You are on page 1of 13

REVIEWS

C y t o s k e l e ta l M o t o r s

Non-muscle myosin II takes centre


stage in cell adhesion and migration
Miguel Vicente‑Manzanares*, Xuefei Ma‡, Robert S. Adelstein‡§ and
Alan Rick Horwitz*§
Abstract | Non-muscle myosin II (NM II) is an actin-binding protein that has actin cross-linking
and contractile properties and is regulated by the phosphorylation of its light and heavy
chains. The three mammalian NM II isoforms have both overlapping and unique properties.
Owing to its position downstream of convergent signalling pathways, NM II is central in the
control of cell adhesion, cell migration and tissue architecture. Recent insight into the role
of NM II in these processes has been gained from loss-of-function and mutant approaches,
methods that quantitatively measure actin and adhesion dynamics and the discovery of
NM II mutations that cause monogenic diseases.

Actin filament
Myosins constitute a superfamily of motor proteins 17 kDa essential light chains (ELCs) that stabilize the
A strand of polymerized that play important parts in several cellular processes that heavy chain structure (FIG. 1a). Although these myosins
globular actin subunits that require force and translocation1–3. Recent analyses of are referred to as ‘non-muscle’ myosin IIs to disting-
winds around another strand genomic databases have yielded an increasing number uish them from their muscle counterparts, they are
to form a helix. Actin filaments
of myosin classes in eukaryotic cells4,5. Myosin molecules also present in muscle cells, where they have distinct
are one of the three major
cytoskeletal elements of a cell,
can walk along, propel the sliding of or produce tension functions during skeletal muscle development and
along with microtubules and on actin filaments. This requires energy, which is provided differentiation12, as well as in the maintenance of tension
intermediate filaments. by the hydrolysis of ATP, and requires myosins to have in smooth muscle13,14.
catalytic sites with ATPase activity. Myosin catalytic sites NM II has a fundamental role in processes that
Integrin
One of a large family of
are usually found in the amino-terminal (head) region of require cellular reshaping and movement, such as cell
heterodimeric transmembrane the molecule, and they are often activated when myosin adhesion, cell migration and cell division. NM II can use
proteins that functions as a binds to actin. The carboxy-terminal region of some its actin cross-linking and contractile functions, which
receptor for ECM or cell myosins binds to and moves cargo in a cell, whereas the are regulated by phosphorylation and the ability of NM II
adhesion molecules.
C-terminal domains of other myosins self-associate into to form filaments, to regulate the actin cytoskeleton.
filaments, which allows their heads to tether actin fila- In this Review, we provide an overview of the
ments and exert tension. Myosins can also act indirectly structure and regulation of NM II, with emphasis on
through actin to bring adhesion-related proteins, such its central role in cell adhesion and cell migration, and
as integrins, or signal transduction molecules into close also outline the relationship between NM II mutations
proximity 2,3. and disease.
*Department of Cell Biology, Most myosins belong to class II and, together with
University of Virginia School
of Medicine, Charlottesville,
actin, make up the major contractile proteins of car- Domain structure of NM II
Virginia 22908, USA. diac, skeletal and smooth muscle, in which the sliding The two globular head domains of NM II contain a

Laboratory of Molecular crossbridges that connect thick myosin filaments with binding site for both ATP and actin and they are fol-
Cardiology, National Heart, thin actin filaments provide the force to, for example, lowed by neck regions, each of which binds the two
Lung, and Blood Institute,
pump blood, lift objects and expel babies6,7. Importantly, functionally different light chains. The neck domain
National Institutes of Health,
Bethesda, Maryland myosin II molecules that resemble their muscle counter- acts as a lever arm to amplify head rotation while the
20892‑1583, USA. parts, with respect to both structure and function, are chemical energy of ATP is converted into the mechani-
§
These authors contributed also present in all non-muscle eukaryotic cells8–11. Like cal movement of the myosin head1. This neck domain
equally to this work. muscle myosin II, non-muscle myosin II (NM II) mol- is followed by a long α-helical coiled coil, which forms
Correspondence to M.V.‑M.
e‑mail:
ecules are comprised of three pairs of peptides: two an extended rod-shaped domain that effects dimeriza-
miguelvix@virginia.edu heavy chains of 230 kDa, two 20 kDa regulatory light tion between the heavy chains and terminates in a rela-
doi:10.1038/nrm2786 chains (RLCs) that regulate NM II activity and two tively short non-helical tail (FIG. 1a). The rod domains

778 | NovEMbER 2009 | voLuME 10 www.nature.com/reviews/molcellbio

© 2009 Macmillan Publishers Limited. All rights reserved


REVIEWS

a 10S assembly-incompetent NM II 6S assembly-competent NM II


Globular head domain
(actin binding and
Mg2+-ATPase motor domains)
ELC
RLC Heavy chain
RLC phosphorylation

Coiled-coil rod domain


Non-helical tail

S-1
HMM MHC phosphorylation

b Bipolar NM II filaments

Actin filament

Figure 1 | Domain structure of NM II. a | The subunit and domain structure of non-muscle myosin II (NM II), which forms
a dimer through interactions between the α-helical coiled-coil rod domains. The globular
Nature head domain
Reviews contains
| Molecular Cellthe
Biology
actin-binding regions and the enzymatic Mg2+-ATPase motor domains. The essential light chains (ELCs) and the
regulatory light chains (RLCs) bind to the heavy chains at the lever arms that link the head and rod domains. In the absence
of RLC phosphorylation, NM II forms a compact molecule through a head to tail interaction. This results in an assembly-
incompetent form (10S; left) that is unable to associate with other NM II dimers. On RLC phosphorylation, the 10S
structure unfolds and becomes an assembly-competent form (6S). S-1 is a fragment of NM II that contains the motor
domain and neck but lacks the rod domain and is unable to dimerize. Heavy meromyosin (HMM) is a fragment that
contains the motor domain, neck and enough of the rod to effect dimerization. b | NM II molecules assemble into bipolar
filaments through interactions between their rod domains. These filaments bind to actin through their head domains and
the ATPase activity of the head enables a conformational change that moves actin filaments in an anti-parallel manner.
Bipolar myosin filaments link actin filaments together in thick bundles that form cellular structures such as stress fibres.

of NM II self-associate to form bipolar filaments (anti- splicing, and there is currently no known specificity
parallel arrays of myosin molecules), which are consider- of light chains for particular NMHC IIs. The two light
ably smaller than those found in cardiac and skeletal chain pairs are very tightly, but non-covalently, bound
muscle15,16 (FIG. 1b). to α-helical stretches of each heavy chain. Despite hav-
Three different genes in mammalian cells (myosin ing a high degree of homology, particularly in their
heavy chain 9 (MYH9), MYH10 and MYH14) encode actin-binding globular heads, the myosin isoforms are
the NM II heavy chain (NMHC II) proteins (NMHC IIA, spatially segregated in some areas of cells, but clearly
NMHC IIb and NMHC IIC, respectively), although overlap in others20,21. As detailed below, some cellular
there is only one NMHC II gene, zipper, in Drosophila functions are isoform-specific, whereas others are
melanogaster 17. We refer to the whole myosin II molecule redundant 22,23.
(heavy chains and light chains) as NM II and the heavy The short, non-helical tails of NMHC II proteins
chains alone as NMHCs. The NMHC isoform deter- differ sufficiently among the three mammalian iso-
mines the NM II isoform, which are named NM IIA, forms that peptides can be synthesized to mimic these
NM IIb or NM IIC, accordingly. For NM II motors to sequences for the generation of isoform-specific anti-
retain their normal, native and active conformations, bodies24. There is no evidence for heterodimer form-
the light chains must be bound to the heavy chains. ation between the three NMHC II isoforms, and when
Deletion of a specific NMHC II results in the loss of that immunoprecipitation experiments are performed on a
NM II isoform. mixture of isoforms with specific antibodies only the
The mammalian heavy chain pre-mRNAs that are antibody-specific isoform is detected 24. Two impor-
transcribed in humans and mice from MYH10 and tant kinetic properties that differ among the NM II
MYH14 undergo alternative splicing, predominantly isoforms are the actin-activated Mg 2+-ATPase activity
in neuronal tissue, which increases the total number (the increase in ATP hydrolysis by myosin when bound
Coiled coil of expressed NMHC II proteins to nine18. Although to actin) and the duty ratio (the time that myosin is
A structural domain that can there is evidence for alternative splicing of the MYH9 bound to actin in a force-generating state). NM IIA has
mediate oligomerization. The transcripts at locations homologous to those in MYH10 the highest rate of ATP hydrolysis of the three NM II
myosin coiled-coil rod domain
contains two α-helices that
and MYH14, it is not known whether these transcripts isoforms and it propels actin filaments more rapidly
twist around each other to are translated19. The light chains are encoded by a dif- than NM IIb and NM IIC25. NM IIb has a significantly
form a supercoil. ferent set of genes, which can also undergo alternative higher duty ratio than NM IIA26,27. NM IIb also has an

