase is
neuron-specific transcription factor. The area of the Golgi apparatus was a critical regulator of disabled-1 during brain development. Curr. Biol. 13, 9–17.
calculated for each Z-plain (Image Examiner, Zeiss), multiplied by the thick- Asada, N., Sanada, K., and Fukada, Y. (2007). LKB1 regulates neuronal migra-
ness of the section, and summed to determine the volume. tion and neuronal differentiation in the developing neocortex through centro-
somal positioning. J. Neurosci. 27, 11769–11775.
Cell Culture
Baas, A.F., Kuipers, J., van der Wel, N.N., Batlle, E., Koerten, H.K., Peters, P.J.,
Hippocampal neuronal cultures were isolated from embryonic day (E) 17.5
and Clevers, H.C. (2004). Complete polarization of single intestinal epithelial
mice or E18.5 rats and grown in neurobasal samples supplemented with 2%
cells upon activation of LKB1 by STRAD. Cell 116, 457–466.
B27 (Invitrogen, Matsuki et al., 2008). For polarity studies, neurons (1 3 104
cells per cm2) were infected with the respective viruses on the day of culturing Barnes, A.P., Lilley, B.N., Pan, Y.A., Plummer, L.J., Powell, A.W., Raines, A.N.,
and replated 2 days later on poly-L-lysine coated coverslips placed over Sanes, J.R., and Polleux, F. (2007). LKB1 and SAD kinases define a pathway
a monolayer of astrocytes. Axons were quantified at 2 days in vitro (DIV) or required for the polarization of cortical neurons. Cell 129, 549–563.
6DIV as indicated, following standard criteria (Shelly et al., 2007). For Golgi Barnes, A.P., Solecki, D., and Polleux, F. (2008). New insights into the molec-
deployment assays, rat cultured neurons (3 3 105 cells per cm2) were infected ular mechanisms specifying neuronal polarity in vivo. Curr. Opin. Neurobiol.
with low titer virus on day 3 and treated and fixed on day 6 in culture. Similar 18, 44–52.
results were obtained with mouse neurons (data not shown). The control- Barr, F.A., and Short, B. (2003). Golgins in the structure and dynamics of the
and Reelin-conditioned media were collected and concentrated as previously Golgi apparatus. Curr. Opin. Cell Biol. 15, 405–413.
described (Matsuki et al., 2008).
Bisbal, M., Conde, C., Donoso, M., Bollati, F., Sesma, J., Quiroga, S., Dı́az
Añel, A., Malhotra, V., Marzolo, M.P., and Cáceres, A. (2008). Protein kinase
Analysis of In Utero Electroporated Brains
d regulates trafficking of dendritic membrane proteins in developing neurons.
To knock down Stk25 expression, DNA was injected into the lateral ventricle of
J. Neurosci. 28, 9297–9308.
E17.5 embryos of Swiss Webster mice in utero and electroporated (70 mV) as
previously described (Olson et al., 2006) with the electrode paddles oriented to Bock, H.H., and Herz, J. (2003). Reelin activates SRC family tyrosine kinases in
direct the DNA into the hippocampus. Perfused brains were processed for neurons. Curr. Biol. 13, 18–26.
analysis on P7. Floating sections (70–100 mm) were immunostained with anti- Brich, J., Shie, F.S., Howell, B.W., Li, R., Tus, K., Wakeland, E.K., Jin, L.W.,
bodies described in the figure legends. Confocal images were collected with Mumby, M., Churchill, G., Herz, J., and Cooper, J.A. (2003). Genetic modula-
overlapping optical sections through 30 mm, which were flattened for display. tion of tau phosphorylation in the mouse. J. Neurosci. 23, 187–192.
We assessed whether axon initial segments or Golgi elements belonged to Caviness, V.S.J., Jr., and Sidman, R.L. (1973). Retrohippocampal, hippo-
a particular GFP-positive neuron (Figure 2), by examining movies of either campal and related structures of the forebrain in the reeler mutant mouse.
3D-rendered images or Z sections (Movie S1 and Movie S2). Golgi areas J. Comp. Neurol. 147, 235–254.
(Figures 2G and 2H) were produced by thresholding (Adobe Photoshop) flat-
Chai, X., Förster, E., Zhao, S., Bock, H.H., and Frotscher, M. (2009). Reelin
tened, 2D-negative images to match the GRASP65 signal channel in the orig-
stabilizes the actin cytoskeleton of neuronal processes by inducing n-cofilin
inal and discarding the signal extraneous to the GFP-positive cells (Movie S2).
phosphorylation at serine3. J. Neurosci. 29, 288–299.
Process diameters were measured 12 mm from the nucleus (Figure 2K). These
measurements were done using Image Examiner (Zeiss). Measurement of Cooper, J.A. (2008). A mechanism for inside-out lamination in the neocortex.
dendrite lengths was done using the softWoRx (AppliedPrecision). Trends Neurosci. 31, 113–119.
D’Arcangelo, G., Miao, G.G., Chen, S.C., Soares, H.D., Morgan, J.I., and
SUPPLEMENTAL INFORMATION Curran, T. (1995). A protein related to extracellular matrix proteins deleted in
the mouse mutant reeler. Nature 374, 719–723.
Supplemental Information includes Extended Experimental Procedures, four D’Arcangelo, G., Homayouni, R., Keshvara, L., Rice, D.S., Sheldon, M., and
figures, and three movies and can be found with this article online at doi: Curran, T. (1999). Reelin is a ligand for lipoprotein receptors. Neuron 24,
10.1016/j.cell.2010.10.029. 471–479.

Cell 143, 826–836, November 24, 2010 ª2010 Elsevier Inc. 835
de Anda, F.C., Pollarolo, G., Da Silva, J.S., Camoletto, P.G., Feiguin, F., and on Cajal-Retzius neurons is a crucial molecule for laminar organization of
Dotti, C.G. (2005). Centrosome localization determines neuronal polarity. cortical neurons. Neuron 14, 899–912.
Nature 436, 704–708. Olson, E.C., Kim, S., and Walsh, C.A. (2006). Impaired neuronal positioning
de Anda, F.C., Meletis, K., Ge, X., Rei, D., and Tsai, L.H. (2010). Centrosome and dendritogenesis in the neocortex after cell-autonomous Dab1 suppres-
motility is essential for initial axon formation in the neocortex. J. Neurosci. sion. J. Neurosci. 26, 1767–1775.
30, 10391–10406. Pfenninger, K.H. (2009). Plasma membrane expansion: a neuron’s Herculean
Dotti, C.G., and Banker, G.A. (1987). Experimentally induced alteration in the task. Nat. Rev. Neurosci. 10, 251–261.
polarity of developing neurons. Nature 330, 254–256.
Preisinger, C., Short, B., De Corte, V., Bruyneel, E., Haas, A., Kopajtich, R.,
Efimov, A., Kharitonov, A., Efimova, N., Loncarek, J., Miller, P.M., Andreyeva, Gettemans, J., and Barr, F.A. (2004). YSK1 is activated by the Golgi matrix
N., Gleeson, P., Galjart, N., Maia, A.R., McLeod, I.X., et al. (2007). Asymmetric protein GM130 and plays a role in cell migration through its substrate 14-3-
CLASP-dependent nucleation of noncentrosomal microtubules at the trans- 3zeta. J. Cell Biol. 164, 1009–1020.
Golgi network. Dev. Cell 12, 917–930.
Puthenveedu, M.A., Bachert, C., Puri, S., Lanni, F., and Linstedt, A.D. (2006).
Fidalgo, M., Fraile, M., Pires, A., Force, T., Pombo, C., and Zalvide, J. (2010). GM130 and GRASP65-dependent lateral cisternal fusion allows uniform
CCM3/PDCD10 stabilizes GCKIII proteins to promote Golgi assembly and cell Golgi-enzyme distribution. Nat. Cell Biol. 8, 238–248.
orientation. J. Cell Sci. 123, 1274–1284.
Rice, D.S., Nusinowitz, S., Azimi, A.M., Martı́nez, A., Soriano, E., and Curran, T.
Förster, E., Bock, H.H., Herz, J., Chai, X., Frotscher, M., and Zhao, S. (2010). (2001). The reelin pathway modulates the structure and function of retinal
Emerging topics in Reelin function. Eur. J. Neurosci. 31, 1511–1518. synaptic circuitry. Neuron 31, 929–941.
Goffinet, A.M. (1984). Events governing organization of postmigratory
Rosso, S., Bollati, F., Bisbal, M., Peretti, D., Sumi, T., Nakamura, T., Quiroga,
neurons: studies on brain development in normal and reeler mice. Brain Res.
S., Ferreira, A., and Cáceres, A. (2004). LIMK1 regulates Golgi dynamics, traffic
319, 261–296.
of Golgi-derived vesicles, and process extension in primary cultured neurons.
Hiesberger, T., Trommsdorff, M., Howell, B.W., Goffinet, A., Mumby, M.C., Mol. Biol. Cell 15, 3433–3449.
Cooper, J.A., and Herz, J. (1999). Direct binding of Reelin to VLDL receptor
Rubinson, D.A., Dillon, C.P., Kwiatkowski, A.V., Sievers, C., Yang, L., Kopinja,
and ApoE receptor 2 induces tyrosine phosphorylation of disabled-1 and
J., Rooney, D.L., Zhang, M., Ihrig, M.M., McManus, M.T., et al. (2003). A lenti-
modulates tau phosphorylation. Neuron 24, 481–489.
virus-based system to functionally silence genes in primary mammalian cells,
Horton, A.C., Rácz, B., Monson, E.E., Lin, A.L., Weinberg, R.J., and Ehlers, stem cells and transgenic mice by RNA interference. Nat. Genet. 33, 401–406.
M.D. (2005). Polarized secretory trafficking directs cargo for asymmetric
Sanada, K., Gupta, A., and Tsai, L.H. (2004). Disabled-1-regulated adhesion of
dendrite growth and morphogenesis. Neuron 48, 757–771.
migrating neurons to radial glial fiber contributes to neuronal positioning during
Howell, B.W., Hawkes, R., Soriano, P., and Cooper, J.A. (1997). Neuronal posi- early corticogenesis. Neuron 42, 197–211.
tion in the developing brain is regulated by mouse disabled-1. Nature 389,
Sanchez-Ponce, D., Tapia, M., Muñoz, A., and Garrido, J.J. (2008). New role of
733–737.
IKK alpha/beta phosphorylated I kappa B alpha in axon outgrowth and axon
Howell, B.W., Herrick, T.M., Hildebrand, J.D., Zhang, Y., and Cooper, J.A. initial segment development. Mol. Cell. Neurosci. 37, 832–844.
(2000). Dab1 tyrosine phosphorylation sites relay positional signals during
mouse brain development. Curr. Biol. 10, 877–885. Shelly, M., Cancedda, L., Heilshorn, S., Sumbre, G., and Poo, M.M. (2007).
LKB1/STRAD promotes axon initiation during neuronal polarization. Cell
Jacobs, T., Causeret, F., Nishimura, Y.V., Terao, M., Norman, A., Hoshino, M.,
129, 565–577.
, M. (2007). Localized activation of p21-activated kinase controls
and Nikolic
neuronal polarity and morphology. J. Neurosci. 27, 8604–8615. Sütterlin, C., and Colanzi, A. (2010). The Golgi and the centrosome: building
a functional partnership. J. Cell Biol. 188, 621–628.
Kishi, M., Pan, Y.A., Crump, J.G., and Sanes, J.R. (2005). Mammalian SAD
kinases are required for neuronal polarization. Science 307, 929–932. ten Klooster, J.P., Jansen, M., Yuan, J., Oorschot, V., Begthel, H., Di Giacomo,
V., Colland, F., de Koning, J., Maurice, M.M., Hornbeck, P., and Clevers, H.
Lizcano, J.M., Göransson, O., Toth, R., Deak, M., Morrice, N.A., Boudeau, J.,
(2009). Mst4 and Ezrin induce brush borders downstream of the Lkb1/Strad/
Hawley, S.A., Udd, L., Mäkelä, T.P., Hardie, D.G., and Alessi, D.R. (2004).
Mo25 polarization complex. Dev. Cell 16, 551–562.
LKB1 is a master kinase that activates 13 kinases of the AMPK subfamily,
including MARK/PAR-1. EMBO J. 23, 833–843. Trommsdorff, M., Borg, J.P., Margolis, B., and Herz, J. (1998). Interaction of
cytosolic adaptor proteins with neuronal apolipoprotein E receptors and the
Lowe, M., Rabouille, C., Nakamura, N., Watson, R., Jackman, M., Jämsä, E.,
amyloid precursor protein. J. Biol. Chem. 273, 33556–33560.
Rahman, D., Pappin, D.J., and Warren, G. (1998). Cdc2 kinase directly phos-
phorylates the cis-Golgi matrix protein GM130 and is required for Golgi Trommsdorff, M., Gotthardt, M., Hiesberger, T., Shelton, J., Stockinger, W.,
fragmentation in mitosis. Cell 94, 783–793. Nimpf, J., Hammer, R.E., Richardson, J.A., and Herz, J. (1999). Reeler/
Marra, P., Salvatore, L., Mironov, A., Jr., Di Campli, A., Di Tullio, G., Trucco, A., Disabled-like disruption of neuronal migration in knockout mice lacking the
Beznoussenko, G., Mironov, A., and De Matteis, M.A. (2007). The biogenesis of VLDL receptor and ApoE receptor 2. Cell 97, 689–701.
the Golgi ribbon: the roles of membrane input from the ER and of GM130. Mol. Witte, H., Neukirchen, D., and Bradke, F. (2008). Microtubule stabilization
Biol. Cell 18, 1595–1608. specifies initial neuronal polarization. J. Cell Biol. 180, 619–632.
Matsuki, T., Pramatarova, A., and Howell, B.W. (2008). Reduction of Crk and Yadav, S., Puri, S., and Linstedt, A.D. (2009). A primary role for Golgi posi-
CrkL expression blocks reelin-induced dendritogenesis. J. Cell Sci. 121, tioning in directed secretion, cell polarity, and wound healing. Mol. Biol. Cell
1869–1875. 20, 1728–1736.
Nichols, A.J., and Olson, E.C. (2010). Reelin promotes neuronal orientation and Ye, B., Zhang, Y., Song, W., Younger, S.H., Jan, L.Y., and Jan, Y.N. (2007).
dendritogenesis during preplate splitting. Cereb. Cortex 20, 2213–2223. Growing dendrites and axons differ in their reliance on the secretory pathway.
Niu, S., Renfro, A., Quattrocchi, C.C., Sheldon, M., and D’Arcangelo, G. (2004). Cell 130, 717–729.
Reelin promotes hippocampal dendrite development through the VLDLR/ Yin, D.M., Huang, Y.H., Zhu, Y.B., and Wang, Y. (2008). Both the establishment
ApoER2-Dab1 pathway. Neuron 41, 71–84. and maintenance of neuronal polarity require the activity of protein kinase D in
Niwa, H., Yamamura, K., and Miyazaki, J. (1991). Efficient selection for high- the Golgi apparatus. J. Neurosci. 28, 8832–8843.
expression transfectants with a novel eukaryotic vector. Gene 108, 193–199. Zmuda, J.F., and Rivas, R.J. (1998). The Golgi apparatus and the centrosome
Ogawa, M., Miyata, T., Nakajima, K., Yagyu, K., Seike, M., Ikenaka, K., are localized to the sites of newly emerging axons in cerebellar granule
Yamamoto, H., and Mikoshiba, K. (1995). The reeler gene-associated antigen neurons in vitro. Cell Motil. Cytoskeleton 41, 18–38.

