You are on page 1of 3

Immunity

Previews

successive exposure to the same influ- moral immune response in the develop- T.B., and Kelsoe, G. (2016). Immunity 44, this
issue, 542–552.
enza vaccine in humans are recalled ment of broadly protective vaccines.
from the same B cell clonotypes (Andrews McKean, D., Huppi, K., Bell, M., Staudt, L., Ger-
et al., 2015). It is possible that this hard, W., and Weigert, M. (1984). Proc. Natl.
REFERENCES
Acad. Sci. U.S.A. 81, 3180–3184.
discrepancy is due to prior antigen expe-
rience in humans and the early GC Andrews, S.F., Huang, Y., Kaur, K., Popova, L.I., Victora, G.D., and Nussenzweig, M.C. (2012).
Ho, I.Y., Pauli, N.T., Henry Dunand, C.J., Taylor, Annu. Rev. Immunol. 30, 429–457.
response being dominated by memory B
W.M., Lim, S., Huang, M., et al. (2015). Sci. Transl.
cells, which have faster response kinetics Med. 7, 316ra192. Victora, G.D., and Wilson, P.C. (2015). Cell 163,
than the naive B cells. It is not clear how 545–548.
Clarke, S.H., Huppi, K., Ruezinsky, D., Staudt, L.,
this would alter the GC response, but Gerhard, W., and Weigert, M. (1985). J. Exp. Victora, G.D., Schwickert, T.A., Fooksman, D.R.,
these results suggest that there is a point Med. 161, 687–704. Kamphorst, A.O., Meyer-Hermann, M., Dustin,
M.L., and Nussenzweig, M.C. (2010). Cell 143,
where repeated antigen exposure no
Clarke, S.H., Staudt, L.M., Kavaler, J., Schwartz, 592–605.
longer results in increased diversity. D., Gerhard, W.U., and Weigert, M.G. (1990).
Perhaps future research, taking into ac- J. Immunol. 144, 2795–2801. Wrammert, J., Smith, K., Miller, J., Langley, W.A.,
Kokko, K., Larsen, C., Zheng, N.-Y., Mays, I.,
count these new findings, will finally Kuraoka, M., Schmidt, A.G., Nojima, T., Feng, F., Garman, L., Helms, C., et al. (2008). Nature 453,
reveal how to properly harness the hu- Watanabe, A., Kitamura, D., Harrison, S.C., Kepler, 667–671.

Silence of the ROS


Judith Agudo1 and Brian D. Brown1,2,*
1Genetics and Genomic Sciences Department, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
2Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
*Correspondence: brian.brown@mssm.edu
http://dx.doi.org/10.1016/j.immuni.2016.02.027

Reactive oxygen species (ROS) are generated during T cell activation and serve a signaling function but can
also be damaging. In this issue of Immunity, Zhang et al. (2016) show that miR-23a prevents ROS-elicited
necrosis by suppressing cyclophilin D (PPIF), a regulator of ROS escape from mitochondria.

T cell activation sets off a signaling T cells are believed to be due to pro- signaling while preventing ROS-induced
cascade that leads to proliferation and duction by NOX enzymes and the conse- pathological damage. How this is done
production of cytokines, which are key quence of a more active metabolic cell is not well understood.
to the effector phase of the immune state. In this issue of Immunity, Zhang et al.
response. T cell activation also causes ROS are now known to be important for (2016) set out to better understand what
the accumulation of intracellular reactive intracellular signaling through their ability role microRNA (miRNA) have in the CD4+
oxygen species (ROS) (Devadas et al., to mediate reversible oxidation of pro- T cell response to infection. miRNAs are
2002). ROS are highly reactive molecules teins by reacting with the thiol groups small non-coding RNAs that complex
derived from oxygen (O2) and mostly of cysteine residues (Sena and Chandel, with Argonaute proteins to form RNA-
include superoxide (O2) and hydrogen 2012). The ROS produced during acti- induced silencing complexes (RISC). The
peroxide (H2O2). The majority of intracel- vation affects T cell function by modifying miRNA guides RISC to sequences en-
lular ROS are produced either by NAPDH phosphatases, such as Shp1, as well coded on mRNAs that are partially com-
oxydases (NOX) or by the electron trans- as other proteins (Simeoni and Bogeski, plementary to the miRNA (Pasquinelli,
port chain (ETC). NOX enzymes produce 2015). These changes contribute to 2012). miRISC binding prevents transla-
ROS in the cytoplasm by transferring increased cytokine production and prolif- tion of the mRNA and can also accelerate
one electron to oxygen, forming O2, eration. However, ROS can also damage decay of the transcript. The outcome of
which can further react to form H2O2. cell components through oxidative stress miRNA regulation is decreased protein
ROS are also formed in the mitochondria and ultimately lead to cell death. Thus, translation of its target genes. There are
as a by-product of ETC. These ROS accu- although ROS have an important role in more than 1,000 miRNAs encoded in
mulate inside the mitochondria and can T cell activation and proliferation, they the mammalian genome, many of which
be released to the cytoplasm through can also be harmful to the T cells. T cells are completely conserved from mouse
the permeability transition pore (PTP). must somehow balance ROS amounts to humans. miRNAs have been found
The increased ROS amounts in activated within the cell to enable ROS-mediated to regulate different aspects of T cell