NATuRE REvIEWs | Molecular cell BIology voLuME 10 | NovEMbER 2009 | 779

© 2009 Macmillan Publishers Limited. All rights reserved


REVIEWS

extremely high affinity for ADP when compared to There is compelling evidence from in vitro experi-
other myosins, and the release of ADP from NM IIb ments that phosphorylation of the RLC also regulates
is markedly slowed by backward strain on the myosin the assembly of NM II filaments40. In the presence of
head (the force exerted on myosin heads by the actin ATP, unphosphorylated NM II folds into a compact
filaments, opposite to the direction of myosin head conformation, in which one head blocks the second
movement)28. Thus, NM IIb is particularly well suited head of the same molecule through an asymmetrical
to exert tension on actin filaments for longer periods of interaction (FIG. 1a; assembly-incompetent form)35,41–44.
time and with less expenditure of energy than NM IIA. The tail also folds at two points and interacts with the
Indeed, NM IIb has recently been shown to play an heads to further compact the molecule and prevent
ancillary role in smooth muscle contraction, in which filament assembly. Phosphorylation of RLCs disrupts
it may be responsible for a portion of the tension main- the head–head and head–tail interactions and causes the
tenance during tonic contractions13. These differences in compact, faster sedimenting (10s) molecule to form
kinetic properties between NM IIA and NM IIb might an elongated slower sedimenting (6s) species (FIG. 1a),
help to explain why expressing NMHC IIA under which forms bipolar filaments at physiological ionic
the control of the endogenous MYH10 promoter can strengths (FIG. 1b). Although these dramatic changes can
rescue some, but not all, of the abnormalities found in be demonstrated in solution, there is no firm evidence
NM IIb-ablated mice29. that they occur in vivo. The formation of assemblies
Whereas the different enzymatic and motor activities of comprising 14–20 myosin molecules in bipolar fila-
NM IIs reside in their N-terminal domain, the C-terminal ments promotes the interaction of myosin filaments
rod and non-helical tail determine the assembly with actin filaments (FIG. 1b).
of myosin filaments and the intracellular localization of More than a dozen kinases have been reported to
the NM II isoforms. Two assembly-competent domains phosphorylate the RLCs of NM II (FIG. 2), including
in the NM IIb rod that have a crucial role in filament myosin light chain kinase (MLCK; also known as MYLK),
formation30, and two regions in the NM IIb rod that are Rho-associated, coiled coil-containing kinase (RoCK),
involved in isoform-specific filament assembly, have been citron kinase, leucine zipper interacting kinase (ZIPK;
identified31. also known as DAPK3) and myotonic dystrophy kinase-
Experiments using chimaeras of NM IIA and NM IIb related CDC42-binding kinase (MRCK; also known as
have shown that the intracellular localization of these iso- CDC42bP)6,34,45,46. These kinases phosphorylate RLCs on
forms is determined by the C-terminal 179–190 amino ser19, Thr18 or both, to relieve the inhibition imposed
acids, which contain one of the assembly-competent on the myosin molecule by unphosphorylated RLCs
domains and the non-helical tail32,33. and the head–head interaction outlined above. However,
the activating signals for these kinases differ. Ca2+–
regulation of NM II activity calmodulin activates MLCK, which seems to be specific
In contrast to skeletal and cardiac myosins, which are for the NM II RLCs, whereas the small GTP-binding pro-
regulated by a separate set of proteins that are bound tein RHoA activates RoCK and citron kinase. unlike
to the actin filaments, the regulation of Mg 2+-ATP MLCK, RoCK and citron kinase phosphorylate several
hydrolysis and filament formation of NM II involves substrates in addition to the RLCs of NM II. For exam-
the reversible phosphorylation of specific amino acids ple, although RoCK can phosphorylate RLCs directly, it
present in the pair of 20 kDa RLCs and the heavy chains. primarily acts to inhibit the major myosin phosphatase,
The function of the ELC pair is to stabilize the NMHC protein phosphatase 1 (PP1), which is responsible for
and there is no evidence that they undergo reversible the dephosphorylation of NM II47. Phosphorylation of
phosphorylation. the regulatory subunit myosin phosphatase-targeting
subunit 1 (MYPT1; also known as PPP1R12A) of this
Regulation of NM II by RLC phosphorylation. The regu- trimeric phosphatase inactivates the enzymatic activity,
lation of NM IIA, NM IIb, NM IIC and smooth muscle resulting in increased NM II phosphorylation and activa-
myosin, but not cardiac or mammalian skeletal muscle tion. MLCK, at least in some cells, is more peripherally
myosin, depends on the reversible phosphorylation of localized, wherase RoCK is more central48,49. This dif-
the RLC on ser19. This phosphorylation event greatly ferential localization means that the actomyosin struc-
increases the Mg 2+-ATPase activity of myosin in the tures in the cell centre (such as stress fibres and mature
presence of actin34 by controlling the conformation of focal adhesions) are much more stable than those at the
the myosin heads35. However, phosphorylation has little periphery, which are more responsive to upstream stimuli.
Tonic contractions or no effect on the affinity of myosin for actin36. Thr18 Protein kinase C (PKC) differs from the above kinases
Sustained muscular can also be phosphorylated, and diphosphorylation of the in that it phosphorylates RLCs on ser1, ser2 and Thr9,
contractions that develop
RLC on ser19 and Thr18 often occurs in cultured cells37. which renders the RLC a poorer substrate for MLCK
slowly and show a prolonged
phase of relaxation. The additional phosphorylation of Thr18 in the pres- and therefore decreases the activity of NM II50 (FIG. 2).
ence of phosphorylated ser19 (REF. 38) further increases For example, PKC phosphorylation of the RLC on ser1
Actomyosin filaments the total actin-activated Mg 2+-ATPase activity at sub- or ser2 initiated by stimulation of platelet-derived growth
Produced when bipolar myosin saturating actin concentrations, but does not affect the factor receptor inhibits NM II activity to promote the
filaments interact with
polymerized actin filaments to
myosin vmax (the maximal actin-activated Mg 2+-ATPase reorganization of actomyosin filaments51. As noted below,
exert tension or produce activity of myosin at saturating actin concentrations) or in addition to phosphorylating the RLCs, PKC also
movement. the rate of movement of actin filaments in vitro39. phosphorylates the heavy chains.

780 | NovEMbER 2009 | voLuME 10 www.nature.com/reviews/molcellbio

© 2009 Macmillan Publishers Limited. All rights reserved


REVIEWS

Regulatory light chain non-helical NMHC tail is a substrate for phosphoryla-


tion by CK II, and phosphorylation of this residue, or of
Kinases PKC MLCK MRCK
ROCK ZIPK ser1917 by PKC, inhibits the assembly of NM IIA rods
Citron kinase into filaments54.
12 9 18 19 Phosphorylation of NM IIA also affects the binding
0 66.5$.$.77..5345$7619)$0 172 of s100A4 (also known as MTs1) to the NMHC. This
protein, which is a member of the s100 family of Ca2+-
binding proteins, promotes the invasion of metastatic
S100A4
tumour cells. binding of s100A4 to NMHC IIA prevents
filament formation, but s100A4–NMHC IIA binding is
blocked by the phosphorylation of ser1943 by CK II58.
An alpha kinase from the same family as D. discoideum
NMHC kinase A, TRPM7, and its close homologue
TRPM6, phosphorylate NM IIb and NM IIC at several
NMHC IIA sites in the non-helical tails59. Although the effects of
1,803
1,800 1,808 1,916 1,927 1,943 these phosphorylation events on filament assembly have
7 6 6 6 3 6 1,960 not yet been studied, the more specific phosphorylations
1,937 1,939
NMHC IIB 1,935 1,941 catalysed by TRPM7 in the helical portion of the rod
1,810 1,815 1,922 1,933 1,952 1,956 1,960 1,965 1,975 in NM IIA at Thr1800, ser1803 and ser1808 seem to
6 7 6 3 6 6666 6 6 7 6 6 1,976
6 66 6 decrease filament formation and alter the subcellular
NMHC IIC localization of NM IIA55. As yet, there is no evidence for
1,840 1,846 1,936 1,957 1,961 1,964 1,977 1,988 the endogenous phosphorylation of these sites.
7 6 7 3 7 7 6 6 2,003
7 7 Phosphorylation of NM IIb by aPKCζ, which is
Kinases TRPM7 PKCβ CK II regulated by p21-activated kinase 1 (PAK1), was stud-
PKC ied in a prostate cancer cell line. This phosphorylation,
PKCζ which occurs on ser1937 in the non-helical tail region
of NMHC IIb, is stimulated by epidermal growth factor
Figure 2 | regulation of NM II activation and filament formation. Multiple kinases,
(EGF) and leads to slower filament assembly in cultured
including myosin light chain kinase (MLCK; also known as MYLK),
Nature ReviewsRho-associated, coiled
| Molecular Cell Biology
coil-containing kinase (ROCK), citron kinase, myotonic dystrophy kinase-related cells53. Recently, phosphorylation of both the non-
CDC42-binding kinase (MRCK; also known as CDC42BP) and leucine zipper interacting helical tail and the rod of the NMHC has been reported
kinase (ZIPK; also known as DAPK3) can phosphorylate the regulatory light chain (RLC) for NM IIC. Phosphorylation increases the solubility of
of non-muscle myosin II (NM II) on Ser19 or on Thr18 and Ser19 to activate it. Protein NM IIC and also has an effect on cellular localization60.
kinase C (PKC) can phosphorylate the RLC on Ser1, Ser2 and Thr9 to inhibit NM II. Human Thus, it is clear that considerable regulation of NM II
RLC is encoded by myosin light chain 9 (MYL9). In addition to RLC phosphorylation, occurs through phosphorylation of the heavy chains,
NM II filament assembly is regulated by phosphorylation of the NM II heavy chain particularly in the tail region (FIG. 2). The continued
(NMHC II) coiled-coil and tail domains. The phosphorylation sites and the corresponding characterization of the roles of these different phospho-
kinases, including transient receptor potential melastatin 7 (TRPM7), PKC proteins
rylation events on NM II function and isoform-specific
and casein kinase II (CK II), are shown for human NMHC IIA, NMHC IIB and NMHC IIC.
regulation should produce important new insights.
Phosphorylation of NMHC II by CK II blocks the binding of S100A4 (also known as MTS1)
to the tail of NM II to prevent it from inhibiting myosin filament assembly. Human
NMHC IIA is encoded by myosin heavy chain 9 (MYH9), NMHC IIB is encoded by MYH10 NM II in cell migration, adhesion and polarity
and NMHC IIC is encoded by MYH14. Note that each phosphorylation site and the kinase NM II is an important regulator of adhesion and
that phosphorylates it are depicted in the same colour. polarity in cell migration. These processes involve the
dynamic remodelling of the actin cytoskeleton and
the interaction of the cell with its environment. Each of the
Regulation of NM II by NMHC phosphorylation. NM II isoforms affects these processes differently, as
Although phosphorylation of NMHC II plays an impor- discussed below.
tant part in regulating the activity of Dictyostelium dis-
coideum myosin II (reviewed in REF. 52), the roles in NM II regulates protrusion and cell migration. In migrat-
regulating the structure and activity of mammalian NM II ing cells, actin organizes into several distinct structures
Lamellipodium
A 1–2 μm-wide band that is
are still being defined. There are several phosphorylation and its polymerization in cellular protrusions drives
made up of a network of sites near the C-termini of NMHCs, in both the coiled- cell migration. Protrusions generally contain two actin-
dendritic actin filaments and coil domain and the non-helical tail, including sites that based structures: the lamellipodium and the lamellum61.
forms the outer edge of a cell are phosphorylated by PKC53, casein kinase II (CK II)54 Different classes of regulatory molecules organize actin
protrusion.
and transient receptor potential melastatin 7 (TRPM7)55 in these two structures. The actin nucleator Arp2/3
Lamellum (FIG. 2; see below). In each case phosphorylation of the generates the lamellipodium. In vitro, Arp2/3 binds
The cell region immediately NMHC either dissociates myosin filaments or prevents to the sides of actin filaments and generates branches
behind the lamellipodium, their formation in vitro. A number of laboratories have at a fixed angle (70°) to form a dendritic network62.
characterized by the absence reported that ser1917 in rat NMHC IIA (equivalent to However, the in vivo data are still under debate. A recent
of dendritic actin and the
presence of longer, bundled
ser1916 in human NMHC IIA) is phosphorylated by study has reported actin filaments with variable angles
actin filaments and slow PKC, and phosphorylation of this site by PKCβ corre- (15°–90°) in the lamellipodium, which challenges the
retrograde flow. lates with exocytosis in mast cells56,57. ser1943 in the dendritic network notion63. The relative positioning of