836 Cell 143, 826–836, November 24, 2010 ª2010 Elsevier Inc.
Resource

A Human Genome Structural Variation


Sequencing Resource Reveals Insights
into Mutational Mechanisms
Jeffrey M. Kidd,1,4 Tina Graves,2 Tera L. Newman,1,5 Robert Fulton,2 Hillary S. Hayden,1 Maika Malig,1 Joelle Kallicki,2
Rajinder Kaul,1 Richard K. Wilson,2 and Evan E. Eichler1,3,*
1Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
2Washington University Genome Sequencing Center, School of Medicine, St Louis, MO 63108, USA
3Howard Hughes Medical Institute, Seattle, WA 98195, USA
4Present address: Department of Genetics, Stanford University, Stanford, CA 94305, USA
5Present address: iGenix, Seattle, WA 98110, USA

*Correspondence: eee@gs.washington.edu
DOI 10.1016/j.cell.2010.10.027

SUMMARY mation about the precise structure and location of identified vari-
ants. Because of their dependence on the reference genome,
Understanding the prevailing mutational mecha- array-based approaches preferentially detect deletions over
nisms responsible for human genome structural vari- insertions and are unable to directly detect copy-number-neutral
ation requires uniformity in the discovery of allelic events such as inversions. Higher-density array platforms give
variants and precision in terms of breakpoint delinea- a better estimation of variant sizes, but most breakpoints cannot
tion. We develop a resource based on capillary end be resolved at a scale finer than 50 bp regions (Conrad et al.,
2010b), while targeted next-generation sequencing approaches
sequencing of 13.8 million fosmid clones from 17
have difficulty resolving breakpoints within homologous
human genomes and characterize the complete
segments (Conrad et al., 2010a).
sequence of 1054 large structural variants corre- These methodological biases threaten to skew our under-
sponding to 589 deletions, 384 insertions, and 81 standing of the underlying mechanisms responsible for the
inversions. We analyze the 2081 breakpoint junctions formation of structural variation and limit our ability to compre-
and infer potential mechanism of origin. Three hensively discover and genotype this form of genetic variation.
mechanisms account for the bulk of germline struc- We resolve the breakpoints of 1054 structural variants based
tural variation: microhomology-mediated processes on capillary sequencing of clone inserts. The high-quality
involving short (2–20 bp) stretches of sequence sequence of contiguous variant haplotypes allows alternative
(28%), nonallelic homologous recombination (22%), structures to be included in future human genome assemblies
and L1 retrotransposition (19%). The high quality and provides the breakpoint resolution necessary to accurately
genotype these variants in sequence data generated from
and long-range continuity of the sequence reveals
next-generation sequencing platforms. The sequences and the
more complex mutational mechanisms, including
associated clones also provide a resource for assessing future
repeat-mediated inversions and gene conversion, methods for structural variation discovery.
that are most often missed by other methods, such
as comparative genomic hybridization, single nucle- RESULTS
otide polymorphism microarrays, and next-genera-
tion sequencing. The Human Genome Structural Variation Clone
Resource
INTRODUCTION The high quality of the reference human genome is due, in large
part, to the fact that it was assembled based on capillary
Despite significant advances in the discovery and genotyping of sequencing of individual large insert clones whose complete
human genome structural variation, only a small fraction of sequence was resolved prior to final genome assembly. This
common structural variation has been resolved at the sequence strategy allowed complex duplicated and repetitive regions to
level (Conrad et al., 2010b; Freeman et al., 2006; Itsara et al., be incorporated that were missed by other approaches (Istrail
2009; Kidd et al., 2008; Lam et al., 2010; McCarroll et al., et al., 2004; She et al., 2004). Since genome structural variation
2008b; Redon et al., 2006). The majority of human genome struc- is similarly biased to these regions, we proposed that developing
tural variation has been discovered with single nucleotide poly- clone libraries for a modest number of additional genomes would
morphism (SNP) microarrays and array comparative genomic serve as a valuable resource for characterizing complex and
hybridization (arrayCGH), approaches that provide limited infor- difficult-to-assay regions of genome structural variation (Eichler

