You are on page 1of 3

Immunity

Previews

of apoptosis that would promote cytokine contributing to inflammatory responses. REFERENCES


release by dying inflammasome-compe- Given the important role of the NLRP3
tent cells to promote tissue injury- inflammasome in various human patholo- Martinon, F., Mayor, A., and Tschopp, J. (2009).
Annu. Rev. Immunol. 27, 229–265.
mediated inflammation, or whether the gies, the answer to these questions will
release of oxidized mitochondrial DNA probably provide better overall under- Menu, P., and Vince, J.E. (2011). Clin. Exp. Immu-
nol. 166, 1–15.
and NLRP3 activation can occur in the standing of the mechanisms that translate
absence of apoptosis. Although the mito- cellular stress into an inflammatory Nakahira, K., Haspel, J.A., Rathinam, V.A.K., Lee,
S.-J., Dolinay, T., Lam, H.C., Englert, J.A., Rabino-
chondria is emerging as a key organelle reaction. In Choderlos de Laclos’s vitch, M., Cernadas, M., Kim, H.P., et al. (2011).
that senses cellular insults to promote novel Dangerous Liaisons, relationships Nat. Immunol. 12, 222–230.
inflammatory responses, the role of are used as weapons in a game of power
Shimada, K., Crother, T.R., Karlin, J., Dagvadorj,
potassium efflux and the mechanisms and revenge that eventually takes a tragic J., Chiba, N., Chen, S., Ramanujan, V.K., Wolf,
that drive mitochondrial ROS production course. By analogy, when mitochondrial A.J., Vergnes, L., Ojcius, D.M., et al. (2012). Immu-
nity 36, this issue, 401–414.
and control the release of mitochondrial DNA encounters and interacts with
DNA are still unknown. Finally, it remains NLRP3, it initiates a complex cascade of Tschopp, J. (2011). Eur. J. Immunol. 41, 1196–1202.
to be determined whether release of events that start with inflammasome
Zhang, Q., Raoof, M., Chen, Y., Sumi, Y., Sursal,
mitochondrial DNA into the circulation formation and promotes self-defenses T., Junger, W., Brohi, K., Itagaki, K., and Hauser,
upon tissue injury (Zhang et al., 2010) or, in a dramatic twist worthy of Laclos, C.J. (2010). Nature 464, 104–107.
could contribute to inflammasome activa- backfires into a dangerous and harmful Zhou, R., Yazdi, A.S., Menu, P., and Tschopp, J.
tion by recruited phagocytes, thereby inflammation. (2011). Nature 469, 221–225.

B Cells, Not Just for Antibody Anymore


Kamal M. Khanna1 and Leo Lefrançois1,*
1Department of Immunology, Center for Integrated Immunology and Vaccine Research, UCONN Health Center, Farmington, CT 06030, USA

*Correspondence: llefranc@neuron.uchc.edu
DOI 10.1016/j.immuni.2012.02.011

B cell antibody production is thought to be crucial for protection against virus infection. In this issue of
Immunity, Moseman et al. (2012) illustrate an antibody-independent role for B cells in macrophage activation
that prevents virus dissemination after subcutaneous infection.

The production of neutralizing antibodies infection. Thus, in the absence of type I been described (Delale et al., 2005). For
by B cells to protect against viral and interferon receptor signaling, mice are example, the IFN-b response after CMV
bacterial infections is a hallmark of immu- exquisitely sensitive to infection with a infection has been shown to be regulated
nity and one of the primary goals of vacci- number of viruses including vesicular by lymphotoxin (LT) produced by hemato-
nation. However, recent reports including stomatitis virus (VSV) and mouse cyto- poietic cells. In the absence of LTab-LTbR
a paper in this issue of Immunity have re- megalovirus (MCMV) (Garcı́a-Sastre and signaling, mice are highly susceptible to
vealed an additional previously unappre- Biron, 2006). While plasmacytoid den- intravenous MCMV infection, resulting in
ciated critical function of B cells as regula- dritic cells (pDCs) triggered through large part from poor induction of type I
tors of innate immunity to virus infection. pattern recognition receptors, particularly interferons (Benedict et al., 2001). Re-
For some viruses, the very earliest events TLRs, produce copious amounts of IFN-a markably, the relevant source of LTb
after infection that lead to virus clearance in response to virus infection, many other is the splenic B cell (Schneider et al.,
are not well defined. Whereas pre-existing cell types also have the ability to produce 2008). Bone marrow chimera recon-
‘‘natural’’ antibody may in some cases and respond to type I interferons. Never- stitution studies further revealed that the
provide some degree of initial protec- theless, the precise mechanisms by responding cell type is a stromal cell of
tion, the innate immune system including which IFN is induced, especially with re- nonhematopoietic origin. Thus, B cell-
macrophages, NK cells, and neutrophils gard to the initial cell types that encounter derived LTb acting on a potentially in-
are believed to be central to limiting initial and potentially harbor virus replication, fected stromal cell drives IFN-b produc-
pathogen spread while the adaptive im- are unclear. tion that leads to early protection against
mune response ramps up. Moreover, In addition to TLR-mediated IFN-a infection. Additionally, this pathway of
cytokines, in particular type I interferons, induction, TLR-independent pathways IFN production is independent of TLR
are key to early protection against virus for driving IFN-a production have also signaling. Considering that these events