NATuRE REvIEWs | Molecular cell BIology voLuME 10 | NovEMbER 2009 | 781

© 2009 Macmillan Publishers Limited. All rights reserved


REVIEWS

the lamellipodium and the lamellum is also unclear. It mediates this maturation, is not completely under-
was recently proposed that these two actin networks stood. The actin bundling function of NM II seems to
overlap at the leading edge, with the lamellipodium be sufficient, at least for the initial stages of adhesion
superimposed onto the lamellum64, but a more traditional maturation, because motor-deficient NM II mutants
view is that the lamellipodium is spatially anterior to the that still bind to actin support growth and elongation
lamellum61. In the lamellum, actin filaments coalesce of adhesions75.
into thicker bundles (FIG. 3a). The lamellipodium and the An integrin–actin linkage translates the effect of
lamellum are kinetically different: the lamellipodium is NM II to adhesions and mediates adhesion formation
distinguished by a fast retrograde flow of actin, whereas and maturation. Different adhesion proteins seem to
the lamellum exhibits slower retrograde flow. The participate in this linkage, including α-actinin, vinculin
convergent zone between the two is characterized by and talin (FIG. 4). There are two non-exclusive hypotheses
active depolymerization of the dendritic network and for how NM II mediates adhesion maturation. one is
the reorganization of actin64,65 (FIG. 3b). that NM II bundles actin filaments, and as a conse-
NM II does not reside in or seem to play a part in quence adhesion proteins at the ends of these actin fila-
the physical organization of the lamellipodium, but ments are brought together and clustered. This increases
it can affect the net rate of cellular protrusion 22,64,66 molecular interactions between adhesion proteins and
(FIG. 3b). When NM II is knocked out, knocked down results in increased integrin avidity (the combined
by small interfering RNA or inhibited with blebbistatin, strength of multiple integrin–ligand interactions) and
large actin bundles are not observed in the lamellum, signalling. For example, the actomyosin-driven binding
whereas the lamellipodium remains intact 64. In many of vinculin to talin induces integrin clustering 77. The sec-
cells, advancement of the protrusion is interrupted by ond hypothesis is that NM II-generated force induces
NM II-generated pauses67 that are absent when myosin conformational changes that expose cryptic binding
is inhibited or deleted22. or activation sites in key adhesion components 78–80.
one hypothesis for the role of NM II in protrusion Talin, an actin–integrin linkage protein, binds vinculin
formation is that NM II generates the retrograde flow of when mechanically stretched79 and is required to trans-
actin in the lamellum, which is connected to the lamelli- late NM II-generated forces to the substratum81 (FIG. 4).
podium64,68 (FIG. 3b). Inhibition of NM II activity with bleb- The adhesion scaffold protein CRK-associated sub-
bistatin, or genetic deletion of NM II, greatly decreases strate (p130CAs; also known as bCAR1) is also acti-
the rate of actin retrograde flow in the lamellum64,66 vated when stretched78. Furthermore, both ends of the
and inhibits coalescence of actin into proto-bundles adhesive linkage respond to force. Fibronectin under-
at the lamellipodium–lamellum interface69,70, which goes conformational changes induced by mechanical
increases protrusiveness22,66,71 (FIG. 3b). NM II-generated force that result in the exposure of its cryptic sites82,
retrograde flow counters the actin polymerization- and integrins are activated by tension80. Finally, NM II
mediated advancement of the leading edge and thereby itself can also undergo force-dependent conformational
reduces the observed protrusion rate. Thus, periodic changes83. Interestingly, the force-dependent modulation
contractions of the lamellipodium could cyclically inhibit of mechanoreactive NM II molecules also impinges on
the protrusion rate. In this model, the protrusion rate other signalling molecules that are not mechanosensi-
is the difference between the actin polymerization and tive themselves. It has recently been proposed that NM II
the retrograde flow rates. Adhesion modulates this bal- also controls EGF-induced paxillin phosphorylation
ance by coupling actin to the substratum, thereby creating and its subsequent dephosphorylation, which regulates
traction points that counter actin retrograde flow 72. such cellular protrusion and retraction84. In addition, zyxin
a mechanism presumably increases the rate of protrusion associates and dissociates from adhesions in response to
by diverting the forces created by the retrograde flow of extracellular forces85, and focal adhesion kinase (FAK),
the actin to the substratum. Thus, the linkage between a prominent adhesion-associated tyrosine kinase, is
actin and the substratum constitutes a ‘molecular clutch’. required for durotaxis (that is, the tendency of cells to
These observations, first made in neuronal growth cones migrate towards stiffer substrates)86.
but later extended to many other cell types, constitute NM II is also an integral part of the cellular response
the clutch hypothesis72–74, which integrates adhesion, to mechanical stimulation. It reacts to mechanical stim-
retrograde flow and actin polymerization. uli through cellular signalling pathways that regulate its
activation. In this way, NM II provides a convergence
Blebbistatin NM II in integrin-mediated adhesion. NM II is dis- point for external and cell-generated forces. For example,
A small-molecule inhibitor pensable for the assembly and disassembly of nascent application of external forces produces post-translational
with high affinity for myosin II adhesions inside the lamellipodium75,76. However, it is modifications such as phosphorylation, or conforma-
that blocks myosin in an
required for their subsequent maturation, which involves tional changes in different signalling molecules, which
actin-detached state.
elongation and growth. Adhesions mature along thin inhibit protrusion formation and lead to adhesion matu-
Pliability actin bundles that originate near the transition zone at ration and actin filament bundling 87. Conversely, cells
The mechanical properties the lamellipodium and lamellum interface75 (FIG. 3b). The in which NM II is inhibited do not respond to external
of the cellular environment. probability that a nascent adhesion in the lamellipodium forces88. In addition, cells sense substrate pliability, in
The environment can show low
pliability (that is, be elastic or
matures, rather than disassembles, seems to depend on part through the activation of NM II by RLC phos-
compliant) or high pliability the level of active NM II75. However, the mechanism by phorylation89,90. When cells are plated on low-pliability
(that is, be rigid or stiff). which adhesions elongate and grow, and how NM II substrates, NM II activation is low, actin is not highly

782 | NovEMbER 2009 | voLuME 10 www.nature.com/reviews/molcellbio

© 2009 Macmillan Publishers Limited. All rights reserved


REVIEWS

a Polarized migrating fibroblast


Trailing edge

c Trailing edge

Calpain-mediated
proteolysis (release)
Microtubule-mediated
relaxation
NM IIA

DI
Dynamin

AP
AP
Maturing

C B1
H1
adhesion

E
Actin
filament

NM IIA-mediated Microtubule
Leading edge
retraction

b Protrusion
Actin d Nucleus MRCK CDC42
filament Maturing
adhesion
NM IIB PAR3 PAR6

NM II GSK3 PKCζ
NM II-dependent, Stress +
slow retrograde flow fibre
Dorsal
arch
Lamellum
+
α-Actinin Dynein
Dynactin
Maturing Disassembling
adhesion Nucleus
adhesion Microtubule
Interface

NM II-independent,
fast retrograde flow

Lamellipodium Perinuclear
Dendritic Arp2/3 actin
actin Nascent adhesion complex MTOC

Figure 3 | Multiple roles of NM II in cell migration. a | A polarized, migrating fibroblast.


NatureAreas of the
Reviews cell in which
| Molecular Cell Biology
non-muscle myosin II (NM II) has an active role are boxed and expanded in parts b–d. b | NM II regulates retrograde flow
in the lamellum and promotes adhesion maturation, thereby limiting protrusion. Nascent adhesions form in the
lamellipodium, in which dendritic actin branching mediated by the Arp2/3 complex also occurs. At the lamellipodium–
lamellum interface, actin is depolymerized or bundled and adhesions disassemble or mature. A schematic of adhesions
maturing in the lamellum is also shown. NM II localizes to actin bundles contacting growing adhesions, forming a striated
pattern with α-actinin. In other cells, such as in neuronal growth cones, NM II may have a more direct role controlling
retrograde flow in the peripheral zone151. c | NM II participates in adhesion disassembly at the rear of the cell.
NM IIA-mediated contraction, calpain-dependent cleavage of adhesion components and microtubule targeting
coordinately induce adhesion disassembly. d | NM II has a role in nuclear positioning and orienting the microtubule-
organizing centre (MTOC) and Golgi, which are important for cell polarization. NM II is thought to act in concert with the
CDC42–partitioning defective 3 (PAR3) or PAR6–protein kinase Cζ (PKCζ) –glycogen synthase kinase 3 (GSK3) pathway to
polarize the cell. Myotonic dystrophy kinase-related CDC42-binding kinase (MRCK; also known as CDC42BP) activates
NM II and regulates its effect on nucleus repositioning. APC, adenomatous polyposis coli; DIAPH1, diaphanous 1;
EB1, end binding protein 1 (also known as MAPRE1).

NATuRE REvIEWs | Molecular cell BIology voLuME 10 | NovEMbER 2009 | 783

© 2009 Macmillan Publishers Limited. All rights reserved


REVIEWS

Actin Among other functions, Rac triggers actin polymer-


ization through the Arp2/3 complex and also inhibits
NM II NM II activation (FIG. 4). other signalling pathways activ-
Mechano-
responsive
ated by adhesion have the opposite effect and promote
molecules Integrin NM II activation through RHoA94,95. Rac is activated by
Vinculin
ROCK Talin signals generated in small adhesions close to the lead-
FAK
p130CAS
ing edge that actively undergo turnover and reassemble.
RHOA RHoA activation mediates actin filament formation and
Myosin AM MYPT1 adhesion maturation as the protrusion moves away
phosphatase PP1 and the adhesions become more central. This induces
further activation of NM II and the coalescence of thick
Arp2/3
actomyosin bundles. During adhesion maturation, Rac
Vinculin
RhoGEF Rac ix GIT signalling decreases and RHoA-mediated activation of
β-P
Pa
Src in
Zyx
in NM II increases, which results in enhanced actomyosin
xil