Cell 143, 837–847, November 24, 2010 ª2010 Elsevier Inc. 837
et al., 2007). The overall strategy involved the construction of 1994; Richard et al., 2008). VNTR repeat units ranged from
individual genome libraries using a fosmid cloning vector 17 bp to 6.5 kb with copy numbers ranging from 1 to 319
(40 kb inserts) and capillary sequencing of the ends of the inserts copies. We identified 198 events (20% of the total insertions
to generate a high-quality end-sequence pair (ESP). Discrep- and deletions) that we classified as being the result of L1
ancies in the length and orientation of these mapped ESPs retrotransposition. Each of the 198 L1 elements associated with
with respect to the reference genome serve as signatures of the retrotransposition events has a sequence identity of at least
copy-number variation and inversion, respectively. Since the 97.5% when compared to the L1.3 reference sequence, and
underlying clones can be retrieved, the complete sequence 152 are at least 6 kb in size, consistent with full-length elements
context of the discovered structural variant can also be obtained. that may be capable of subsequent retrotransposition (Beck
Previously, we discovered and cloned 1695 structural variants et al., 2010). We find evidence for transduction of flanking
with fosmid libraries derived from nine individuals and presented sequence for 20% (40/198) of the sites, with the transduced
sequence of 261 structural variants (Kidd et al., 2008; Tuzun segment size ranging from 45 to 968 nucleotides (median of
et al., 2005). We expand this resource to include capillary end 81.5) (Goodier et al., 2000; Moran et al., 1999; Pickeral et al.,
sequencing of 4.1 million additional fosmid clones from eight 2000). Using the transduced sequence as a marker, we identi-
additional human genomes (Table S1, available online). fied the potential donor location for 30 of these retrotransposi-
The combined set includes 13.8 million clones derived from the tions (20 insertions in the fosmid source sample and 10
genomes of six Yoruba Nigerians, five CEPH Europeans, three insertions in the reference genome). We identified three posi-
Japanese, two Han Chinese, and one individual of unknown tions that have each given rise to multiple LINE insertions (Fig-
ancestry. ure 2B), suggesting the presence of L1 donor hotspots. We
note that 11 of the 20 L1 insertions in the fosmid source
Structural Variant Alleles (including the three recurrent L1 donors) correspond to
Using this resource, we searched for clusters of clones that elements that have been functionally determined to represent
suggest a structural difference when compared to the reference. hot L1s, according to assays performed by Beck et al. (2010).
We discovered a total of 2051 discordant regions (Table S1) We found two events consistent with the insertion of an intact
having support from multiple clones for a structure different HERV-K element: one insertion in the reference sequence (as
from the reference genome. The size distribution of the fosmid indicated by clone AC209281) and an insertion contained in
clone inserts limited us to the detection of structural variants clone AC226770. Both events showed less than 1% divergence
greater than 5 kb in length. Inversions also tend to be biased to from the HERV-K sequence (Dewannieux et al., 2006) and were
larger events because of the probability of capturing a breakpoint flanked by long terminal repeats (Tristem, 2000). Our discovery
by a pair of end sequences. While there is no upper bound in the size thresholds (>5 kb) preclude the identification of smaller ret-
detection of deletions and inversions, the direct capturing of rotransposition events arising from SVA or Alu repeats that are
insertions larger than the insert size of the clone (40 kb) requires common when smaller structural variants are considered (Ben-
specialized approaches. For example, new tandem duplications nett et al., 2008; Korbel et al., 2007; Lam et al., 2010; Mills et al.,
may be identified with an everted clone mapping signature (Fig- 2006).
ure S1) (Cooper et al., 2008) and insertions of novel human We divided the remaining 824 structural variants into two
sequence may be identified by read pairs for which only one broad categories. Class I consists of variants with no additional
end maps (Kidd et al., 2010). sequence at the breakpoint junction (Figures 4A–4D and
We targeted 1054 structural variants (Table S1) from nine Figure S2). Class II variants contain an additional sequence,
human genomes and completely sequenced the inserts of found across the variant junction, that is not present at either
1167 fosmid clones (46.4 Mb of sequence). We identified 81 of the other variant breakpoints (Figures 4E–4G). We also
loci for which breakpoints could not be resolved because of diffi- assessed the presence of extended sequence homology
culty in clone assembly and the limits of 40 kb fosmid inserts (see and the extent of matching sequence at the breakpoints. We
Supplemental Experimental Procedures). We defined break- note that microhomology is a qualitative term without clear delin-
points relative to the reference genome assembly following eation as 1 or 2 bp matches are expected to occur often by
a two-stage procedure (Kidd et al., 2010) (Figure 1 and Table chance (Figure 4) and a range of homologous match lengths is
S2). We initially distinguished copy-number changes (n = 973 observed (Conrad et al., 2010a; Lam et al., 2010). Similarly, there
insertion and/or deletions) from balanced genome structural is ambiguity in assigning events to potential mechanisms based
variants (81 inversions) (Figure 2). The analyzed variants altered solely on the length of homologous segments. Consequently, we
95 gene structures. We estimate that 1.04% (11/1054) of the categorize events based on observed ranges of homology and
sequenced alleles are already known risk factors for common consider assignment to specific mechanisms as speculative.
and rare human diseases (Figure 3 and Table S3). Among the class I events, 49% (289/590) of copy-number
variants contain 2–20 bp of matching sequence, indicating that
Breakpoint Features microhomology-mediated mechanisms, such as microhomol-
Using the 40 kb of clone-based sequence, we examined the ogy-mediated end joining (MMEJ), contribute to a substantial
sequence features and inferred potential mechanism of origin fraction (30%) of human structural variation (Table 1) (Hastings
for these variants (Table 1). We identified 30 variants associated et al., 2009; McVey and Lee, 2008; Payen et al., 2008; Roth
with the expansion or contraction of a variable number of and Wilson, 1986). Although there is large overlap in the variant
tandem repeats (VNTRs) (Buard et al., 2000; Jeffreys et al., size when broken down by extent of homologous sequence

838 Cell 143, 837–847, November 24, 2010 ª2010 Elsevier Inc.
A C

10 kb deletion

break 1 break 2

deletion junction

B
Align against common
junction sequence Combine pairwise
alignments Assess identity
|||||||| |||--||--|| |||||||| |||--||--||
|||||||| |||--||--|| |||||||| |||
|||| |||--||--||
| || ||

| |----| || |||| |||| | |----| || |||| ||||


| |----| || |||||||| | |----| || |||| ||||
*1*1111* **122** **22

Figure 1. Sequence and Breakpoint Analyses


Variant breakpoints were defined based on alignments of sequences from the sequenced insertion and deletion alleles. For example, (A) the sequence of fosmid
clone AC207429 is compared with sequence from the corresponding region on chr2. A 10 kb deletion, relative to the reference sequence, is readily apparent
(indicated by the red bracket). The position of segmental duplications, common repeats (LINEs are green, SINEs are purple, and LTR elements are orange),
and RefSeq exons are shown. Sequence segments corresponding to three different breakpoint regions (red, green, and purple bars) are extracted for further
analysis.
(B) The sequence across the variant junction is aligned against each of the other two sequences and the resulting pairwise alignments are merged. The pattern of
sequence identity is assessed to identify the positions where the junction sequence switches from being a better match to the first breakpoint to being a better
match to sequence from the second breakpoint. The breakpoint coordinates correspond to the innermost positions that can be confidently assigned to be before
and after the variant boundary.
(C) The result of aligning the three segments depicted in (A). Alignment columns where the junction sequence matches the sequence from the first (leftmost)
breakpoint are indicated by a 1 while alignment columns where the junction sequence matches the second (rightmost) breakpoint are indicated by a 2. Positions
where all three sequences are the same are indicated by an asterisk (*). The red square highlights the position of the breakpoint coordinates (highlighted in red and
green text). The two breakpoints are separated by seven nucleotides found at both breakpoints with perfect identity (blue text). Highlighted in gray is a 293 bp
segment present at both breakpoints with a sequence identity of 91%. See also Tables S2 and S7.

(Figure 4C), we find that, as a class, the mean size of events size. Overall, we find that younger Alu events and segmental
associated with microhomology (2–20 bp of matching sequence, duplications contribute most significantly to the process of
n = 289, mean size is 9.7 kb) is significantly smaller (p = 0.02926, NAHR (Table S4), as expected because of their higher levels of
two sample t test) than those showing a hallmark of nonallelic sequence identity. The strongest enrichment is found for paired
homologous recombination (NAHR) (R200 bp of matching Alu repeats at each breakpoint (5.2-fold enrichment). If each
sequence, n = 177, mean size is 21.0 kb). The analyzed inver- breakpoint is treated separately, rather than requiring that an
sions are overwhelming driven by large homologous segments element of the same subfamily be present at both breakpoints
with 69% (56/81) of all analyzed inversions containing stretches of a variant, then AluY also shows a substantial degree of enrich-
of matching sequence at least 200 bp in length. In contrast, only ment (2.6-fold, Table S4). Since AluY is the most recently active
30% (177/590) of the class I copy-number variants contain Alu family, dispersed AluY elements are expected to have
matching breakpoint sequences of at least that length. It is a higher degree of sequence identity than other Alu families (Bat-
important to note, however, that our clone end-sequence zer and Deininger, 2002; Cordaux and Batzer, 2009). Closer
mapping strategy is biased toward the detection of larger inver- examination of the distribution of breakpoints within individual
sions when compared to copy-number variants. This is a direct Alus reveals a nonuniform pattern of breakpoint density (Fig-
consequence of the probability of capturing a breakpoint that ure 3D). The highest density of breakpoints occurs near the posi-
diminishes when inversions become smaller than the clone insert tion of a sequence motif (CCNCCNTNNCCNC) that has been

Cell 143, 837–847, November 24, 2010 ª2010 Elsevier Inc. 839
A B

Figure 2. Sequenced Structural Variant Alleles


(A) Size distribution for 1054 sequenced structural variants. Insertions, deletions, and inversions relative to the genome reference assembly are depicted sepa-
rately. Note that the bins are not of equal sizes. The mean size of the sequenced variants is 14.9 kb for deletions, 6.1 kb for insertions, and 196 kb for inversions.
Our variant selection methodology largely identifies deletions greater than 5 kb and insertions from 5 kb to 40 kb in size and is biased against inversions
smaller than 40 kb.
(B) The relationship between the donor site of transduced sequences and LINE insertion position are given for 30 events with a match to hg18 using BLAT. Rela-
tionships are shown for 20 LINE insertions in library source individuals relative to the reference (blue lines) and for 10 insertions in the genome reference (red lines).
The blue circles represent three different loci associated with multiple distinct LINE insertions. See also Figure S1 and Table S1.

associated with meiotic recombination hotspots, is found in stretches of homologous sequence flanking the breakpoint
some Alu elements (Myers et al., 2008), and has also been insertion confirming they arose by mechanisms other than
observed for rearrangements between human and chimpanzee NAHR. Interestingly, if we examine the sequence context of
(Han et al., 2007; Sen et al., 2006). these regions, we find that 20% (30/153) of class II events
We find that 16% (153/973) of the insertion and deletion vari- map within 5 kb of a segmental duplication. This represents
ants and 9% (7/81) of the inversions contain additional a significant enrichment for proximity to duplicated sequence
sequence at the variant breakpoints (class II events; Figure 4). (p < 0.002 based on comparisons with randomly sampled
Many of the additional insertion sequences are relatively short sequences) indicating that regions flanking segmental duplica-
in length, consistent with nontemplate-directed repair associ- tions may be generally more unstable and susceptible to
ated with nonhomologous end joining (Figure 4B). For these multiple mutational processes such as template switching
shorter sequences, no inference could be made as to the source during replication (Itsara et al., 2009; Lee et al., 2007; Payen
of the additions. However, 41% of all class II variants (66/160) et al., 2008).
contain additional sequence at the junction at least 20 bp in
length. Of these longer fragments, 88% (58/66) map to another Gene Conversion and Structural Variation
location within the human genome. Since we are limited in this During our analysis of putative NAHR events, we identified
study to directly capturing the breakpoints of insertions smaller 10 structural variants having a complex pattern of exchange
than 40 kb, we repeated this comparison with only deletions inconsistent with a simple model of unequal crossover. The
relative to the assembly where we expect to have less of breakpoint region contains an interleaved pattern of alternating
a bias in terms of variant size. We find that the additional junction patches of sequences from flanking homologous segments
sequences for 30 of 39 class II deletion events at least 20 bp (Figure 5). These patterns are reminiscent of multiple rounds of
long map elsewhere in the genome. Seventy-three percent gene conversion, although each of these events was also asso-
(22/30) are found on the same chromosome as the variant. ciated with a copy-number variant event. Using paralogous
In fact, eight of the insertions map less than 1 kb away from sequence variants that distinguish the 50 and 30 homologous
the variant breakpoint (Figure 4G and Table S5) and all 22 are segments, we investigated the overall extent of this nonallelic
less than 250 kb from the breakpoint. This pattern suggests exchange (referred to as the conversion tract length), and the
the action of a replication-associated process that involves number of switches before unambiguous homology to the 50 or
template switching or strand invasion (Hastings et al., 2009; 30 end was re-established. We determined that most (6/10) of
Lee et al., 2007; Smith et al., 2007). In contrast to the class I the conversion tracts were relatively short (200–600 bp in length)
events, only 2% of the class II events (3/160) contained with a relatively consistent number (4–6) and length (30–40 bp) of