520 Immunity 44, March 15, 2016 ª2016 Elsevier Inc.


Immunity

Previews

generally occurs subsequent to initial


T cell expansion, and thus the early death
of the activated Mir23afl/fl-cre CD4+
T cells was unexpected. Further analysis
indicated that the cells were dying not
by apoptosis but by necrosis associated
with mitochondrial swelling and rupture.
This appeared to be caused by excessive
ROS, because overexpression of the
ROS scavenger GPX1 rescued activated
Mir23afl/fl-cre CD4+ T cells from prema-
ture necrotic death. These findings indi-
cate that early after T cell activation, mito-
chondrial ROS rise to a threshold that
can precipitate necrosis, but that this is
controlled by the concomitant increase
in miR-23a. This helps explain why
Listeria infection of the Mir23afl/fl-cre
Figure 1. miR-23a Regulates ROS Leakage from the Mitochondria of Activated T Cells mice led to liver pathology and sys-
The amount of reactive oxygen species (ROS) in the mitochondria increases upon CD4+ T cell activation
due to Ca2+ influx and increased metabolism. ROS can leak out through the mitochondria permeability temic inflammation because the activated
transition pore (PTP), which is opened by peptidylprolyl isomerase F (PPIF or Cyclophilin D) (right). Mir23afl/fl-cre T cells were unable to pre-
Excessive ROS release can lead to cell necrosis. To prevent this, T cells increase miR-23a concentration vent ROS-induced necrosis.
during activation, which then suppresses PPIF by binding to PPIF’s 30 UTR and preventing translation. The
reduction in PPIF keeps the mitonchondria PTP closed and reduces the escape of ROS (left), which
Because miRNAs regulate mRNA, and
maintains CD4+ T cell survival during the early hypermetabolic and inflammatory state that accompanies not ROS, Zhang et al. (2016) looked for
activation. predicted targets of miR-23a that are
known to have a role in necrosis. They
biology, including T cell development, expression of miR-27a or miR-24. Loss of found two conserved target sites for
proliferation, and cytokine production, miR-23a did not affect T cell development miR-23a in the 30 UTR of peptidylprolyl
but the role of most miRNAs expressed or homeostatic maintenance, as shown isomerase F (PPIF or Cyclophilin D), which
in T cells is still unknown. by the facts that T cell numbers were is a vital pore-opening regulator of
Zhang et al. (2016) started by profiling normal in the Mir23afl/fl-cre mice and that the PTP that controls ROS flow from
miRNA expression in Listeria-specific the cells proliferated similar to wild-type the mitochondria (Baines et al., 2005).
CD4+ T cells (LLO118 TCR trans- T cells in response to activation. Quite They confirmed that the PPIF mRNA
genic) at different time points after infec- strikingly, the Mir23afl/fl-cre mice died was a direct target of miR-23a and
tion of mice with Listeria monocyto- within 4–7 days of being infected with a that the PPIF protein was significantly
genes. They found that there was a dose of Listeria that was relatively well upregulated in miR-23a-deficient cells.
group of 45 miRNAs whose expres- tolerated by wild-type mice. Surprisingly, Importantly, when they reversed this
sion gradually increased during the early mortality was not the consequence of upregulation by silencing the expression
expansion phase of activation, then increased bacterial loads; the Listeria of PPIF in Mir23afl/fl-cre cells, they re-
rapidly decreased during contraction, colonies were actually lower in Mir23afl/fl- balanced the intracellular ROS amounts
and increased again as the effector cre mice compared to wild-type mice. and prevented the activated T cells from
T cells transitioned to memory. Among Instead, Zhang et al. (2016) found that prematurely dying. Thus, it appears that
this group was miR-23a, which is en- there were large areas of necrosis in the PPIF, and by extension mitochondrial
coded in the genome as a part of a miRNA liver and elevations of the inflammatory release of ROS, is the main regulatory
cluster that also contains Mir27a and cytokines IL-6 and TNF in the serum of target of miR-23a in preventing T cell ne-
Mir24 and has a paralog cluster en- the Mir23afl/fl-cre mice infected with Liste- crosis during the early effector phase of
coded elsewhere in the genome that is ria, which suggested that the cause of activation (Figure 1). This helps to explain
comprised of Mir23b, Mir27b, and Mir24. death was due to a severe inflammatory how T cells handle the burden of rapid
The three miRNAs in each cluster are response. Closer analysis indicated that ROS production during the high meta-
co-expressed on a polycistronic tran- miR-23a-deficient CD4+ T cells died at a bolic demands of activation and prolifera-
script, but each has a different sequence substantially higher frequency than wild- tion. It will be interesting to explore the
and thus target different sequences. type cells, and as quickly as 6 hr after acti- relevance of miR-23a in CD4+ T cell sur-
To determine whether miR-23a had any vation. Thus, they found that miR-23a was vival in additional contexts associated
function in CD4+ T cells, Zhang et al. (2016) important for the survival of activated with high amounts of ROS, such as radia-
generated mice in which Mir23a was spe- T cells. tion therapy and cancer.
cifically ablated in CD4+ cells by floxing T cell death is a normal and pro- Recently, two different groups have
Mir23a and crossing with Cd4-cre mice grammed consequence of T cell activa- shown that deletion of the entire Mir23a-
(Mir23afl/fl-cre). The gene for miR-23a tion that occurs by apoptosis. However, Mir27a-Mir24-2 cluster alters the commit-
was floxed in a manner that did not disrupt apoptotic activation-induced cell death ment of CD4+ T cells to become T helper