K
l

CRK p130CAS FA Talin


α-Actinin bundling 87,89,96.
DOCK180
NM II is a dual regulator of protrusion, through its
effects on actin retrograde flow and adhesion-generated
Plasma membrane signalling 22,66. Increased activation of NM II results
β-Integrin α-Integrin
subunit subunit in large actin bundles and stable adhesions, decreased
signalling to Rac and decreased protrusion. Lower levels
of active NM II result in less actin bundling and increased
ECM protrusion. This can explain, in part, the underlying
Figure 4 | NM II in integrin-mediated adhesion. Integrins that are bound to the differences in migration among different cell types.
extracellular matrix (ECM) are linked to the actin cytoskeleton through
Nature Reviews an actin
| Molecular Celllinkage
Biology
specifically, highly migratory cells such as leukocytes do
that is formed by multiple molecules, including talin, vinculin and α-actinin. Kinases such not exhibit large adhesions97, probably reflecting low levels
as focal adhesion kinase (FAK) and Src, and adaptors such as paxillin, are also recruited of NM II activation or an intrinsic inability to rearrange
and trigger the downstream activation of Rho GTPases such as Rac through adaptor and their actin cytoskeleton into large bundles, whereas slow
activating proteins. Representative pathways and associations are shown, including moving cells such as fibroblasts have adhesions that tend
the activation of Rac through paxillin by the CRK-associated substrate (p130CAS; also to mature into large, elongated structures as a result of
known as BCAR1)– CRK–dedicator of cytokinesis 1 (DOCK1; also known as DOCK180) NM II activation and robust actin bundling.
and G protein-coupled receptor kinase interacting ArfGAP (GIT)–β−Pix (also known as NM II activation promotes adhesion maturation
ARHGEF7) pathways. Activated Rac induces actin polymerization through the Arp2/3
through its actin bundling and contractile activities.
complex, which can also interact with some of the molecules of the actin linkage, such as
vinculin and FAK. Rac is also thought to locally inhibit NM II activation. The activation of Cycles of Rac and Rho activation inactivate and acti-
RhoGEFs by integrins, and the subsequent activation of RHOA and Rho-associated, vate NM II, resulting in protrusion and actin bundling,
coiled coil-containing kinase (ROCK), activates NM II. ROCK activates NM II directly respectively. The role of NM II in adhesion depends on
by phosphorylating the regulatory light chains (RLCs) or by inactivating myosin its ability to exert force on adhesions, even though it is
phosphatase, which in turn promotes RLC dephosphorylation. The pathways are spatially not physically present in the adhesions but attaches to
and temporally regulated. Additionally, the activation and inactivation of NM II itself the actin bundles with which adhesions are associated22.
affects adhesive signalling by triggering conformational changes in the mechano- Thus, NM II influences adhesion from a distance.
responsive molecules shown (pink boxes), which induces the clustering of the indicated
adhesion proteins (blue boxes) by reinforcing or weakening the linkage of the adhesion NM II determines the polarity of migrating cells.
and the actin cytoskeleton. AM, adaptor module; MYPT1, myosin phosphatase-targeting
Directional migration requires an initial symmetry break
subunit 1 (also known as PPP1R12A); PP1, protein phosphatase 1.
and the polarization of the migratory machinery. There
is strong evidence that NM II contractility initiates sym-
metry breaking by forming the prospective rear of the
bundled and adhesions are small. As rigidity increases, cell98–100, which allows for protrusion to occur at the oppo-
NM II activation increases, actin is visibly bundled and site end. The signals leading to specification of the cell
adhesions become large and elongated. rear and protrusion are spatially segregated101.
The leading protrusion resides at the cell front, and
Adhesive signalling in NM II activation. NM II influ- the cell rear often adopts a defined morphology, such
ences adhesive signalling through clustering and/or as flat and parallel to the leading edge in keratocytes, or
conformational changes, but adhesive signalling also lagging and triangular in some fibroblasts. The molecu-
controls NM II activation (FIG. 4). For example, integrin lar composition and organization of these front and rear
activation induces Tyr phosphorylation of the adhesion compartments is distinct 102, particularly with regards to
adaptor paxillin on Tyr31 and Tyr118 and of FAK on the actin cytoskeleton and the adhesions in these regions.
Tyr397. These phosphorylations trigger the activation The tail region is usually comprised of larger actin fila-
and recruitment to adhesions of signalling intermediates ment bundles that support adhesion disassembly and
such as the p130CAs–CRK and G protein-coupled recep- retraction of the rear, and large, stable adhesions that
tor kinase interacting ArfGAP (GIT)–β-Pix (also known do not signal to Rac. by contrast, adhesions at the front
as ARHGEF7) complexes, relaying activation signals to are smaller and more dynamic and mediate robust Rac
Rho GTPases, particularly Rac (reviewed in REFS 91–93). signalling, which drives protrusion.

784 | NovEMbER 2009 | voLuME 10 www.nature.com/reviews/molcellbio

© 2009 Macmillan Publishers Limited. All rights reserved


REVIEWS

1 Apical constriction the non-protrusive parts of the cell (the centre and the
(contraction) trailing edge), whereas dynamic filaments in protruding
regions of the cell are comprised of NM IIA alone 33.
NM IIA specifically mediates retraction of the trailing
edge during migration105 (FIG. 3c). When NM IIA is
inhibited, cells become elongated owing to their failure
to retract 105,106.
In addition to differences in actin and adhesion
organization and function, the position of the nucleus,
the microtubule-organizing centre (MToC) and the Golgi
Cadherin apparatus are also hallmarks of migratory cell polariza-
Contact RHOA tion. The MToC and Golgi apparatus reposition in front
3 Contact compaction (NM II)

initiation NM IIA of the nucleus towards the direction of protrusion 107.


(actin 2 Nuclear positioning Data from experiments in which NM II activity was
polymerization) (NM IIB)
inhibited by blebbistatin reveal that NM II also regulates
+ Rac-driven actin nucleus repositioning 108 (FIG. 3d). NM IIb seems to play a
polymerization prominent part in nuclear dynamics during migration as
Cadherin cells depleted of NM IIb exhibit multidirectional protru-
clustering
(NM IIA) sions109, lack a clear front and back, have a mislocalized
Nucleus
RHOA Golgi and MToC, exhibit inappropriate rotation of the
NM IIA nucleus and fail to reposition the nucleus properly dur-
ing polarized migration22. one hypothesis is that NM IIb
Actin directly anchors the perinuclear actin at the nucleus by
interacting with nuclear transmembrane receptors such
Cadherin signalling 4 Integrin as nesprins, which are involved in nuclear redistribu-
(contraction) clustering tion during cell migration110,111. Another possibility is
that NM IIb provides tension or structural rigidity to the
perinuclear actin and this force prevents inappropriate
rotation of the nucleus and moves it forwards (FIG. 3d).
Integrin

NM II in cell–cell adhesion and morphogenesis. NM II


ECM
Fibrillogenesis is not only pivotal in controlling integrin-mediated
adhesion and migration, it also regulates epithelial
Figure 5 | roles of NM II in epithelial cell polarization. The different roles of cell adhesion, polarization and morphogenesis. Well-
NatureNM
non-muscle myosin II (NM II) in epithelial cell polarization. Reviews | Molecular
II is involved Cell Biology
in apical
defined cell–cell junctions are a key feature of epithelial
constriction (step 1), which leads to important morphogenic movements such as dorsal
sheets and represent a different type of adhesive structure
closure (closure of the epidermis over the amnioserosa during embryogenesis) in
Drosophila melanogaster. In addition, NM II regulates nuclear positioning (step 2), in a that is controlled by NM II. Although these cell–cell junc-
similar manner to how it does this in fibroblasts (see FIG 3). NM II and RHOA signalling tions use cadherins as the main adhesion receptors, they
also stabilize cell–cell contacts by reinforcing them through actin cross-linking (known contain scaffolds and signalling intermediates analogous
as contact compaction; step 3). The initial contacts are formed as a result of Rac-driven to those found in integrin-mediated complexes. In aggre-
actin polymerization, but NM IIA is required for contact formation and reinforcement gates, epithelial cells have apical and basolateral regions
and cadherin clustering. NM II also mediates crosstalk between homophilic cadherin as well as integrin-based adhesions to the extracellular
contact-initiated signalling and extracellular matrix (ECM) remodelling triggered by matrix (ECM) at the basal surface (FIG. 5). Epithelial cell
integrin activation and clustering (step 4). sheets can move as multicellular cohorts, with the leading
cells showing protrusions and the trailing cells retract-
ing, or they can detach from the sheet and move away
NM II activation is essential for the formation of a as single cells. In free edges of the sheet, the migrating
defined rear compartment. NM II localizes to the poste- cells undergo a polarity switch from apicobasal (FIG. 5) to
rior part of migrating leukocytes and D. discoideum103,104. front–back (FIG. 3).
Here, it creates stable adhesions and highly bundled NM II controls the formation and stability of the
Microtubule-organizing actin, which prevent protrusion and thereby generate cell–cell junctions. NM IIA is required for cadherin
centre non-protrusive areas that constitute the sides and the clustering 112,113 (FIG. 5) and the stability of the cell–cell
A eukaryotic cell structure from trailing edge of the migrating cell33,98. junction. The initiation of the contact is driven by actin
which microtubules emanate.
In migrating fibroblasts, the NM II isoforms have polymerization114, and NM II-driven contractility medi-
During mitosis, the MTOC
organizes the mitotic spindle. different roles in cell polarization. NM IIA is dynamic ates the later stages of contact formation (compaction).
and assembles actomyosin bundles in protrusions22,66. Active RHoA and phosphorylated RLCs localize to the
Cadherin by contrast, NM IIb incorporates into preformed actin periphery of the junction and drive the contraction and
A member of a family of type I bundles and remains stationary, defining the centre compaction of the cell–cell contact 114 (FIG. 5). RHoA
transmembrane receptors that
mediate cell–cell adhesion
and rear of the migrating cell33. In this manner, the activation, which activates NM II, also mediates contact
through homophilic cooperative functions of NM IIA and NM IIb induce inhibition of movement (that is, the inhibition of pro-
interactions. big, non-dynamic actomyosin structures that define trusion by cell–cell contact)115. NM IIA probably plays