840 Cell 143, 837–847, November 24, 2010 ª2010 Elsevier Inc.
A C
NEGR1 MST150
IRGM

chr1 chr5

AC210916 AC207974
10 20 30 40 kb 10 20 30 40 kb
Repeats

SegDupMasker

TMEM50A
B D
C1orf63 RHD LCE3D LCE3C LCE3B LCE3A
LCE3E

chr1 chr1

AC196511 AC196522
10 20 30 40 kb 10 20 30 40 kb
0.0
Repeats

SegDupMasker

Figure 3. Examples of Sequenced Variants


Examples of the complete sequence of structural variant alleles that have been associated with disease risk, including (A) a 45.5 kb deletion upstream of NEGR1,
(B) a 72 kb deletion of RHD, (C) a 3.9 kb and a 20.1 kb deletion upstream of IRGM, and (D) a 32 kb deletion of LCE3C. See also Table S3.

switches before clear boundaries at the 50 and 30 could be re-es- Comparison with Other Genome-wide Studies
tablished (Figure S3). Seven of these events have breakpoints and Ascertainment Biases
that map within segmental duplications, and the remaining three In this study we focused on systematically characterizing large
have breakpoints that map within LINEs. Three of the variants structural variants at the single base-pair level. In order to identify
contained at least ten switches. One variant (AC212911) showed events that may have been missed by the fosmid ESP approach,
the largest associated conversion tract with a remarkable 182 we compared our set of structural variants to other studies that
switches extending over 7.9 kb (Figure 5D). We sequenced the have discovered and genotyped copy-number variants in the
deletion allele with fosmids derived from three different individ- same DNA samples. We focused on five individuals analyzed
uals for one event (AC226182). Each of the three deletion haplo- by fosmid end sequencing (Kidd et al., 2008), Affymetrix 6.0 mi-
types contained identical patterns of interleaved sequence, croarray (McCarroll et al., 2008b), and high-density oligonucleo-
a finding that is consistent with the creation of the pattern at tide arrayCGH (Conrad et al., 2010b). A comparison of the three
the time of variant formation, or shortly thereafter, rather than studies shows that 11%–65% of discovered variants are unique
as a result of a continual conversion process between deletion to a single study and corresponding experimental platform (Fig-
and insertion alleles leading to a diverse set of related molecules ure 6). The limited overlap should not be surprising since each
over time (Figure S3). It is also possible that the conversion approach preferentially identifies a subset of the total collection
pattern arose before the formation of the structural variant and of genomic variation. For example, the fosmid ESP mapping
that the pattern we observe in sequenced variants is merely approach can detect insertions of sequence not represented in
incidental or the result of a series of mismatch repair processes the genome assembly (Kidd et al., 2008, 2010), as well as
prior to variant formation. Nevertheless, the observed switch balanced events such as inversions (not depicted in Figure 6),
pattern is reminiscent of patterns of toggling previously whereas array approaches can more readily detect copy-
observed at some LINE insertions (Gilbert et al., 2005, 2002; number variation caused by large duplications.
Symer et al., 2002) and suggests a mechanism of serial strand Differences in ascertainment extend to the resolution of break-
invasion/repair during the rearrangement process. point sequences. The sequenced variants described in this

Cell 143, 837–847, November 24, 2010 ª2010 Elsevier Inc. 841
Table 1. Summary of Events and Inferred Mechanisms
Event Classification Insertions and Deletions Inversions Potential Mechanisms
Retroelements
L1 198 (20.3%) NA Retrotransposition
HERV-K 2 (0.2%) NA Retrotransposition
VNTR 30 (3.1%) Minisatellite, NAHR
Class I (no additional sequence at breakpoint) 590 (60.6%) 74 (91.3%)
0 or 1 matching nucleotides 82 (8.4%) 10 (12.3%) NHEJ
2–20 matching nucleotides 289 (29.7%) 8 (9.9%) NHEJ, MMEJ
21–100 matching nucleotides 28 (2.9%) 0 NAHR, other
101–199 matching nucleotides 14 (1.4%) 0 NAHR, other
R200 (NAHR) 177 (18.2%) 56 (69.1%) NAHR
Class 2 (additional sequence at breakpoint) 153 (15.7%) 7 (8.6%)
1–10 additional nucleotides 76 (7.8%) 2 (2.5%) NHEJ
>10 additional nucleotides 77 (7.9%) 5 (6.2%) NHEJ, FoSTeS,template switching
Total 973 81
The number of events that fall into each breakpoint class is given. The following abbreviations are used: NHEJ, nonhomologous end joining; FoSTeS,
fork stalling and template switching. See also Table S6.

manuscript include 237 of the regions targeted for array capture with segmental duplications, including ten examples of tandem
and 454 sequencing (Conrad et al., 2010a). Seventy of these duplications. We note that 23 of these duplication-containing
targeted events were successfully resolved by breakpoint loci map near gaps in the National Center for Biotechnology
array-capture experiments (Table S6), with none of the events Information (NCBI) build36 genome assembly or to sequences
containing extended breakpoint homology successfully resolved that have been assigned to a chromosome but not fully inte-
by next-generation sequencing. grated into the genome reference sequence. Duplication-medi-
We also reassessed regions discovered by other studies that ated copy-number variation remains underascertained in terms
were missed by the fosmid ESP approach. With the standard of sequence-level resolution of variant haplotypes and muta-
fosmid analysis criteria (two or more discordant clones with suffi- tional mechanism analysis. If we adjust for these biases, we esti-
cient quality) (Tuzun et al., 2005), an overlapping deletion site is mate that the fosmid ESP approach has minimally missed at
only identified for 53% (631/1193) of the corresponding deletion least 106 structural variants associated with segmental
genotypes reported by Conrad et al. (2010b). The intersection duplications.
rate increases to 75% (900/1193 sample-level genotypes) if indi-
vidual deletion clones are considered with reduced quality DISCUSSION
thresholds. This suggests that much of the variation missed by
the fosmid ESP approach is a result of random fluctuations in We describe a clone resource from 17 human DNA samples that
the level of clone coverage and the quality of individual provides 135-fold physical coverage of the human genome. The
sequencing reads (Cooper et al., 2008). corresponding catalog and clones can be used to further charac-
Experimental approaches to discover structural variation can terize almost any segment of human euchromatin. We used this
have reduced sensitivity in regions of segmental duplication resource to assess breakpoint characteristics of 1054 events.
because of difficulty in uniquely mapping reads or designing The nature of our experimental design permitted us to discover
array probes (Cooper et al., 2008; Kidd et al., 2008; Tuzun more events mediated by larger segments of homology, providing
et al., 2005). We compared the validated structural variants a more complete assessment of human genetic variation. Of
from Kidd et al. (2008) with those found by read-depth particular interest are complex events whose sequence features
approaches (Alkan et al., 2009). Alkan et al. (2009) identified have been difficult to previously assess at a genome-wide level.
113 genes that differ in copy number among three individuals. The high quality and length of the sequenced fosmids combined
Only 38% of the genes greater than 5 kb (26/69) and identified with defined paralogous sequence events allowed us to quantify
as copy-number variable by read-depth intersect with a struc- alternating sequence matches suggestive of interlocus gene
tural variant (reported in Kidd et al.[2008]). This result indicates conversion (Bayés et al., 2003; Lagerstedt et al., 1997; Reiter
that even the fosmid ESP approach has underascertained et al., 1997; Visser et al., 2005).
copy-number variation associated with the most variable dupli- Using this resource, we obtained the complete structure of
cated sequences. several alleles that have been associated with disease, including
We identified 81 loci during our sequence analysis with a deletion variant upstream of the NEGR1 gene associated with
evidence for a nonreference structure for which we could not increased body mass index (Willer et al., 2009) (clone
unambiguously define the variant breakpoint (see Supplemental AC210916), two deletion polymorphisms upstream of the
Experimental Procedures). Of these 81 loci, 63 are associated IRGM gene associated with Crohn’s disease (Barrett et al.,

842 Cell 143, 837–847, November 24, 2010 ª2010 Elsevier Inc.
A B

C D

0.12

0.10

Breakpoint Density
0.08

0.06

0.04

0.02

50 100 150 200 250 300


Position in Alu

E G
5’bkpnt CAAATGCAATGTTTATTAAGCAGGTACTTTGTGCTCAAGAGTATGATACAGAGCACTAT
AC209239 CAAATGCAATGTTTATTAAGCAGGTACTTTGTGCTCAAGAGTATGATACAGAGCACTAT

5’bkpnt GCTGGG
AC209239 GCTGGGATTTGGCAGAGGGGGATTTGGCAGGGTCATAGGACAACAGCGGAGGGAAGGTC

AC209239 AGCTCAGGAGGCTTAGGCATGAGAATCACTTGAACCTGGTAGGCA
3’bkpnt CTCAGGAGGCTTAGGCATGAGAATCACTTGAACCTGGTAGGCA

Figure 4. Variant Breakpoint Analyses


(A–D) Class I variants are defined as those without additional nucleotides at the breakpoint. (A) A histogram of the extent of matching breakpoint sequence (black)
and extended breakpoint homology (gray) is shown for 590 class I copy-number events. The red line corresponds to the expected distribution of breakpoint
match lengths found from 100 random permutations. Note that bin sizes are not equal. The increase in extended homology segments 250–299 bp in length corre-
sponds to variants having Alus at their breakpoints. (B) As in (A) zoomed in to show variants having a matching sequence of 20 bp or less. (C) Box plot of variant
size partitioned by length of extended breakpoint homology for 590 class I copy-number variants (red line: median; blue box: interquartile range; whiskers: within
1.53 interquartile range). (D) Breakpoint density map within a consensus Alu repeat sequence based on 269 copy-number variant events (blue box: RNA pol III
promoter; black boxes: AT-rich segment between the two monomers that make up the Alu element and the poly A tail; purple box: position of motif
(CCNCCNTNNCCNC) found in some Alus and associated with recombination hotspots [Myers et al., 2008]).
(E–G) Class II variants contain additional sequence across the breakpoint junction. (E) A class II variant containing a 55 nucleotide-long stretch of additional
sequence (in blue) that is not found at either breakpoint. (F) Histogram of the length of additional sequence found at variant breakpoints (black) and the length
of detected extended homology between breakpoint sequences (gray) for 153 class II copy-number variants. (G) Genomic location for class II unmatched
sequences (>20 bp) associated with deletions. The black lines connect the positions of a class II deletion variant (relative to the genome assembly) and the cor-
responding location where the additional sequence across the variant breakpoint can be found. The relationship for 31 deletion variants is depicted. One event
involves a match to unlocalized sequence on chromosome 1 (chr1_rand). See also Figure S2 and Tables S4 and S5.