Immunity 44, March 15, 2016 ª2016 Elsevier Inc. 521


Immunity

Previews

2 (Th2) cells and affects airway allergic functions, which will need to be further Pasquinelli, A.E. (2012). Nat. Rev. Genet. 13,
271–282, http://dx.doi.org/10.1038/nrg3162nrg3162.
reactions (Cho et al., 2016; Pua et al., explored. It will be equally important to
2016). The effects were predominantly determine whether miR-23a, or the other
dependent upon miR-24 and miR-27, miRNAs in the cluster, has a similar role in Pua, H.H., Steiner, D.F., Patel, S., Gonzalez, J.R.,
Ortiz-Carpena, J.F., Kageyama, R., Chiou, N.-T.,
which were found to directly and indirectly humans and whether their dysregulation Gallman, A., de Kouchkovsky, D., Jeker, L.T.,
regulate IL-4 and Gata3, and no increase in contributes to pathology. et al. (2016). Immunity 44. Published
activated CD4+ T cell death was reported. online February 2, 2016. S1074-7613(16)00004-2.
http://dx.doi.org/10.1016/j.immuni.2016.01.003.
The results are not necessarily contradic- REFERENCES
tory with Zhang et al. (2016) because the
aforementioned studies focused on Th2 Baines, C.P., Kaiser, R.A., Purcell, N.H., Blair, N.S., Sena, L.A., and Chandel, N.S. (2012). Mol. Cell 48,
Osinska, H., Hambleton, M.A., Brunskill, E.W., 158–167, http://dx.doi.org/10.1016/j.molcel.2012.
cell responses and used OVA sensitization Sayen, M.R., Gottlieb, R.A., Dorn, G.W., et al. 09.025.
and did not involve a pathogen such as (2005). Nature 434, 658–662, http://dx.doi.org/10.
Listeria, which might induce higher ROS 1038/nature03434.
Simeoni, L., and Bogeski, I. (2015). Biol. Chem.
production in CD4+ T cells. There might Cho, S., Wu, C.-J., Yasuda, T., Cruz, L.O., 396, 555–568, http://dx.doi.org/10.1515/hsz-
also be competing effects of the miRNAs Khan, A.A., Lin, L.-L., Nguyen, D.T., Miller, M., 2014-0312.
that are lost when all three were deleted. Lee, H.-M., Kuo, M.-L., et al. (2016). J. Exp.
Med. 213, 235–249, http://dx.doi.org/10.1084/
The findings of these three new studies jem.20150990. Zhang, B., Liu, S.-Q., Li, C., Lykken, E., Jiang, S.,
highlight the breadth of control mediated Wong, E., Gong, Z., Tao, Z., Zhu, B., Wan, Y.,
Devadas, S., Zaritskaya, L., Rhee, S.G., Oberley, and Li, Q.-J. (2016). Immunity 44. Published online
by the miR-23a cluster on CD4+ T cell L., and Williams, M.S. (2002). J. Exp. Med. 195, February 23, 2016. S1074-7613(16)00031-5, this
biology and suggest context-dependent 59–70. issue.