NATuRE REvIEWs | Molecular cell BIology voLuME 10 | NovEMbER 2009 | 785

© 2009 Macmillan Publishers Limited. All rights reserved


REVIEWS

a similar part during the formation of T cell–antigen- patients with these diseases were heterozygous for
presenting cell conjugates in antigenic presentation116. MYH9 mutations. one of the most common abnormali-
NM II-driven mechanisms also govern the three- ties in patients with MYH9-related diseases is associated
dimensional organization of epithelial tissues. In three with dysfunction of the blood platelets, which play an
dimensions, the force applied to a single cell is potentially important part in blood clotting and clot retraction, and
multi-directional, rather than planar or one-dimensional. affected patients suffer from macrothrombocytopenia.
The rigidity of the cellular microenvironment is crucial. Platelets from patients with mutated NM IIA have an
There is much evidence that NM II is part of the mecha- abnormal cytoskeletal composition and fail to form
nism by which the cell both generates and responds to and/or reorganize cytoskeletal structures on stimula-
force. Three dimensional responses have been studied in tion with the thrombin receptor activating peptide137.
Xenopus laevis and D. melanogaster during early embry- The initial shape change and internal contraction of the
onic development and organogenesis. In both systems, platelets following stimulation requires NM IIA activity,
the same general principles of regulation of NM II apply. which in turn is activated by RLC phosphorylation138,139.
In D. melanogaster, NM II (NMHC encoded by zipper) baculoviral-expressed catalytic fragments (heavy mero-
is activated by phosphorylation of the RLCs (encoded myosins) of NM IIA mutants that mimic the human
by spaghetti squash), which is under the control of Rho mutations in the motor domain (N93K and R702C
kinase (RoK)117–119. NM II is also implicated in epithelial in NMHC IIA), have a reduced actin-activated Mg 2+-
processes involving contraction, such as gastrulation120, ATPase activity and an inability to translocate actin fila-
trachea formation121,122, ventral furrow formation123,124, ments in vitro140. It is therefore likely that impairment
dorsal closure117 and morphogenesis of the vertebrate of NM IIA motor function contributes to the platelet
neural tube124–126, as well as in the reorganization of cell defects in these patients.
contacts in the plane of the epithelium during convergent both haploinsufficiency and the ability of mutant
extension127,128. In D. melanogaster eye imaginal disc myosin to interfere with wild-type myosin (dominant-
formation, morphogenetic signalling is under the con- negative effects) have been proposed as mechanisms by
trol of Hedgehog, which activates NM II through the which mutated NM IIA leads to MYH9-related diseases.
RHoA–RoK pathway 129. A reduction in wild-type NM IIA, accompanied by no
Finally, NM II provides the tension and traction that or very low levels of the mutant form of NM IIA, has
Convergent extension cells use to form and remodel a functional ECM during been observed in platelets and their precursors (mega-
A phase of gastrulation in tissue morphogenesis and wound repair. This includes karyocytes) from patients with MYH9-related diseases.
which layers of cells intercalate NM II-powered collagen fibre remodelling 130 and However, both haploinsufficiency and dominant-
(converge) and become longer fibronectin fibrillogenesis82,131. Recently, a link between negative effects have been reported in different tissues
(extend). Extension is driven by
a rearrangement of the cells of
cell–cell contact organization and fibrillo genesis from the same patient. Haploinsufficiency was shown
the ventral part of the was proposed. Actomyosin reorganization driven by to be the mechanism by which mutated NM IIA leads to
epithelium, which converge cadherins translates into ECM remodelling through MYH9-related diseases in platelets, and dominant-
towards the ventral midline. integrins, suggesting that cadherin-generated signals negative effects were shown to be the mechanism
regulate integrin activation by modulating cytoskeletal in granulocytes141,142. Mice with NM IIA specifically
Fibrillogenesis
A cell-induced reorganization tension132 (FIG. 5). ablated from megakaryocytes have thrombocytopenia
of the surrounding ECM with large platelets similar to those in patients with
molecules into bundled fibres. NM II mutations and disease MYH9-related diseases, as well as defects in cytoskeletal
An early indication that NM IIA, NM IIb and NM IIC reorganization143,144.
Macrothrombocytopenia
A condition that is
have different roles during development came from stud- so far there have been no reports of mutations in
characterized by enlarged ies in mice showing that disruption of the genes encod- MYH10, which encodes NMHC IIb, associated with
blood platelets that are ing the heavy chains of these proteins (MYH9, MYH10 human disease. To establish a putative mouse model
approximately the size of red and MYH14, respectively) leads to different phenotypes. for such diseases, mice carrying the R709C mutation
blood cells, are reduced in
NMHC IIA germline-ablated mice fail to form a polar- in NMHC IIb (a homologue of the R702C muta-
number and result in
prolonged bleeding times. ized visceral endoderm and show defects in cell–cell tion in human NMHC IIA) have been generated 145.
adhesion that result in death by embryonic day (E) 6.5, Homozygous mutant mice die by E14.5 and show
Haploinsufficiency before organ formation133. Mice in which NMHC IIb major morphogenetic defects, including a failure to
A state in which the loss of only has been deleted or mutated survive to E14.5 and show close the ventral body wall, resulting in a cleft palate,
one allele of a gene detectably
disables the encoded protein’s
cardiac and brain defects, which reflects the enrichment ectopia cordis and a large omphalocele, as well as defects
function. of the NM IIb isoform in these organs in wild-type in heart and diaphragm development (X.M. and R.s.A.,
mice134,135. As noted below, the brain defects were due to unpublished observations). These abnormalities are
Ectopia cordis failures in both cell adhesion and cell migration. reminiscent of a rare human syndrome known as
A congenital displacement of
To decipher the role of NM II in adhesion and migra- Pentalogy of Cantrell146.
the heart outside the thoracic
cavity and chest wall. tion, several investigators used mutants that mimic the Mice ablated for NM IIb, in contrast to mice with
mutations found in humans in MYH9 and MYH14 and point mutations in NMHC IIb, develop hydrocephalus
Omphalocele expressed the mutant NMHC IIA and NMHC IIC pro- (in which circulation of the cerebral spinal fluid is
A protrusion that occurs at teins in mice, D. melanogaster and cultured cells. To blocked), one of the most common congenital diseases
birth, whereby part of the
intestine protrudes through a
date, almost 40 mutations in NMHC IIA, distributed that affects humans (1–3 per 1,000 live births). This is
large defect in the abdominal among the head and rod domains, have been reported because loss of NM IIb causes disruption of apical cell–cell
wall at the umbilicus. to cause MYH9-related diseases136 (TABLE 1). All of the adhesion of the neuroepithelial cells that border the

786 | NovEMbER 2009 | voLuME 10 www.nature.com/reviews/molcellbio

© 2009 Macmillan Publishers Limited. All rights reserved


REVIEWS

Table 1 | the 39 MYH9 mutations associated with MYH9-related diseases* demonstrates markedly increased enzymatic motor
properties compared with NM IIb, as well as a decreased
exon Mutation clinical features (in addition to duty ratio (see above). Therefore it is likely that the
macrothrombocytopenia)
structural properties of NM IIb, rather than its enzy-
Head matic motor activity, are essential for maintaining the
Exon 1 W33C and P35A152 or A95T Döhle-like inclusions‡ integrity of cell–cell adhesion in the neuroepithelial cells
N93K Döhle-like inclusions and deafness that line the spinal canal10.
Finally, five mutations in MYH14, which encodes
S96L Deafness and nephritis
NMHC IIC, have been reported to occur in both the
Exon 10 K373N§ Döhle-like inclusions myosin head and tail domains. All of these mutations
Exon 16 R702C Döhle-like inclusions, deafness, nephritis and result in deafness148,149. The in vivo function of NM IIC
cataracts is largely unknown and mice ablated for NM IIC show
R702H Deafness and nephritis no obvious abnormalities (X.M. and R.s.A., unpublished
observations).
R705H Deafness
The pathophysiological roles of NM II are still not
Q706E or R718W|| Döhle-like inclusions fully understood. Although there is no evidence for
Rod heterodimer formation among the NMHC isoforms,
Exon 24 G1055_Q1068del153 Döhle-like inclusions, deafness and nephritis there is evidence for colocalization of NM II isoforms
in filaments. This raises the interesting possibility that a
E1066_A1072del Döhle-like inclusions, deafness and cataracts
mutant isoform might interfere with a second (or third)
E1066_A1072dup154 Döhle-like inclusions and cataracts wild-type isoform.
E1084del 153
Döhle-like inclusions studies from genetically manipulated mouse models
Exon 25 V1092_R1162del or Döhle-like inclusions demonstrate that NM II is involved in cell–cell adhesion
T1155A|| and cell migration in vivo. In maintaining the integrity of
S1114P Nephritis cell–cell adhesions, NM IIA and NM IIb are functionally
interchangeable and this function seems to be independent
T1155I Döhle-like inclusions, deafness and nephritis of their actin-activated Mg 2+-ATPase motor activity. In
Exon 26 R1165C Döhle-like inclusions, deafness and cataracts driving cell migration, however, the ATPase activity of
R1165L Döhle-like inclusions, deafness and nephritis the myosin motor is essential and NM IIA and NM IIb
cannot substitute for each other.
L1205_Q1207del Döhle-like inclusions
Exon 30 R1400W Nephritis Concluding remarks
D1424Y, D1424N, D1447V ||
Döhle-like inclusions Through its effects on actin bundling and contractility,
or D1447H NM II acts as a master integrator of the processes that
D1424H Döhle-like inclusions, deafness, nephritis and drive cell migration and adhesion. It is also an impor-
cataracts tant end point on which many signalling pathways
K910Q and D1424H155 Döhle-like inclusions, deafness, nephritis and converge, largely through Rho GTPases. NM II itself
hypertension is tightly regulated at different levels, including at the
Exon 31 V1516L Döhle-like inclusions level of folding, myosin filament assembly and disas-
sembly, actin binding and ATPase and motor activity.
Exon 37 I1816V Döhle-like inclusions and nephritis
The regulation of the actin cytoskeleton by NM II con-
Exon 38 E1841K Döhle-like inclusions, deafness, nephritis and trols multiple interrelated processes, such as migration,
cataracts
cell–cell and cell–matrix adhesion, cell differentiation,
Exon 40 G1924fs or D1925fs¶ Döhle-like inclusions tissue morphogenesis and development. Furthermore,
Non-helical tail NM II is at one end of the regulatory feedback loops
Exon 40 P1927fs, R1933fs|| or Döhle-like inclusions that control the activation of NM II’s own upstream
D1941fs signalling pathways, including the conformational activ-
ation of adhesion molecules and actin organization. The
R1933X Döhle-like inclusions and nephritis
importance of NM II is highlighted by the profound and
E1945X Döhle-like inclusions and deafness varied physiological effects of genetic deletion of NM II
*See also REF. 136. ‡Basophilic spindle-shaped inclusion bodies in the granulocytes. §Originally isoforms in animal models and the high number of
reported as K371N. ||Clinical features listed are based on common findings for myosin heavy
chain 9 (MYH9)-related diseases, but were not described in the reference. ¶Deletion of two naturally occurring mutations in the genes encoding the
different nucleotides both cause D1925fs. del, deletion; dup, duplication; fs, frame shift. NMHC II proteins that cause human disease.
The recent interest in the cellular and tissue functions
of NM II has produced many revealing and insightful
spinal canal147. Interestingly, this phenotype can be observations, although many basic and translational ques-
rescued by expression of the NMHC IIb point mutant tions remain. For example, the spatiotemporal regulation
R709C, which has a motor that is deficient in enzyme of NM II by subcellular localization and activation of its
activity, or by using gene substitution to replace NM regulating kinases in different cells and tissues has impor-
IIb with NM IIA29,147. NM IIA is normally not detected tant ramifications in controlling the NM II function but
in the neuroepithelial apical adhesion complex and needs further investigation. Emerging evidence strongly

NATuRE REvIEWs | Molecular cell BIology voLuME 10 | NovEMbER 2009 | 787

© 2009 Macmillan Publishers Limited. All rights reserved


REVIEWS

indicates that mechanical forces probably remodel well as migration, opens up the possibility of manipu-
the tumour cell microenvironment through NM II to lating both the rigidity of the microenvironment and the
affect tumour progression and metastasis150, but the expression and activation of NM II to guide cell prolifera-
precise mechanism by which NM II responds to and gen- tion and differentiation during tissue regeneration and
erates the microenvironment remains to be elucidated. stem cell transplantation. In this way, the manipulation of
In addition, the notion that NM II is a central integrator NM II and its regulators constitutes a potentially valuable
of external force during cell and tissue differentiation, as approach in regenerative medicine.