Cell 143, 837–847, November 24, 2010 ª2010 Elsevier Inc. 843
A Figure 5. Breakpoint Assessment Using Paralogous
Insertion Sequence Variants
Allele
(A) Schematic comparison of the structures of the insertion
and deletion haplotypes of a putative NAHR variant. The blue
Deletion and red boxes represent homologous sequences present at
Allele
the breakpoints, which mediate the rearrangement. The blue
and red vertical lines identify paralogous sequence variants
B that distinguish the 50 and 30 copy of the matching sequence.
AC216822
Scanning along the deletion allele, which is missing the inter-
AC216064 vening sequence, one observes single nucleotides specific
with the 50 breakpoint, followed by a stretch of sequence
AC206476
that matches both, then sequences that match the 30 break-
point.
1 200 400 600 800 1000
(B) Representation for three variants showing a classic NAHR
pattern. Each line represents the deletion allele corresponding
to the indicated variant. We note a single unexpected paralo-
C gous sequence variant mismatch located 145 bp past the 30
AC212994
breakpoint, which could correspond to a SNP, short gene
AC225624
conversion, or alignment artifact because of the placement
of indels between 50 and 30 segments.
AC225305 (C) Representation of four variants having breakpoints that
show a pattern of alternating sequences that match the 30
AC203608 then 50 breakpoints.
(D) An extreme pattern of alternating matches that contains
1 200 400 600 800 1000 1200 1400 1600 1800 182 switches spanning over a 7.9 kb interval.
(E) Rearrangements associated with gene conversion. See
D also Figure S3.
AC212911

1 1000 2000 3000 4000 5000 6000 7000 8000

E the variants are uniquely identifiable with a read


Number of Conversion Variant
Accession
switches tract (bp) Size (kb) length of 36 bp and uniqueness threshold permit-
AC225832 4 2,632 27.6
AC225305 4 632
ting up to one substitution. This includes 32 inver-
6.7
AC216797 4 250 8.7 sions—balanced events that are invisible to array-
AC215992 4 116 16.2 based genotyping approaches. As read lengths
AC211399 6 211 10.1
AC212994 6 205
increase to 100 bp, we estimate that 88% (902/
3.9
AC226182 6 122 108.7 1024) of these variants could be genotyped. The
AC203608 10 1,249 20.6 construction of complete alternative haplotypes
AC225624 14 454 5.9
then facilitates the use of read-pair information to
AC212911 182 7,899 30.7
distinguish among distinct structural configurations
(Antonacci et al., 2010).
Although, short read technologies may miss
some of the breakpoint sequences, there are
2008; Bekpen et al., 2009; McCarroll et al., 2008a) (clone many advantages to the application of short read technology
AC207974), and the deletion of the LCE3B and LCE3C genes. to genome structural variation. This includes the detection of
In total, we conservatively estimate that 1.04% (11/1,054) of thousands more events per individual genome, especially vari-
the discovered variants are associated with disease. This yield ants below the detection threshold of the fosmid ESP approach.
of disease-causing alleles rivals that found by genome-wide The dynamic range response and the sequence specificity of
association studies using SNPs, which have identified 779 next-generation sequencing allow absolute copy number and
genome-wide associations based on genotyping of at least the identity of duplicated genes to be accurately predicted.
100,000 SNPs (http://www.genome.gov/multimedia/illustrations/ One of the strengths of this clone resource, however, is that it
GWAS2010-3.pdf). permits the iterative assessment of predicted variants. Clones
Although the functional significance of many of the other struc- may be retrieved corresponding to structural variants discovered
tural variants remains to be determined, the clone resource and by other methods applied to these 17 individuals, including
availability of the complete sequence of variant haplotypes will newly developed approaches such as methods for identifying
facilitate future disease association through the rapid design of transposon insertions (Huang et al., 2010; Witherspoon et al.,
assays to test for association with disease (Abe et al., 2009; An 2010). Sequencing would provide complete information
et al., 2009; Kidd et al., 2007) or direct comparison with short regarding the structure of additional events, thereby providing
sequencing reads from next-generation sequence platforms a resource set of sequenced variant haplotypes. The availability
(Kidd et al., 2010; Lam et al., 2010). of the underlying clones and potential location of the variant
We investigated this approach for 1024 non-VNTR sequenced within a specific DNA sample provides an approach for more
structural variants (Table S7) and found that 71% (726/1024) of fully exploring the genetic architecture and mutational properties

844 Cell 143, 837–847, November 24, 2010 ª2010 Elsevier Inc.
(Parsons, 1995) with a match threshold of s 400. Images summarizing these
Conrad et al.
Kidd et al. comparisons that included annotations of the repeat content, predicted and
N=1,128
N=1,206 observed segmental duplications (with DupMasker [Jiang et al., 2008]), and
RefSeq exons were prepared and examined to identify clones harboring
a structural difference relative to the build36. Clones that mapped to unas-
signed or random parts of the reference genome or that do not contain an
790 283 634 entire event (such as clones that contain one edge of a tandem duplication)
278 were omitted from analysis. Approximate variant breakpoints were determined
utilizing the context provided by long stretches of contiguous matching
sequence. In many cases, the pattern of common repeats or segmental dupli-
cations was a useful aid in this assessment.
128 For each variant, three sequences were extracted and aligned. In the case of
132 a deletion, two sequences at the variant boundaries are extracted from the
130 genome assembly and one sequence (termed the deletion junction sequence)
84
is extracted from the clone. For insertions, the junction sequence is extracted
5 76
5 from the genome assembly and two sequences corresponding to the variant
25 boundaries in the fosmid clone are extracted. For inversions, a single break-
point is directly captured in the sequenced clone. However, the position of
McCarroll et al. the other breakpoint can be inferred based on a comparison with the genome
N=236 assembly. Thus, for inversions, two sequences are extracted from the
assembly at the edges of the inferred inversion and the third sequence is
Figure 6. Comparison of Events Detected from Three Studies extracted from the clone. For inversion analysis, one of the chromosome-
Only variants estimated to be >5 kb are included. The Kidd et al. (2008) set derived segments is reverse-complimented prior to alignment.
includes sites of insertion or deletion in one of the five samples relative to An alignment is then constructed from the extracted breakpoint segments
the genome assembly; the Conrad et al. (2010b) set includes gains and losses (Kidd et al., 2010). First, an optimal global alignment is computed between
in at least one of the five samples relative to a reference arrayCGH sample; and the junction fragment and each of the other two fragments with the program
the McCarroll et al. (2008b) set includes CNVs that were successfully geno- needle with default parameters (Rice et al., 2000). These alignments are then
typed on the Affymetrix 6.0 platform and are variable among the five included merged to yield a single, three-sequence alignment. From this alignment,
samples. Prior to comparison, the variant sets within each study were merged the innermost positions that can be confidently assigned to be before and after
into a single, nonredundant interval set, and any overlap among regions the structural variant are identified. The resulting positions are used to define
between studies was sufficient regardless of which sample a variant was membership as a class I or class II variant and correspond to the breakpoint
detected in. match length depicted in Figure 4. Extended breakpoint homology was deter-
mined with both cross_match (http://www.phrap.org/, -minmatch 4 -max-
match 4 -minscore 20 -masklevel 100 -raw -word_raw) without complexity-
of these regions. Thus, we predict that such a resource will be adjusted scoring (Chiaromonte et al., 2002) and bl2seq (-W 7 -g F -F F -S 1
a valuable complement for understanding the true complexity -e 20) to identify the longest extent and identity of additional matching
of human genetic variation as human genomes become routinely sequence (termed extended breakpoint homology) that included the two
sequenced using short read sequencing technology. breakpoints. For putative NAHR events, we additionally determined the
longest stretch of 100% perfect identity as well as a parsimonious matching
metric to account for mutations after the time of variant formation (Figure S2).
EXPERIMENTAL PROCEDURES
VNTR and Retroelement Analysis
Identifying and Sequencing Variant Clones
Events associated with tandem repeats were characterized with the output
Sites of structural variation, relative to the reference genome assembly, were
from miropeats (Parsons, 1995), tandem repeats finder (Benson, 1999),
identified through fosmid ESP mapping. Briefly, genomic DNA was obtained
DupMasker (Jiang et al., 2008), and RepeatMasker (Smit et al., 1996–2004).
from transformed lymphoblastoid cell lines (available from the Coriell Cell
Potential L1 insertions were characterized with both the TSDfinder program
Repository) and approximately 1 million 40 kb fragments from each individual
(Szak et al., 2002) and the results of the breakpoint identification and charac-
were cloned into fosmid vectors. Paired end sequences were obtained from
terization process.
both ends of each fragment with standard capillary sequencing. The resulting
ESPs were mapped onto the reference assembly to identify clusters of multiple
clones from a single individual showing the same type of discordancy (Tuzun Genotyping Structural Variants with Diagnostic K-mers
et al., 2005). We previously identified 1695 structural variants that have been Diagnostic k-mers were identified for each variant (Table S7) by extracting
experimentally validated (Kidd et al., 2008). In this manuscript, we focus on overlapping k-mers of the indicated size across each sequenced breakpoint.
1054 events for which complete, finished clone sequence is available. High- K-mers were then searched against the build36 genome sequence and a set
quality finished sequence was obtained for all fosmid inserts with capillary- of sequenced fosmids with mrsFAST (http://mrfast.sourceforge.net/). To be
based shotgun sequencing and assembly with the procedures established considered diagnostic, a k-mer must be unique (within the given edit distance
for sequencing clones as part of the Human Genome Project. Some sequenced threshold) to the allele variant from which it was derived (Kidd et al., 2010).
clones contain gaps in simple sequence repeats that are not related to the
detected structural variants. For one individual, NA18956, additional clones ACCESSION NUMBERS
were selected with a relaxed threshold of two standard deviations larger or
smaller than the observed mean insert. In some cases, multiple clones were All sequence data have been deposited in GenBank under project ID 29893.
sequenced for a single event, whereas in other loci a single clone sequence
appeared to contain multiple distinct variants relative to the genome reference.
SUPPLEMENTAL INFORMATION

Identifying Variant Breakpoints Supplemental Information includes Supplemental Experimental Procedures,


Sequences of individual fosmid inserts were initially compared to the NCBI three figures, and eight tables and can be found with this article online at
build36 (UCSC hg18) genome reference assembly with the program miropeats doi:10.1016/j.cell.2010.10.027.