Chitosan: An Adjuvant with an Unanticipated STING


Nicolas Riteau1 and Alan Sher1,*
1Immunobiology Section, Laboratory of Parasitic Diseases, National Institute for Allergy and Infectious Diseases, National Institutes of Health,

Bethesda, MD 20892, USA


*Correspondence: asher@niaid.nih.gov
http://dx.doi.org/10.1016/j.immuni.2016.03.002

Adjuvants promote adaptive immunity through the triggering of innate signals that are largely poorly under-
stood. In this issue of Immunity, Lavelle and colleagues describe an unexpected role for the DNA sensing
cGAS-STING pathway in the mechanism of action of the Th1 cell-promoting polysaccharide adjuvant chitosan.

Adjuvants are an important gateway to 1989). The discovery of pattern-recogni- tantly, they present intriguing evidence
better vaccines. A superb illustration of tion receptors (PRRs) and their corre- that the chitosan-induced trigger of this
this point is the experimental recombinant sponding microbial PAMPs has helped response is host mitochondrial DNA that
RTS,S vaccine used for vaccination define the molecular basis of innate im- is likely recognized as a danger-associ-
against malaria that became significantly mune recognition in adjuvant function ated molecular pattern (DAMP).
protective in humans only after formula- and has led to the development of new Chitosan is a polysaccharide derived
tion with the appropriate lipid-based adju- adjuvant products. However, the mecha- from the partial deacetylation of chitin,
vants (Hoffman et al., 2015). Nevertheless, nism of action of many natural adjuvants the second most abundant natural poly-
the science of adjuvant development re- remains poorly defined, a situation that mer. There are a vast variety of chitosan
mains a backwater of translational immu- might reflect the underappreciated contri- products that differ in their biochemical
nology because of our poor understanding bution of host-derived ‘‘danger signals.’’ and physical characteristics. The chitosan
of how to selectively trigger and boost In this issue of Immunity, Carroll et al. preparation utilized in this study was pre-
different classes of immune responses. (2016) dissect the pathway by which a viously shown by the authors to promote
It is generally thought that adjuvants natural adjuvant, chitosan, induces den- peritoneal Th1 and Th17 cell responses
work by linking innate and adaptive immu- dritic cell (DC) activation to promote T to ovalbumin. In the current study, they
nity and, indeed, Charles Janeway was helper 1 (Th1) cell immune responses. In demonstrate that the same polymer
clearly thinking of adjuvants when he so doing, they uncover an unanticipated triggers highly polarized Th1 cell cyto-
coined this mechanism as the ‘‘dirty little role for the cytosolic DNA sensing cGAS- kine and immunoglobulin G2c (IgG2c)
secret’’ of the immune system (Janeway, STING pathway in this process. Impor- antibody responses to a recombinant

522 Immunity 44, March 15, 2016 ª2016 Elsevier Inc.

You might also like