1. Holmes, K. C. in Myosins (ed. Coluccio, L. M.) 35–54 21. Kolega, J. Cytoplasmic dynamics of myosin IIA and IIB: dephosphorylation of a second site for myosin light
(Springer, The Netherlands, 2007). spatial ‘sorting’ of isoforms in locomoting cells. J. Cell chain kinase on the 20,000-dalton light chain of
Provides an in depth survey of selective myosin Sci. 111, 2085–2095 (1998). smooth muscle myosin. J. Biol. Chem. 261, 36–39
class members, including NM IIs. See also 22. Vicente-Manzanares, M., Zareno, J., Whitmore, L., (1986).
references 2, 3, 6, 7 and 9. Choi, C. K. & Horwitz, A. F. Regulation of protrusion, 39. Umemoto, S., Bengur, A. R. & Sellers, J. R. Effect of
2. Mooseker, M. S. & Foth, B. J. in Myosins adhesion dynamics, and polarity by myosins IIA and multiple phosphorylations of smooth muscle and
(ed. Coluccio, L. M.) 1–34 (Springer, The Netherlands, IIB in migrating cells. J. Cell Biol. 176, 573–580 cytoplasmic myosins on movement in an in vitro
2007). (2007). motility assay. J. Biol. Chem. 264, 1431–1436
3. El-Mezgueldi, M. & Bagshaw, C. R. in Myosins 23. Bao, J., Jana, S. S. & Adelstein, R. S. Vertebrate (1989).
(ed. Coluccio, L. M.) 55–93 (Springer, The nonmuscle myosin II isoforms rescue small interfering 40. Scholey, J. M., Taylor, K. A. & Kendrick-Jones, J.
Netherlands, 2007). RNA-induced defects in COS-7 cell cytokinesis. J. Biol. Regulation of non-muscle myosin assembly by
4. Richards, T. A. & Cavalier-Smith, T. Myosin domain Chem. 280, 19594–19599 (2005). calmodulin-dependent light chain kinase. Nature 287,
evolution and the primary divergence of eukaryotes. 24. Golomb, E. et al. Identification and characterization of 233–235 (1980).
Nature 436, 1113–1118 (2005). nonmuscle myosin II-C, a new member of the myosin II 41. Woodhead, J. L. et al. Atomic model of a myosin
5. Odronitz, F. & Kollmar, M. Drawing the tree of family. J. Biol. Chem. 279, 2800–2808 (2004). filament in the relaxed state. Nature 436,
eukaryotic life based on the analysis of 2,269 25. Kim, K. Y., Kovacs, M., Kawamoto, S., Sellers, J. R. & 1195–1199 (2005).
manually annotated myosins from 328 species. Adelstein, R. S. Disease-associated mutations and 42. Jung, H. S., Komatsu, S., Ikebe, M. & Craig, R.
Genome Biol. 8, R196 (2007). alternative splicing alter the enzymatic and motile Head–head and head–tail interaction: a general
6. Cremo, C. R. & Hartshorne, D. J. in Myosins (ed. activity of nonmuscle myosins II-B and II-C. J. Biol. mechanism for switching off myosin II activity in cells.
Coluccio, L. M.) 171–222 (Springer, The Netherlands, Chem. 280, 22769–22775 (2005). Mol. Biol. Cell 19, 3234–3242 (2008).
2007). 26. Kovacs, M., Wang, F., Hu, A., Zhang, Y. & Sellers, J. R. 43. Craig, R. & Woodhead, J. L. Structure and function
7. Reggiani, C. & Bottinelli, R. in Myosins (ed. Coluccio, Functional divergence of human cytoplasmic myosin II: of myosin filaments. Curr. Opin. Struct. Biol. 16,
L. M.) 125–169 (Springer, The Netherlands, 2007). kinetic characterization of the non-muscle IIA isoform. 204–212 (2006).
8. Clark, K., Langeslag, M., Figdor, C. G. & van Leeuwen, J. Biol. Chem. 278, 38132–38140 (2003). 44. Burgess, S. A. et al. Structures of smooth muscle
F. N. Myosin II and mechanotransduction: a balancing 27. Wang, F. et al. Kinetic mechanism of non-muscle myosin and heavy meromyosin in the folded, shutdown
act. Trends Cell Biol. 17, 178–186 (2007). myosin IIB: functional adaptations for tension state. J. Mol. Biol. 372, 1165–1178 (2007).
9. Conti, M. A., Kawamoto, S. & Adelstein, R. S. in generation and maintenance. J. Biol. Chem. 278, Together with references 35 and 41–43, this is an
Myosins (ed. Coluccio, L. M.) 223–264 (Springer, The 27439–27448 (2003). outstanding structural study that shows the
Netherlands, 2007). Together with reference 26, this paper details the transformation of the folded, blocked (10S) state
10. Conti, M. A. & Adelstein, R. S. Nonmuscle myosin II differences in the kinetic properties of NM IIA and to the unfolded, activated (6S) state of single NM II
moves in new directions. J. Cell Sci. 121, 11–18 NM IIB. These studies provide a mechanistic molecules following RLC phosphorylation. These
(2008). framework for understanding the putative in vivo studies also show the interaction of the two myosin
11. Krendel, M. & Mooseker, M. S. Myosins: tails (and function of two of the three NM II isoforms. heads in the inactive state.
heads) of functional diversity. Physiology 20, 28. Kovacs, M., Thirumurugan, K., Knight, P. J. & 45. Matsumura, F. Regulation of myosin II during
239–251 (2005). Sellers, J. R. Load-dependent mechanism of cytokinesis in higher eukaryotes. Trends Cell Biol. 15,
12. Swailes, N. T., Colegrave, M., Knight, P. J. & nonmuscle myosin 2. Proc. Natl Acad. Sci. USA 104, 371–377 (2005).
Peckham, M. Non-muscle myosins 2A and 2B drive 9994–9999 (2007). 46. Tan, I., Yong, J., Dong, J. M., Lim, L. & Leung, T.
changes in cell morphology that occur as myoblasts 29. Bao, J., Ma, X., Liu, C. & Adelstein, R. S. Replacement A tripartite complex containing MRCK modulates
align and fuse. J. Cell Sci. 119, 3561–3570 (2006). of nonmuscle myosin II-B with II-A rescues brain but lamellar actomyosin retrograde flow. Cell 135,
13. Yuen, S. L., Ogut, O. & Brozovich, F. V. Nonmuscle not cardiac defects in mice. J. Biol. Chem. 282, 123–136 (2008).
myosin is regulated during smooth muscle contraction. 22102–22111 (2007). 47. Matsumura, F. & Hartshorne, D. J. Myosin
Am. J. Physiol. Heart Circ. Physiol. 297, H191–H199 30. Nakasawa, T. et al. Critical regions for assembly of phosphatase target subunit: Many roles in cell
(2009). vertebrate nonmuscle myosin II. Biochemistry 44, function. Biochem. Biophys. Res. Commun. 369,
14. Morano, I. et al. Smooth-muscle contraction without 174–183 (2005). 149–156 (2008).
smooth-muscle myosin. Nature Cell Biol. 2, 371–375 31. Sato, M. K., Takahashi, M. & Yazawa, M. Two regions 48. Totsukawa, G. et al. Distinct roles of ROCK (Rho-
(2000). of the tail are necessary for the isoform-specific kinase) and MLCK in spatial regulation of MLC
The first demonstration that NM II can play a part functions of nonmuscle myosin IIB. Mol. Biol. Cell 18, phosphorylation for assembly of stress fibers and focal
in smooth muscle contraction. The authors make 1009–1017 (2007). adhesions in 3T3 fibroblasts. J. Cell Biol. 150,
use of smooth muscle myosin knockout mice to 32. Sandquist, J. C. & Means, A. R. The C-terminal tail 797–806 (2000).
study smooth muscle contraction and show that the region of nonmuscle myosin II directs isoform-specific 49. Totsukawa, G. et al. Distinct roles of MLCK and ROCK
sustained phase of contraction is due to NM II. See distribution in migrating cells. Mol. Biol. Cell 19, in the regulation of membrane protrusions and focal
reference 13 for recent work in this area which 5156–5167 (2008). adhesion dynamics during cell migration of fibroblasts.
suggests that NM IIB might contribute to the 33. Vicente-Manzanares, M., Koach, M. A., Whitmore, L., J. Cell Biol. 164, 427–439 (2004).
smooth muscle ‘latch’ state. Lamers, M. L. & Horwitz, A. F. Segregation and 50. Nishikawa, M., Sellers, J. R., Adelstein, R. S. &
15. Niederman, R. & Pollard, T. D. Human platelet myosin. activation of myosin IIB creates a rear in migrating Hidaka, H. Protein kinase C modulates in vitro
II. In vitro assembly and structure of myosin filaments. cells. J. Cell Biol. 183, 543–554 (2008). phosphorylation of the smooth muscle heavy
J. Cell Biol. 67, 72–92 (1975). 34. Somlyo, A. P. & Somlyo, A. V. Ca2+ sensitivity of meromyosin by myosin light chain kinase. J. Biol.
16. Svitkina, T. M., Verkhovsky, A. B. & Borisy, G. G. smooth muscle and nonmuscle myosin II: modulated Chem. 259, 8808–8814 (1984).
Improved procedures for electron microscopic by G proteins, kinases, and myosin phosphatase. 51. Komatsu, S. & Ikebe, M. The phosphorylation of
visualization of the cytoskeleton of cultured cells. Physiol. Rev. 83, 1325–1358 (2003). myosin II at the Ser1 and Ser2 is critical for normal
J. Struct. Biol. 115, 290–303 (1995). 35. Wendt, T., Taylor, D., Trybus, K. M. & Taylor, K. platelet-derived growth factor induced reorganization
17. Mansfield, S. G., al-Shirawi, D. Y., Ketchum, A. S., Three-dimensional image reconstruction of of myosin filaments. Mol. Biol. Cell 18, 5081–5090
Newbern, E. C. & Kiehart, D. P. Molecular organization dephosphorylated smooth muscle heavy meromyosin (2007).
and alternative splicing in Zipper, the gene that reveals asymmetry in the interaction between 52. Bosgraaf, L. & van Haastert, P. J. The regulation of
encodes the Drosophila non-muscle myosin II heavy myosin heads and placement of subfragment 2. Proc. myosin II in Dictyostelium. Eur. J. Cell Biol. 85,
chain. J. Mol. Biol. 255, 98–109 (1996). Natl Acad. Sci. USA 98, 4361–4366 (2001). 969–979 (2006).
18. Jana, S. S. et al. An alternatively spliced isoform of 36. Sellers, J. R., Eisenberg, E. & Adelstein, R. S. 53. Even-Faitelson, L. & Ravid, S. PAK1 and aPKCζ
nonmuscle myosin II-C is not regulated by myosin light The binding of smooth muscle heavy meromyosin to regulate myosin II-B phosphorylation: a novel signaling
chain phosphorylation. J. Biol. Chem. 284, actin in the presence of ATP. Effect of phosphorylation. pathway regulating filament assembly. Mol. Biol. Cell
11563–11571 (2009). J. Biol. Chem. 257, 13880–13883 (1982). 17, 2869–2881 (2006).
19. Li, Y., Lalwani, A. K. & Mhatre, A. N. Alternative splice 37. Hirata, N., Takahashi, M. & Yazawa, M. 54. Dulyaninova, N. G., Malashkevich, V. N., Almo, S. C. &
variants of MYH9. DNA Cell Biol. 27, 117–125 Diphosphorylation of regulatory light chain of Bresnick, A. R. Regulation of myosin-IIA assembly and
(2008). myosin IIA is responsible for proper cell spreading. Mts1 binding by heavy chain phosphorylation.
20. Maupin, P., Phillips, C. L., Adelstein, R. S. & Biochem. Biophys. Res. Commun. 381, 682–687 Biochemistry 44, 6867–6876 (2005).
Pollard, T. D. Differential localization of myosin-II (2009). 55. Clark, K. et al. TRPM7 regulates myosin IIA filament
isozymes in human cultured cells and blood cells. 38. Ikebe, M., Hartshorne, D. J. & Elzinga, M. stability and protein localization by heavy chain
J. Cell Sci. 107, 3077–3090 (1994). Identification, phosphorylation, and phosphorylation. J. Mol. Biol. 378, 790–803 (2008).