Cell 143, 837–847, November 24, 2010 ª2010 Elsevier Inc. 845
ACKNOWLEDGMENTS Conrad, D.F., Bird, C., Blackburne, B., Lindsay, S., Mamanova, L., Lee, C.,
Turner, D.J., and Hurles, M.E. (2010a). Mutation spectrum revealed by break-
We thank D. Smith and the staff at Agencourt Biosciences for library produc- point sequencing of human germline CNVs. Nat. Genet. 42, 385–391.
tion, E. Kirkness and staff of the J. Craig Venter Institute for end-sequence data Conrad, D.F., Pinto, D., Redon, R., Feuk, L., Gokcumen, O., Zhang, Y., Aerts,
from the JVCI library, and L. Chen for computational assistance in the mapping J., Andrews, T.D., Barnes, C., Campbell, P., et al; Wellcome Trust Case Control
of end-sequence data. We thank S. Girirajan, J. Moran, and C. Payen for Consortium. (2010b). Origins and functional impact of copy number variation in
thoughtful discussion; T. Brown for manuscript preparation assistance; and the human genome. Nature 464, 704–712.
members of the University of Washington and Washington University Genome
Cooper, G.M., Zerr, T., Kidd, J.M., Eichler, E.E., and Nickerson, D.A. (2008).
Centers for assistance with data generation. J.M.K. is supported by a National
Systematic assessment of copy number variant detection via genome-wide
Science Foundation Graduate Research Fellowship. This work was supported
SNP genotyping. Nat. Genet. 40, 1199–1203.
by the National Institutes of Health Grant HG004120 to E.E.E., who is an inves-
tigator of the Howard Hughes Medical Institute. E.E.E is on the scientific Cordaux, R., and Batzer, M.A. (2009). The impact of retrotransposons on
advisory board for Pacific Biosciences. T.L.N. is an employee and founder of human genome evolution. Nat. Rev. Genet. 10, 691–703.
iGenix Inc. Dewannieux, M., Harper, F., Richaud, A., Letzelter, C., Ribet, D., Pierron, G.,
and Heidmann, T. (2006). Identification of an infectious progenitor for the
Received: July 6, 2010 multiple-copy HERV-K human endogenous retroelements. Genome Res. 16,
Revised: September 15, 2010 1548–1556.
Accepted: October 15, 2010 Eichler, E.E., Nickerson, D.A., Altshuler, D., Bowcock, A.M., Brooks, L.D.,
Published: November 24, 2010 Carter, N.P., Church, D.M., Felsenfeld, A., Guyer, M., Lee, C., et al; Human
Genome Structural Variation Working Group. (2007). Completing the map of
REFERENCES human genetic variation. Nature 447, 161–165.
Freeman, J.L., Perry, G.H., Feuk, L., Redon, R., McCarroll, S.A., Altshuler,
Abe, H., Ochi, H., Maekawa, T., Hatakeyama, T., Tsuge, M., Kitamura, S., Ki-
D.M., Aburatani, H., Jones, K.W., Tyler-Smith, C., Hurles, M.E., et al. (2006).
mura, T., Miki, D., Mitsui, F., Hiraga, N., et al. (2009). Effects of structural vari-
Copy number variation: new insights in genome diversity. Genome Res. 16,
ations of APOBEC3A and APOBEC3B genes in chronic hepatitis B virus infec-
949–961.
tion. Hepatol. Res. 39, 1159–1168.
Gilbert, N., Lutz-Prigge, S., and Moran, J.V. (2002). Genomic deletions created
Alkan, C., Kidd, J.M., Marques-Bonet, T., Aksay, G., Antonacci, F., Hormoz-
upon LINE-1 retrotransposition. Cell 110, 315–325.
diari, F., Kitzman, J.O., Baker, C., Malig, M., Mutlu, O., et al. (2009). Personal-
ized copy number and segmental duplication maps using next-generation Gilbert, N., Lutz, S., Morrish, T.A., and Moran, J.V. (2005). Multiple fates of L1
sequencing. Nat. Genet. 41, 1061–1067. retrotransposition intermediates in cultured human cells. Mol. Cell. Biol. 25,
7780–7795.
An, P., Johnson, R., Phair, J., Kirk, G.D., Yu, X.F., Donfield, S., Buchbinder, S.,
Goedert, J.J., and Winkler, C.A. (2009). APOBEC3B deletion and risk of HIV-1 Goodier, J.L., Ostertag, E.M., and Kazazian, H.H., Jr. (2000). Transduction of
acquisition. J. Infect. Dis. 200, 1054–1058. 30 -flanking sequences is common in L1 retrotransposition. Hum. Mol. Genet.
Antonacci, F., Kidd, J.M., Marques-Bonet, T., Teague, B., Ventura, M., Girira- 9, 653–657.
jan, S., Alkan, C., Campbell, C.D., Vives, L., Malig, M., et al. (2010). A large and Han, K., Lee, J., Meyer, T.J., Wang, J., Sen, S.K., Srikanta, D., Liang, P., and
complex structural polymorphism at 16p12.1 underlies microdeletion disease Batzer, M.A. (2007). Alu recombination-mediated structural deletions in the
risk. Nat. Genet. 42, 745–750. chimpanzee genome. PLoS Genet. 3, 1939–1949.
Barrett, J.C., Hansoul, S., Nicolae, D.L., Cho, J.H., Duerr, R.H., Rioux, J.D., Hastings, P.J., Ira, G., and Lupski, J.R. (2009). A microhomology-mediated
Brant, S.R., Silverberg, M.S., Taylor, K.D., Barmada, M.M., et al; NIDDK IBD break-induced replication model for the origin of human copy number varia-
Genetics Consortium; Belgian-French IBD Consortium; Wellcome Trust tion. PLoS Genet. 5, e1000327.
Case Control Consortium. (2008). Genome-wide association defines more Huang, C.R., Schneider, A.M., Lu, Y., Niranjan, T., Shen, P., Robinson, M.A.,
than 30 distinct susceptibility loci for Crohn’s disease. Nat. Genet. 40, Steranka, J.P., Valle, D., Civin, C.I., Wang, T., et al. (2010). Mobile interspersed
955–962. repeats are major structural variants in the human genome. Cell 141, 1171–
Batzer, M.A., and Deininger, P.L. (2002). Alu repeats and human genomic 1182.
diversity. Nat. Rev. Genet. 3, 370–379. Istrail, S., Sutton, G.G., Florea, L., Halpern, A.L., Mobarry, C.M., Lippert, R.,
Bayés, M., Magano, L.F., Rivera, N., Flores, R., and Pérez Jurado, L.A. (2003). Walenz, B., Shatkay, H., Dew, I., Miller, J.R., et al. (2004). Whole-genome
Mutational mechanisms of Williams-Beuren syndrome deletions. Am. J. Hum. shotgun assembly and comparison of human genome assemblies. Proc.
Genet. 73, 131–151. Natl. Acad. Sci. USA 101, 1916–1921.
Beck, C.R., Collier, P., Macfarlane, C., Malig, M., Kidd, J.M., Eichler, E.E., Itsara, A., Cooper, G.M., Baker, C., Girirajan, S., Li, J., Absher, D., Krauss,
Badge, R.M., and Moran, J.V. (2010). LINE-1 retrotransposition activity in R.M., Myers, R.M., Ridker, P.M., Chasman, D.I., et al. (2009). Population anal-
human genomes. Cell 141, 1159–1170. ysis of large copy number variants and hotspots of human genetic disease.
Bekpen, C., Marques-Bonet, T., Alkan, C., Antonacci, F., Leogrande, M.B., Am. J. Hum. Genet. 84, 148–161.
Ventura, M., Kidd, J.M., Siswara, P., Howard, J.C., and Eichler, E.E. (2009). Jeffreys, A.J., Tamaki, K., MacLeod, A., Monckton, D.G., Neil, D.L., and
Death and resurrection of the human IRGM gene. PLoS Genet. 5, e1000403. Armour, J.A.L. (1994). Complex gene conversion events in germline mutation
Bennett, E.A., Keller, H., Mills, R.E., Schmidt, S., Moran, J.V., Weichenrieder, at human minisatellites. Nat. Genet. 6, 136–145.
O., and Devine, S.E. (2008). Active Alu retrotransposons in the human genome. Jiang, Z., Hubley, R., Smit, A., and Eichler, E.E. (2008). DupMasker: a tool for
Genome Res. 18, 1875–1883. annotating primate segmental duplications. Genome Res. 18, 1362–1368.
Benson, G. (1999). Tandem repeats finder: a program to analyze DNA Kidd, J.M., Newman, T.L., Tuzun, E., Kaul, R., and Eichler, E.E. (2007). Popu-
sequences. Nucleic Acids Res. 27, 573–580. lation stratification of a common APOBEC gene deletion polymorphism. PLoS
Buard, J., Shone, A.C., and Jeffreys, A.J. (2000). Meiotic recombination and Genet. 3, e63.
flanking marker exchange at the highly unstable human minisatellite CEB1 Kidd, J.M., Cooper, G.M., Donahue, W.F., Hayden, H.S., Sampas, N., Graves,
(D2S90). Am. J. Hum. Genet. 67, 333–344. T., Hansen, N., Teague, B., Alkan, C., Antonacci, F., et al. (2008). Mapping and
Chiaromonte, F., Yap, V.B., and Miller, W. (2002). Scoring pairwise genomic sequencing of structural variation from eight human genomes. Nature 453,
sequence alignments. Pacific Symposium on Biocomputing 7, 115–126. 56–64.

846 Cell 143, 837–847, November 24, 2010 ª2010 Elsevier Inc.
Kidd, J.M., Sampas, N., Antonacci, F., Graves, T., Fulton, R., Hayden, H.S., Al- Reiter, L.T., Murakami, T., Koeuth, T., Gibbs, R.A., and Lupski, J.R. (1997). The
kan, C., Malig, M., Ventura, M., Giannuzzi, G., et al. (2010). Characterization of human COX10 gene is disrupted during homologous recombination between
missing human genome sequences and copy-number polymorphic insertions. the 24 kb proximal and distal CMT1A-REPs. Hum. Mol. Genet. 6, 1595–1603.
Nat. Methods 7, 365–371. Rice, P., Longden, I., and Bleasby, A. (2000). EMBOSS: the European Molec-
Korbel, J.O., Urban, A.E., Affourtit, J.P., Godwin, B., Grubert, F., Simons, J.F., ular Biology Open Software Suite. Trends Genet. 16, 276–277.
Kim, P.M., Palejev, D., Carriero, N.J., Du, L., et al. (2007). Paired-end mapping
Richard, G.F., Kerrest, A., and Dujon, B. (2008). Comparative genomics and
reveals extensive structural variation in the human genome. Science 318,
molecular dynamics of DNA repeats in eukaryotes. Microbiol. Mol. Biol. Rev.
420–426.
72, 686–727.
Lagerstedt, K., Karsten, S.L., Carlberg, B.M., Kleijer, W.J., Tönnesen, T., Pet-
tersson, U., and Bondeson, M.L. (1997). Double-strand breaks may initiate the Roth, D.B., and Wilson, J.H. (1986). Nonhomologous recombination in
inversion mutation causing the Hunter syndrome. Hum. Mol. Genet. 6, mammalian cells: role for short sequence homologies in the joining reaction.
627–633. Mol. Cell. Biol. 6, 4295–4304.