788 | NovEMbER 2009 | voLuME 10 www.nature.com/reviews/molcellbio

© 2009 Macmillan Publishers Limited. All rights reserved


REVIEWS

56. Conti, M. A., Sellers, J. R., Adelstein, R. S. & Elzinga, 79. del Rio, A. et al. Stretching single talin rod molecules 105. Chrzanowska-Wodnicka, M. & Burridge, K. Rho-
M. Identification of the serine residue phosphorylated activates vinculin binding. Science 323, 638–641 stimulated contractility drives the formation of stress
by protein kinase C in vertebrate nonmuscle myosin (2009). fibers and focal adhesions. J. Cell Biol. 133,
heavy chains. Biochemistry 30, 966–970 (1991). 80. Friedland, J. C., Lee, M. H. & Boettiger, D. 1403–1415 (1996).
57. Ludowyke, R. I. et al. Phosphorylation of nonmuscle Mechanically activated integrin switch controls α5β1 Implicates NM II in trailing edge retraction.
myosin heavy chain IIA on Ser1917 is mediated by function. Science 323, 642–644 (2009). 106. Worthylake, R. A., Lemoine, S., Watson, J. M. &
protein kinase C βII and coincides with the onset of 81. Jiang, G., Giannone, G., Critchley, D. R., Fukumoto, E. Burridge, K. RhoA is required for monocyte tail
stimulated degranulation of RBL-2H3 mast cells. & Sheetz, M. P. Two-piconewton slip bond between retraction during transendothelial migration. J. Cell
J. Immunol. 177, 1492–1499 (2006). fibronectin and the cytoskeleton depends on talin. Biol. 154, 147–160 (2001).
58. Li, Z. H. & Bresnick, A. R. The S100A4 metastasis Nature 424, 334–337 (2003). 107. Etienne-Manneville, S. & Hall, A. Integrin-mediated
factor regulates cellular motility via a direct interaction 82. Zhong, C. et al. Rho-mediated contractility exposes activation of Cdc42 controls cell polarity in migrating
with myosin-IIA. Cancer Res. 66, 5173–5180 (2006). a cryptic site in fibronectin and induces fibronectin astrocytes through PKCζ. Cell 106, 489–498
59. Clark, K. et al. The α-kinases TRPM6 and TRPM7, matrix assembly. J. Cell Biol. 141, 539–551 (2001).
but not eEF-2 kinase, phosphorylate the assembly (1998). 108. Gomes, E. R., Jani, S. & Gundersen, G. G. Nuclear
domain of myosin IIA, IIB and IIC. FEBS Lett. 582, 83. Schwaiger, I., Sattler, C., Hostetter, D. R. & Rief, M. movement regulated by Cdc42, MRCK, myosin, and
2993–2997 (2008). The myosin coiled-coil is a truly elastic protein actin flow establishes MTOC polarization in migrating
60. Ronen, D. & Ravid, S. Myosin II tailpiece determines structure. Nature Mater. 1, 232–235 (2002). cells. Cell 121, 451–463 (2005).
its paracrystal structure, filament assembly properties, 84. Schneider, I. C., Hays, C. K. & Waterman, C. M. Demonstrates the role of NM II in nuclear
and cellular localization. J. Biol. Chem. 284, Epidermal growth factor-induced contraction regulates repositioning during cell migration.
24948–24957 (2009). paxillin phosphorylation to temporally separate 109. Lo, C. M. et al. Nonmuscle myosin IIb is involved in the
61. Heath, J. P. & Holifield, B. F. Cell locomotion: new traction generation from de-adhesion. Mol. Biol. Cell guidance of fibroblast migration. Mol. Biol. Cell 15,
research tests old ideas on membrane and 20, 3155-3167 (2009). 982–989 (2004).
cytoskeletal flow. Cell. Motil. Cytoskeleton 18, 85. Yoshigi, M., Hoffman, L. M., Jensen, C. C., Yost, H. J. & 110. Warren, D. T., Zhang, Q., Weissberg, P. L. &
245–257 (1991). Beckerle, M. C. Mechanical force mobilizes zyxin from Shanahan, C. M. Nesprins: intracellular scaffolds
62. Pollard, T. D. & Borisy, G. G. Cellular motility driven by focal adhesions to actin filaments and regulates that maintain cell architecture and coordinate
assembly and disassembly of actin filaments. Cell 112, cytoskeletal reinforcement. J. Cell Biol. 171, 209–215 cell function? Expert Rev. Mol. Med. 7, 1–15
453–465 (2003). (2005). (2005).
63. Koestler, S. A., Auinger, S., Vinzenz, M., Rottner, K. & 86. Wang, H. B., Dembo, M., Hanks, S. K. & Wang, Y. 111. Nery, F. C. et al. TorsinA binds the KASH domain of
Small, J. V. Differentially oriented populations of actin Focal adhesion kinase is involved in mechanosensing nesprins and participates in linkage between
filaments generated in lamellipodia collaborate in during fibroblast migration. Proc. Natl Acad. Sci. USA nuclear envelope and cytoskeleton. J. Cell Sci. 121,
pushing and pausing at the cell front. Nature Cell Biol. 98, 11295–11300 (2001). 3476–3486 (2008).
10, 306–313 (2008). 87. Galbraith, C. G., Yamada, K. M. & Sheetz, M. P. The 112. Shewan, A. M. et al. Myosin 2 is a key Rho kinase
64. Ponti, A., Machacek, M., Gupton, S. L., Waterman- relationship between force and focal complex target necessary for the local concentration of
Storer, C. M. & Danuser, G. Two distinct actin networks development. J. Cell Biol. 159, 695–705 (2002). E-cadherin at cell-cell contacts. Mol. Biol. Cell 16,
drive the protrusion of migrating cells. Science 305, 88. Chen, C. S. Mechanotransduction — a field pulling 4531–4542 (2005).
1782–1786 (2004). together? J. Cell Sci. 121, 3285–3292 (2008). 113. Ivanov, A. I. et al. A unique role for nonmuscle myosin
Describes the kinetic and compositional 89. Beningo, K. A., Dembo, M., Kaverina, I., Small, J. V. & heavy chain IIA in regulation of epithelial apical
distinction between the lamellipodium and the Wang, Y. L. Nascent focal adhesions are responsible junctions. PLoS ONE 2, e658 (2007).
lamellum — the two actin networks in for the generation of strong propulsive forces in 114. Yamada, S. & Nelson, W. J. Localized zones of Rho
protrusions. migrating fibroblasts. J. Cell Biol. 153, 881–888 and Rac activities drive initiation and expansion of
65. Delorme, V. et al. Cofilin activity downstream of Pak1 (2001). epithelial cell-cell adhesion. J. Cell Biol. 178,
regulates cell protrusion efficiency by organizing 90. Beningo, K. A., Hamao, K., Dembo, M., Wang, Y. L. & 517–527 (2007).
lamellipodium and lamella actin networks. Dev. Cell Hosoya, H. Traction forces of fibroblasts are regulated 115. Carmona-Fontaine, C. et al. Contact inhibition of
13, 646–662 (2007). by the Rho-dependent kinase but not by the myosin locomotion in vivo controls neural crest directional
66. Cai, Y. et al. Nonmuscle myosin IIA-dependent force light chain kinase. Arch. Biochem. Biophys. 456, migration. Nature 456, 957–961 (2008).
inhibits cell spreading and drives F-actin flow. Biophys. 224–231 (2006). 116. Ilani, T., Vasiliver-Shamis, G., Vardhana, S.,
J. 91, 3907–3920 (2006). 91. Parsons, J. T. Focal adhesion kinase: the first ten years. Bretscher, A. & Dustin, M. L. T cell antigen receptor
67. Giannone, G. et al. Periodic lamellipodial contractions J. Cell Sci. 116, 1409–1416 (2003). signaling and immunological synapse stability
correlate with rearward actin waves. Cell 116, 92. Deakin, N. O. & Turner, C. E. Paxillin comes of age. require myosin IIA. Nature Immunol. 10, 531–539
431–443 (2004). J. Cell Sci. 121, 2435–2444 (2008). (2009).
68. Giannone, G. et al. Lamellipodial actin mechanically 93. Mitra, S. K. & Schlaepfer, D. D. Integrin-regulated 117. Young, P. E., Richman, A. M., Ketchum, A. S. &
links myosin activity with adhesion-site formation. FAK–Src signaling in normal and cancer cells. Curr. Kiehart, D. P. Morphogenesis in Drosophila requires
Cell 128, 561–575 (2007). Opin. Cell Biol. 18, 516–523 (2006). nonmuscle myosin heavy chain function. Genes Dev. 7,
69. Anderson, T. W., Vaughan, A. N. & Cramer, L. P. 94. Geiger, B., Spatz, J. P. & Bershadsky, A. D. 29–41 (1993).
Retrograde flow and myosin II activity within the Environmental sensing through focal adhesions. 118. Karess, R. E. et al. The regulatory light chain of
leading cell edge deliver F-actin to the lamella to seed Nature Rev. Mol. Cell Biol. 10, 21–33 (2009). nonmuscle myosin is encoded by spaghetti‑squash, a
the formation of graded polarity actomyosin II filament 95. Vicente-Manzanares, M., Choi, C. K. & Horwitz, A. R. gene required for cytokinesis in Drosophila. Cell 65,
bundles in migrating fibroblasts. Mol. Biol. Cell 19, Integrins in cell migration — the actin connection. 1177–1189 (1991).
5006–5018 (2008). J. Cell Sci. 122, 199–206 (2009). 119. Winter, C. G. et al. Drosophila Rho-associated kinase
70. Nemethova, M., Auinger, S. & Small, J. V. Building the 96. Ballestrem, C. et al. Molecular mapping of tyrosine- (Drok) links Frizzled-mediated planar cell polarity
actin cytoskeleton: filopodia contribute to the phosphorylated proteins in focal adhesions using signaling to the actin cytoskeleton. Cell 105, 81–91
construction of contractile bundles in the lamella. fluorescence resonance energy transfer. J. Cell Sci. (2001).
J. Cell Biol. 180, 1233–1244 (2008). 119, 866–875 (2006). 120. Skoglund, P., Rolo, A., Chen, X., Gumbiner, B. M. &
71. Even-Ram, S. et al. Myosin IIA regulates cell motility 97. Smith, A. et al. A talin-dependent LFA-1 focal zone is Keller, R. Convergence and extension at gastrulation
and actomyosin-microtubule crosstalk. Nature Cell formed by rapidly migrating T lymphocytes. J. Cell require a myosin IIB-dependent cortical actin network.
Biol. 9, 299–309 (2007). Biol. 170, 141–151 (2005). Development 135, 2435–2444 (2008).
Establishes the role of NM IIA in the co-regulation 98. Verkhovsky, A. B., Svitkina, T. M. & Borisy, G. G. Self- 121. Brodu, V. & Casanova, J. The RhoGAP crossveinless-c
of actin and microtubule functions in motile polarization and directional motility of cytoplasm. links trachealess and EGFR signaling to cell shape
cells. Curr. Biol. 9, 11–20 (1999). remodeling in Drosophila tracheal invagination.
72. Mitchison, T. & Kirschner, M. Cytoskeletal dynamics 99. Yam, P. T. et al. Actin-myosin network reorganization Genes Dev. 20, 1817–1828 (2006).
and nerve growth. Neuron 1, 761–772 (1988). breaks symmetry at the cell rear to spontaneously 122. Myat, M. M. Making tubes in the Drosophila embryo.
73. Lin, C. H. & Forscher, P. Growth cone advance is initiate polarized cell motility. J. Cell Biol. 178, Dev. Dyn. 232, 617–632 (2005).
inversely proportional to retrograde F-actin flow. 1207–1221 (2007). 123. Barrett, K., Leptin, M. & Settleman, J. The Rho
Neuron 14, 763–771 (1995). 100. Mseka, T., Bamburg, J. R. & Cramer, L. P. ADF/cofilin GTPase and a putative RhoGEF mediate a signaling
74. Jay, P. Y., Pham, P. A., Wong, S. A. & Elson, E. L. family proteins control formation of oriented actin- pathway for the cell shape changes in Drosophila
A mechanical function of myosin II in cell motility. filament bundles in the cell body to trigger fibroblast gastrulation. Cell 91, 905–915 (1997).
J. Cell Sci. 108, 387–393 (1995). polarization. J. Cell Sci. 120, 4332–4344 (2007). 124. Pilot, F. & Lecuit, T. Compartmentalized
75. Choi, C. K. et al. Actin and α-actinin orchestrate the 101. Xu, J. et al. Divergent signals and cytoskeletal morphogenesis in epithelia: from cell to tissue shape.
assembly and maturation of nascent adhesions in a assemblies regulate self-organizing polarity in Dev. Dyn. 232, 685–694 (2005).
myosin II motor-independent manner. Nature Cell Biol. neutrophils. Cell 114, 201–214 (2003). 125. Rolo, A., Skoglund, P. & Keller, R. Morphogenetic
10, 1039–1050 (2008). 102. Ridley, A. J. et al. Cell migration: integrating signals movements driving neural tube closure in Xenopus
76. Alexandrova, A. Y. et al. Comparative dynamics of from front to back. Science 302, 1704–1709 require myosin IIB. Dev. Biol. 327, 327–338
retrograde actin flow and focal adhesions: formation of (2003). (2009).
nascent adhesions triggers transition from fast to slow 103. Eddy, R. J., Pierini, L. M., Matsumura, F. & 126. Hildebrand, J. D. & Soriano, P. Shroom, a PDZ
flow. PLoS ONE 3, e3234 (2008). Maxfield, F. R. Ca2+-dependent myosin II activation is domain-containing actin-binding protein, is required
77. Humphries, J. D. et al. Vinculin controls focal adhesion required for uropod retraction during neutrophil for neural tube morphogenesis in mice. Cell 99,
formation by direct interactions with talin and actin. migration. J. Cell Sci. 113, 1287–1298 (2000). 485–497 (1999).
J. Cell Biol. 179, 1043–1057 (2007). 104. Kolega, J. Asymmetry in the distribution of free 127. Bertet, C., Sulak, L. & Lecuit, T. Myosin-dependent
78. Sawada, Y. et al. Force sensing by mechanical versus cytoskeletal myosin II in locomoting junction remodelling controls planar cell
extension of the Src family kinase substrate p130Cas. microcapillary endothelial cells. Exp. Cell Res. 231, intercalation and axis elongation. Nature 429,
Cell 127, 1015–26 (2006). 66–82 (1997). 667–671 (2004).

NATuRE REvIEWs | Molecular cell BIology voLuME 10 | NovEMbER 2009 | 789

© 2009 Macmillan Publishers Limited. All rights reserved


REVIEWS

128. Yamamoto, N., Okano, T., Ma, X., Adelstein, R. S. & 140. Hu, A., Wang, F. & Sellers, J. R. Mutations in human 150. Butcher, D. T., Alliston, T. & Weaver, V. M. A tense
Kelley, M. W. Myosin II regulates extension, growth nonmuscle myosin IIA found in patients with May- situation: forcing tumour progression. Nature Rev.
and patterning in the mammalian cochlear duct. Hegglin anomaly and Fechtner syndrome result in Cancer 9, 108–122 (2009).
Development 136, 1977–1986 (2009). impaired enzymatic function. J. Biol. Chem. 277, 151. Medeiros, N. A., Burnette, D. T. & Forscher, P.
129. Corrigall, D., Walther, R. F., Rodriguez, L., Fichelson, P. 46512–46517 (2002). Myosin II functions in actin-bundle turnover in
& Pichaud, F. Hedgehog signaling is a principal 141. Kunishima, S., Hamaguchi, M. & Saito, H. Differential neuronal growth cones. Nature Cell Biol. 8, 215–226
inducer of Myosin-II-driven cell ingression in expression of wild-type and mutant NMMHC-IIA (2006).
Drosophila epithelia. Dev. Cell 13, 730–742 polypeptides in blood cells suggests cell-specific 152. Miyajima, Y. & Kunishima, S. Identification of the first
(2007). regulation mechanisms in MYH9 disorders. Blood in cis mutations in MYH9 disorder. Eur. J. Haematol.
130. Meshel, A. S., Wei, Q., Adelstein, R. S. & Sheetz, M. P. 111, 3015–3023 (2008). 82, 288–291 (2009).
Basic mechanism of three-dimensional collagen fibre 142. Pecci, A. et al. Pathogenetic mechanisms of 153. Miyazaki, K., Kunishima, S., Fujii, W. & Higashihara,
transport by fibroblasts. Nature Cell Biol. 7, 157–164 hematological abnormalities of patients with MYH9 M. Identification of three in-frame deletion
(2005). mutations. Hum. Mol. Genet. 14, 3169–3178 mutations in MYH9 disorders suggesting an
131. Zhang, Q., Magnusson, M. K. & Mosher, D. F. (2005). important hot spot for small rearrangements in
Lysophosphatidic acid and microtubule-destabilizing This study analyses 11 patients from 6 families, MYH9 exon 24. Eur. J. Haematol. 83, 230–234
agents stimulate fibronectin matrix assembly through with 6 different NM IIA mutations, and provides (2009).
Rho-dependent actin stress fiber formation and cell evidence that defects in the megakaryocytic lineage 154. De Rocco, D. et al. Identification of the first duplication
contraction. Mol. Biol. Cell 8, 1415–1425 (1997). arise from haploinsufficiency of NM IIA, whereas in MYH9-related disease: A hot spot for hot unequal
132. Dzamba, B. J., Jakab, K. R., Marsden, M., the inclusion bodies in granulocytes are due to the crossing-over within exon 24 of the MYH9 gene. Eur.
Schwartz, M. A. & DeSimone, D. W. Cadherin mutant form of NM IIA interfering with the J. Med. Genet. 52, 191–194 (2009).
adhesion, tissue tension, and noncanonical Wnt wild-type form. 155. Capria, M. et al. Double nucleotidic mutation of the
signaling regulate fibronectin matrix organization. 143. Leon, C. et al. Megakaryocyte-restricted MYH9 MYH9 gene in a young patient with end-stage renal
Dev. Cell 16, 421–432 (2009). inactivation dramatically affects hemostasis while disease. Nephrol Dial Transplant 19, 249–251
133. Conti, M. A., Even-Ram, S., Liu, C., Yamada, K. M. & preserving platelet aggregation and secretion. Blood (2004).
Adelstein, R. S. Defects in cell adhesion and the 110, 3183–3191 (2007).
visceral endoderm following ablation of nonmuscle 144. Eckly, A. et al. Abnormal megakaryocyte morphology Acknowledgements
myosin heavy chain II-A in mice. J. Biol. Chem. 279, and proplatelet formation in mice with megakaryocyte- The authors apologize for the numerous studies left out owing
41263–41266 (2004). restricted MYH9 inactivation. Blood 113, 3182–3189 to journal limits on the number of references. We thank M. A.
134. Tullio, A. N. et al. Nonmuscle myosin II-B is required (2008). Conti and J. R. Sellers (National Heart, Lung and Blood
for normal development of the mouse heart. Proc. 145. Ma, X., Kawamoto, S., Hara, Y. & Adelstein, R. S. Institute (NHLBI)) for helpful suggestions and critical reading
Natl Acad. Sci. USA 94, 12407–12412 (1997). A point mutation in the motor domain of of the manuscript. This work was supported by the US
135. Tullio, A. N. et al. Structural abnormalities develop in nonmuscle myosin II-B impairs migration of distinct National Institutes of Health (NIH) grant GM23244, the Cell
the brain after ablation of the gene encoding groups of neurons. Mol. Biol. Cell 15, 2568–2579 Migration Consortium grant U54-GM064346 (A.R.H.), and
nonmuscle myosin II-B heavy chain. J. Comp. Neurol. (2004). the Division of Intramural Research, NHLBI, NIH (R.S.A.).
433, 62–74 (2001). 146. Cantrell, J. R., Haller, J. A. & Ravitch, M. M. A
136. Burt, R. A., Joseph, J. E., Milliken, S., Collinge, J. E. & syndrome of congenital defects involving the
Kile, B. T. Description of a novel mutation leading to abdominal wall, sternum, diaphragm, pericardium, DataBases
MYH9-related disease. Thromb. Res. 122, 861–863 and heart. Surg. Gynecol. Obstet. 107, 602–614 Entrez Gene: http://www.ncbi.nlm.nih.gov/entrez/query.
(2008). (1958). fcgi?db=gene
137. Canobbio, I. et al. Altered cytoskeleton organization in 147. Ma, X., Bao, J. & Adelstein, R. S. Loss of cell adhesion MYH9 | MYH10 | MYH14 | MYL9
platelets from patients with MYH9-related disease. causes hydrocephalus in nonmuscle myosin II-B-ablated UniProtKB: http://www.uniprot.org
J. Thromb. Haemost. 3, 1026–1035 (2005). and mutated mice. Mol. Biol. Cell 18, 2305–2312 EGF | MLCK | MYPT1 | p130CAS | PAK1 | RHOA | S100A4 |
138. Johnson, G. J., Leis, L. A., Krumwiede, M. D. & (2007). TRPM7 | ZIPK | zyxin
White, J. G. The critical role of myosin IIA in platelet 148. Donaudy, F. et al. Nonmuscle myosin heavy-chain gene
internal contraction. J. Thromb. Haemost. 5, MYH14 is expressed in cochlea and mutated in FUrtHer INForMatIoN
1516–1529 (2007). patients affected by autosomal dominant hearing Alan Rick Horwitz’s homepage: http://people.virginia.
139. Calaminus, S. D. et al. MyosinIIa contractility is impairment (DFNA4). Am. J. Hum. Genet. 74, edu/~afh2n/
required for maintenance of platelet structure during 770–776 (2004). Robert S. Adelstein’s homepage: http://dir.nhlbi.nih.gov/
spreading on collagen and contributes to thrombus 149. Yang, T. et al. Genetic heterogeneity of deafness labs/lmc
stability. J. Thromb. Haemost. 5, 2136–2145 phenotypes linked to DFNA4. Am. J. Med. Genet. A all lINks are actIve IN the oNlINe pDf
(2007). 139, 9–12 (2005).

790 | NovEMbER 2009 | voLuME 10 www.nature.com/reviews/molcellbio

© 2009 Macmillan Publishers Limited. All rights reserved

You might also like