Lam, H.Y., Mu, X.J., Stütz, A.M., Tanzer, A., Cayting, P.D., Snyder, M., Kim, Sen, S.K., Han, K., Wang, J., Lee, J., Wang, H., Callinan, P.A., Dyer, M.,
P.M., Korbel, J.O., and Gerstein, M.B. (2010). Nucleotide-resolution analysis Cordaux, R., Liang, P., and Batzer, M.A. (2006). Human genomic deletions
of structural variants using BreakSeq and a breakpoint library. Nat. Biotechnol. mediated by recombination between Alu elements. Am. J. Hum. Genet. 79,
28, 47–55. 41–53.
Lee, J.A., Carvalho, C.M., and Lupski, J.R. (2007). A DNA replication mecha- She, X., Jiang, Z., Clark, R.A., Liu, G., Cheng, Z., Tuzun, E., Church, D.M.,
nism for generating nonrecurrent rearrangements associated with genomic Sutton, G., Halpern, A.L., and Eichler, E.E. (2004). Shotgun sequence
disorders. Cell 131, 1235–1247. assembly and recent segmental duplications within the human genome.
McCarroll, S.A., Huett, A., Kuballa, P., Chilewski, S.D., Landry, A., Goyette, P., Nature 431, 927–930.
Zody, M.C., Hall, J.L., Brant, S.R., Cho, J.H., et al. (2008a). Deletion polymor- Smit, A., Hubley, R., and Green, P. (1996–2004). RepeatMasker Open-3.0.
phism upstream of IRGM associated with altered IRGM expression and http://www.repeatmasker.org/cgi-bin/WEBRepeatMasker.
Crohn’s disease. Nat. Genet. 40, 1107–1112.
Smith, C.E., Llorente, B., and Symington, L.S. (2007). Template switching
McCarroll, S.A., Kuruvilla, F.G., Korn, J.M., Cawley, S., Nemesh, J., Wysoker, during break-induced replication. Nature 447, 102–105.
A., Shapero, M.H., de Bakker, P.I., Maller, J.B., Kirby, A., et al. (2008b). Inte-
Symer, D.E., Connelly, C., Szak, S.T., Caputo, E.M., Cost, G.J., Parmigiani, G.,
grated detection and population-genetic analysis of SNPs and copy number
and Boeke, J.D. (2002). Human l1 retrotransposition is associated with genetic
variation. Nat. Genet. 40, 1166–1174.
instability in vivo. Cell 110, 327–338.
McVey, M., and Lee, S.E. (2008). MMEJ repair of double-strand breaks (direc-
tor’s cut): deleted sequences and alternative endings. Trends Genet. 24, Szak, S.T., Pickeral, O.K., Makalowski, W., Boguski, M.S., Landsman, D., and
529–538. Boeke, J.D. (2002). Molecular archeology of L1 insertions in the human
genome. Genome Biol. 3, research0052.
Mills, R.E., Luttig, C.T., Larkins, C.E., Beauchamp, A., Tsui, C., Pittard, W.S.,
and Devine, S.E. (2006). An initial map of insertion and deletion (INDEL) varia- Tristem, M. (2000). Identification and characterization of novel human endog-
tion in the human genome. Genome Res. 16, 1182–1190. enous retrovirus families by phylogenetic screening of the human genome
Moran, J.V., DeBerardinis, R.J., and Kazazian, H.H., Jr. (1999). Exon shuffling mapping project database. J. Virol. 74, 3715–3730.
by L1 retrotransposition. Science 283, 1530–1534. Tuzun, E., Sharp, A.J., Bailey, J.A., Kaul, R., Morrison, V.A., Pertz, L.M.,
Myers, S., Freeman, C., Auton, A., Donnelly, P., and McVean, G. (2008). Haugen, E., Hayden, H., Albertson, D., Pinkel, D., et al. (2005). Fine-scale
A common sequence motif associated with recombination hot spots and structural variation of the human genome. Nat. Genet. 37, 727–732.
genome instability in humans. Nat. Genet. 40, 1124–1129. Visser, R., Shimokawa, O., Harada, N., Kinoshita, A., Ohta, T., Niikawa, N., and
Parsons, J.D. (1995). Miropeats: graphical DNA sequence comparisons. Com- Matsumoto, N. (2005). Identification of a 3.0-kb major recombination hotspot
put. Appl. Biosci. 11, 615–619. in patients with Sotos syndrome who carry a common 1.9-Mb microdeletion.
Payen, C., Koszul, R., Dujon, B., and Fischer, G. (2008). Segmental duplica- Am. J. Hum. Genet. 76, 52–67.
tions arise from Pol32-dependent repair of broken forks through two alterna- Willer, C.J., Speliotes, E.K., Loos, R.J., Li, S., Lindgren, C.M., Heid, I.M.,
tive replication-based mechanisms. PLoS Genet. 4, e1000175. Berndt, S.I., Elliott, A.L., Jackson, A.U., Lamina, C., et al; Wellcome Trust
Pickeral, O.K., Maka1owski, W., Boguski, M.S., and Boeke, J.D. (2000). Case Control Consortium; Genetic Investigation of ANthropometric Traits
Frequent human genomic DNA transduction driven by LINE-1 retrotransposi- Consortium. (2009). Six new loci associated with body mass index highlight
tion. Genome Res. 10, 411–415. a neuronal influence on body weight regulation. Nat. Genet. 41, 25–34.
Redon, R., Ishikawa, S., Fitch, K.R., Feuk, L., Perry, G.H., Andrews, T.D., Fie- Witherspoon, D.J., Xing, J., Zhang, Y., Watkins, W.S., Batzer, M.A., and Jorde,
gler, H., Shapero, M.H., Carson, A.R., Chen, W., et al. (2006). Global variation L.B. (2010). Mobile element scanning (ME-Scan) by targeted high-throughput
in copy number in the human genome. Nature 444, 444–454. sequencing. BMC Genomics 11, 410.

Cell 143, 837–847, November 24, 2010 ª2010 Elsevier Inc. 847
Scientific Editor, Cell Press
Cell Press seeks to appoint three Scientific Editors with dual roles covering scientific
editing and the review material. These positions will be associated with the Cell Press
titles Cancer Cell, Current Biology, Developmental Cell, and Neuron, and expertise in
any of the relevant areas covered by these journals will be considered. Working closely
with the research community, you will be acquiring, managing, and developing new
editorial content for the Cell Press research titles. These positions will also work closely
with other aspects of the business, including production, business development,
marketing, and commercial sales, and, therefore, provide an excellent entry opportunity
to science publishing. You will work as part of a highly dynamic and collaborative
editorial group in the Cambridge, MA office. These positions are an exciting opportunity
to stay at the forefront of the latest scientific advances while developing a new career in
an exciting publishing environment.

Minimum qualifications are a PhD in a relevant life science discipline, and additional
postdoctoral or other experience is a plus. Ideal candidates would have a strong
scientific background and broad research interests, excellent writing and communica-
tion skills, strong organizational and interpersonal skills, as well as creative energy and
enthusiasm for science and science communication. Prior publishing or editorial
experience is an advantage but is not a requirement.

To apply
Please submit to the url below a CV and cover letter explaining your interest in an
editorial position and describing your qualifications, research interests, and reasons for
pursuing a career in scientific publishing. Applications will be accepted on an ongoing
basis through December 1, 2010.

http://reedelsevier.taleo.net/careersection/51/jobdetail.ftl?lang=en&job=SCI00063.

No phone inquiries. Elsevier-Cell Press is an Equal Opportunity Employer.


Scientific Editor, Molecular Cell
Molecular Cell is seeking a full-time scientific editor to join its editorial team. We will
consider qualified candidates with scientific expertise in any area that the journal
covers. The minimum qualification for this position is a PhD in a relevant area of
biomedical research, although additional experience is preferred. This is a superb
opportunity for a talented individual to play a critical role in the research community
away from the bench.

As a scientific editor, you would be responsible for assessing submitted research


papers, overseeing the refereeing process, and choosing and commissioning review
material. You would also travel frequently to scientific conferences to follow develop-
ments in research and establish and maintain close ties with the scientific community.
The key qualities we look for are breadth of scientific interest and the ability to think
critically about a wide range of scientific issues. The successful candidate will also be
highly motivated and creative and able to work independently as well as in a team.

This is a full-time in-house editorial position, based at the Cell Press office in Cambridge,
Massachusetts. Cell Press offers an attractive salary and benefits package and a
stimulating working environment. Applications will be held in the strictest of confidence
and will be considered on an ongoing basis until the position is filled.

To apply
Please submit a CV and cover letter describing your qualifications, research interests,
and reasons for pursuing a career in scientific publishing, as soon as possible, to our
online jobs site:
http://www.elsevier.com/wps/find/job_search.careers. Click on “search for US jobs”
and select “Massachusetts.”
Or:
http://reedelsevier.taleo.net/careersection/51/jobdetail.ftl?lang=en&job=SCI0005X.

No phone inquiries, please. Cell Press is an equal opportunity/affirmative action


employer, M/F/D/V.
Scientific Editor, Cell Metabolism
Cell Metabolism is seeking a full-time scientific editor to join its editorial team. Cell
Metabolism publishes metabolic research with an emphasis on molecular mechanisms
and translational medicine. The minimum qualification for this position is a PhD in a
relevant area of biomedical research, although additional postdoctoral and/or editorial
experience is preferred. This is a superb opportunity for a talented individual to play a
critical role in promoting science by helping researchers shape and disseminate their
findings to the wider community.

The scientific editor is responsible for assessing submitted research papers, overseeing
the refereeing process, and choosing, commissioning, and editing review material. The
scientific editor frequently travels to scientific conferences to follow developments in
research and establish and maintain close ties with the scientific community. The key
qualities we look for are breadth of scientific interest, the ability to think critically about
a wide range of scientific issues, and strong communication skills. The successful
candidate will also be highly motivated and creative and able to work independently
as well as in a team and should have opportunities to pioneer and contribute to new
trends in scientific publishing.

This is a full-time in-house editorial position, based at the Cell Press office in Cambridge,
Massachusetts. Cell Press offers an attractive salary and benefits package and a
stimulating working environment that encourages innovation.

Please submit a CV and cover letter describing your qualifications, general research
interests, and motivation for pursuing a career in scientific publishing. Applications will
be considered on an ongoing basis until the closing date of November 15th, 2010.

To apply, visit
http://reedelsevier.taleo.net/careersection/51/jobdetail.ftl?lang=en&job=SCI0005Y.

No phone inquiries. Elsevier-Cell Press is an Equal Opportunity Employer.


Scientific Editor, Neuron
Neuron is seeking an additional full-time scientific editor to join its editorial team based
in Cambridge, Massachusetts. Neuron publishes across a range of disciplines includ-
ing developmental, molecular, cellular, systems, and cognitive neuroscience.

As a scientific editor, you would be responsible for assessing submitted research


manuscripts, overseeing the review process, and commissioning and editing review
material for the journal. You would also travel frequently to scientific conferences to
follow developments in research and to establish and maintain close ties with the
scientific community.

The minimum qualification for this position is a PhD in a relevant area of biomedical
research, although previous editorial experience is beneficial. This is a superb opportu-
nity for a talented individual to play a critical role in the research community away from
the bench. The key qualities we are looking for are breadth of scientific interest and the
ability to think critically about a wide range of scientific issues. The successful candi-
date will also be highly motivated and creative, possess strong communication skills,
and be able to both work independently and as part of a team.

This is a full-time, in-house editorial position, based at Cell Press headquarters in


Cambridge, Massachusetts. Cell Press offers an attractive salary and benefits package
and a stimulating work environment. Applications will be held in the strictest of confi-
dence and will be considered on an ongoing basis.

To apply
Please submit a cover letter describing your background, interests, and a candid
appraisal of the strengths and weaknesses of Neuron, along with your CV, to
http://reedelsevier.taleo.net/careersection/51/jobdetail.ftl?lang=en&job=SCI0006F.
Applications will be accepted through December 1st, 2010.
The American Journal of Human Genetics
Editor Position Available

The American Society of Human Genetics is seeking an Editor for The American Journal of Human
Genetics. The Editor leads one of the world’s oldest and most prestigious journals publishing pri-
mary human genetics research.

Among the Editor’s responsibilities are determining the scope and direction of the scientific con-
tent of The Journal, overseeing manuscripts submitted for review and their publication, selecting
and supervising a staff consisting of an Editorial Assistant and doctoral-level Deputy Editor, direct-
ing interactions with the publisher (currently Cell Press), reviewing quarterly reports provided by the
publisher, evaluating the performance of the publisher, and if required, supervising the process of
the selection a new publisher. The Editor serves as a member of the Board of Directors of the Ameri-
can Society of Human Genetics (ASHG), as well as the ASHG Finance Committee, and presents
semiannual reports to the Board. All Associate Editors of The Journal are appointed by the Editor,
who also determines their duties. At the ASHG annual meeting, the Editor presides over a meeting
of the Associate Editors and presents an annual report to the ASHG membership.

The term of the appointment is five years and includes a yearly stipend. The new Editor will be
selected by the end of 2010 and will begin receiving manuscripts approximately in September 2011;
there will be partial overlap with the Boston office. Applicants should be accomplished scientists
in the field of human genetics and should have a broad knowledge and appreciation of the field.
Nominations, as well as applications consisting of a letter of interest and curriculum vitae, should be
sent to:

AJHG Editorial Search Committee


American Society of Human Genetics
9650 Rockville Pike
Bethesda, MD 20814
Editor: Trends in Molecular Medicine
We are seeking to appoint a new Editor for Trends in Molecular Medicine, to be based
in the Cell Press offices in Cambridge, Massachusetts.

As Editor of Trends in Molecular Medicine, you will be responsible for the strategic
development and content management of the journal. You will be acquiring and devel-
oping the very best editorial content, making use of a network of contacts in academia
plus information gathered at international conferences, to ensure that Trends in
Molecular Medicine maintains its market-leading position.

This is an exciting and challenging role that provides an opportunity to stay close to the
cutting edge of scientific advances while developing a new career away from the
bench. You will work in a highly dynamic and collaborative publishing environment that
includes 14 Trends titles and 12 Cell Press titles. You will also collaborate with your Cell
Press colleagues to maximize quality and efficiency of content commissioning and
participate in exciting new non-journal-based initiatives.

The minimum qualification is a doctoral degree in a relevant discipline, and post-


doctoral training is an advantage. Previous publishing experience is not necessary—we
will make sure you get the training and development you need. Good interpersonal
skills are essential because the role involves networking in the wider scientific commu-
nity and collaboration with other parts of the business.

To apply
Please submit a CV and cover letter describing your qualifications, research interests,
current salary, and reasons for pursuing a career in publishing at
http://reedelsevier.taleo.net/careersection/51/jobdetail.ftl?lang=en&job=SCI0006D. No
phone inquiries, please. Cell Press is an equal opportunity employer.

Applications will be considered on an ongoing basis until the closing date of November
26th, 2010.
Cell Press Business Project Editor
Position Available

Cell Press is seeking a Business Project Editor to plan, develop, and implement projects that have
commercial or sponsorship potential. By drawing on existing content or developing new material, the
Editor will work with Cell Press’s commercial sales group to create collections of content in print or
online that will be attractive to readers and sponsors. The Editor will also be responsible for leverag-
ing new online opportunities for engaging the readers of Cell Press journals.

The successful candidate will have a PhD in the biological sciences, broad scientific interests, a
fascination with technology, good commercial instincts, and a true passion for both science and
science communication. They should be highly organized and dedicated, with excellent written and
oral communication skills, and should be willing to work to tight deadlines.

The position is full time and based in Cambridge, MA. Cell Press offers an attractive salary and
benefits package and a stimulating work environment. Applications will be considered on a rolling
basis. For consideration, please apply online and include a cover letter and resume. To apply, visit
the career page at http://www.elsevier.com and search on keywords “Business Project Editor.”
EDITOR-IN-CHIEF SENIOR EDITORS ASSOCIATE EDITORS
F.E. Bloom J.F. Baker G. Aston-Jones T.A. Milner
La Jolla, CA, USA Chicago, IL, USA Charleston, SC, USA New York, NY, USA
P.R. Hof J.S. Baizer S.D. Moore
New York, NY, USA Buffalo, NY, USA Durham, NC, USA
G.R. Mangun J.D. Cohen T.H. Moran
Davis, CA, USA Princeton, NJ, USA Baltimore, MD, USA
J.I. Morgan B.M. Davis T.F. Münte
Memphis, TN, USA Pittsburgh, PA, USA Magdeburg, Germany
F.R. Sharp J. De Felipe K-C. Sonntag
Sacramento, CA, USA Madrid, Spain Belmont, MA, USA
R.J.Smeyne M.A. Dyer R.J. Valentino
Memphis, TN, USA Memphis, TN, USA Philadelphia, PA, USA
A.F. Sved M.S. Gold C.L. Williams
Pittsburgh, PA, USA Pittsburgh, PA, USA Durham,NC, USA
G.F. Koob
La Jolla, CA, USA

1
23 Twenty-three to
the Power of One.
One re-unified journal, nine specialist sections, 23 receiving Editors ←
Authors receive first editorial decision within 30 days of submission ←
“Young Investigator Awards” for innovative work by a new generation of researchers ←

Brain Research take another look


www.elsevier.com/locate/brainres
Announcements/Positions Available

Columbia University’s CCTI is seeking a qualified


Associate Research Scientist who will have significant
research responsibilities which include directing the
large animal operations/facility. Incumbent will be
responsible for the research infrastructure of the CCTI.
Will monitor and develop standard operating procedures
for the research operation. The candidate is required to
have a MD. or Ph.D. in biology with significant
research experience in transplantation immunology.
Salary offered is $84,000 but will commensurate with
experience. Interested applicants should send a CV,
letter of interest and names of three references to:

Mayra Marte-Miraz
Columbia University Medical Center
630W. 168th Street
New York, NY 10032

or via email at mm18@columbia.edu.

CUMC is an EOE.
Positions Available

Bowes Research Fellows


University of California Berkeley

The Bowes Research Fellows Program at the University of California, Berkeley, is seeking nominations
of outstanding recent or imminent Ph.D. and M.D. graduates to be given the freedom to establish an
independent research program as an alternative to the traditional postdoctoral experience. Bowes
Fellows must have demonstrated exceptional promise and maturity in their graduate careers and be
eager to engage the frontiers of biomedical and life sciences. Fellows will receive funding and space
sufficient to maintain a laboratory of two to three members for a term of up to five years, free from the
need to obtain grant support or the distractions of classroom teaching. Fellows will have principal
investigator status, making them eligible to obtain outside funding from grants or other sources as their
research programs expand.

Bowes Fellows benefit from the mentorship of our faculty, as well as from the exceptional breadth of
our scientific resources and the highly interactive nature of the Berkeley community. In turn, our
community benefits from the creative approaches Bowes Fellows take to solving important problems.
Because interdisciplinary interactions are key to innovation, we seek to attract individuals who have
broad interests in the life sciences and who have diverse expertise in experimental, theoretical and/or
computational approaches.

Candidates must be nominated by their current mentor or by another senior investigator who can
provide an in-depth analysis of their accomplishments and future potential. Refer potential reviewers
to the UC Berkeley Statement of Confidentiality found at: http://apo.chance.berkeley.edu/evalltr.html.

Selected candidates will be asked to submit a brief research plan and to arrange for additional letters
of recommendation. Finalists will be invited to interview on the UC Berkeley campus. Nominations
must be received by December 15, 2010 and should be sent to (email submissions are preferred):

Michael Eisen
Chair, Bowes Research Fellows Selection Committee
Department of Molecular & Cell Biology
University of California, Berkeley
Stanley Hall 304B
Berkeley CA 94720-3220
mbeisen@berkeley.edu

The University of California is an affirmative action, equal opportunity employer.


4HEå"IOPHYSICALå3OCIETYå
ANDå#ELLå0RESS
!åNEWåPARTNERSHIP

3EEåWHATSåNEWåAT
WWWBIOPHYSJORG
EuPA now has
http://www.eupa.org/
its own journal!

Editor in Chief:
Juan J. Calvete, Valencia, Spain Covered by
PubMed
Executive Editors:
Proteomics in Cell Biology
Jean-Jacques Diaz, Lyon, France

Proteomics in Microbiology
Concha Gil, Madrid, Spain

Proteomics in Plant Systems


Jesus V. Jorrín, Córdoba, Spain

Proteomics in Animal Models


Dario Neri, Zürich, Switzerland

Proteomics in Protein Science


Jasna Peter-Katalinic, Münster,
Germany

Biomedical Applications of
Proteomics and Congress
Proceedings
Jean-Charles Sanchez, Geneva,
Switzerland

Proteomics of Body Fluids and


Proteomic Technologies
Pier Giorgio Righetti, Milan, Italy Submitting Authors
Manuscripts can be submitted to the
Bioinformatics in Proteomics Journal of Proteomics at
Peter Højrup, Odense, Denmark http://ees.elsevier.com/jprot
For a complete listing of the
editorial board, visit the
journal’s homepage

To receive more information register at:


http://www.elsevier.com/locate/jprot
SnapShot: The SUMO System
Sandrine Creton and Stefan Jentsch
Max Planck Institute of Biochemistry, Martinsried 82152, Germany

848 Cell 143, November 24, 2010 ©2010 Elsevier Inc. DOI 10.1016/j.cell.2010.11.026 See online version for legend and references.
.%7

Reach Your Ideal Candidate!


så0OSTåONEåORåMULTIPLEåå
JOBåOPENINGSå

så0ROMOTEåANåUPCOMINGåå
CONFERENCEåORåEVENTå

så!NNOUNCEåAåGRANTAWARDå

så%LEVATEåYOURåå
ORGANIZATIONSåPROlLE

careers.cell.com
Nanocrystal Vision
Conjugate your protein in the morning …
See results in the afternoon.
Add the power of nanocrystals
to your research. New eFluor®
Nanocrystal Conjugation Kits allow you to
create your own nanocrystal-conjugated
proteins more quickly and simply than
ever before.

eFluor® Nanocrystal Conjugation Kits offer:


• Intrinsic Brightness – photostable
fluorophore-conjugated proteins
• Streamlined Protocols – under 30 min hands-
on time
• Maximum Versatility – label both polyclonal
and monoclonal antibodies
• Flexibility – use eFluor® reagents for ICC, IF,
IHC and Flow Cytometry

MCF-7 cells stained with eFluor® Nanocrystals. Top: Cytokeratin-605NC


(pseudocolor green) and Tubulin-650NC (red) conjugated using eFluor®
NC Conjugation Kit – Sulfhydryl Reactive. Ki67 staining also shown (blue).
Bottom: Cytokeratin-605NC (pseudocolor green) and Tubulin-650NC
(blue) conjugated using eFluor® NC Conjugation Kit – Amine Reactive. Ki67
staining also shown (red).

Make your data nanocrystal clear today and


see more results. Visit www.eBioscience.com.

full spectrum cell analysis


www.eBioscience.com ORDER 888.999.1371 • TECH 888.810.6168 • INQUIRIES info@eBioscience.com

You might also like