Immunity 36, March 23, 2012 ª2012 Elsevier Inc. 315


Immunity

Previews

Now, in this issue of Immunity, Mose-


Presence of B cells Absence of B cells man et al. (2012) further dissect the role
VSV replication VSV replication of B cells, LT, type I interferons, and
+
CD169 macrophage (increased) +
CD169 macrophage (decreased) CD169+ macrophages in the draining LN
after subcutaneous VSV infection. First,
they show that B cells but not antibodies
are essential for protection against sub-
cutaneous VSV infection. Surprisingly,
although mice lacking immunoglobulin
LTα1β2 but harboring B cells die from intravenous
Type I IFN B cell Type I IFN (decreased) VSV infection, such mice are protected
from subcutaneous infection. In contrast,
Prevents VSV access VSV access mice lacking B cells succumbed to
to pheripheral nerves to pheripheral nerves and CNS subcutaneous VSV infection through virus
entry into the central nervous system via
peripheral nerves. Thus, infection via the
Figure 1. After Subcutaneous Infection with VSV, the Virus Is Initially Captured by CD169+ skin, perhaps mimicking virus transmis-
Macrophages in the Draining Lymph Node sion by an insect bite, reveals a clear
The release of LTa1b2 by B cells allows the CD169+ macrophages to tolerate increased replication of VSV, role for B cells in providing protection
possibly by inducing Usp18. This increased VSV replication induces the release of type I IFN by the
CD169+ macrophages, which in turn prevents the virus from accessing the peripheral nerves and traveling against a highly cytopathic virus, without
to the CNS by an undefined mechanism. In the absence of B cells there is a lack of LTa1b2 and without a requirement for antibody. Next, the
this, the virus fails to replicate in the CD169+ macrophages and type I IFN secretion is severely diminished. authors demonstrate that B cells but
The lack of type I IFN allows the virus to access the peripheral nerves, leading to dissemination of VSV to
the CNS.
again not antibody are required for
macrophage-dependent type I interferon
production. In this model, type I interferon
occur within minutes to a few hours after infection with VSV (Iannacone et al., is thought to confer protection via its
infection, these studies establish B cells 2010). These CD169+ macrophages are action on intranodal nerves to inhibit viral
as key regulators of early innate immunity the LN counterparts to the splenic replication. The precise mechanisms by
to CMV infection, independent of anti- CD169+ macrophages. Thus, this macro- which type I interferon affords this protec-
body production. phage subset represents a critical initial tion remain to be defined. Moreover, in the
In the case of vesicular stomatitis virus, barrier to viral and probably bacterial absence of B cells, LN macrophages did
a neurotropic member of the Rhabdovira- infection either through hematogenous not allow virus replication. Similar to
dae family that also includes rabies virus, delivery or through introduction into the studies in the spleen with CD169+ macro-
protection during primary and sec- skin. But do these cells directly destroy phages, previous work shows that VSV
ondary infections has long been ascribed virus or are cooperative efforts needed preferentially replicates in SCS macro-
to early neutralizing natural antibodies fol- from other innate and adaptive immune phages, rather than their medullary
lowed by induction of VSV-glycoprotein components? In the spleen, CD169+ counterparts. Indeed, in B cell-replete
(G)-specific IgM and subsequent class macrophages actually appear to promote but immunoglobulin-deficient mice, this
switching leading to IgG production. After viral replication (Honke et al., 2012). is precisely what is observed. Taken
intravenous infection, marginal zone met- Splenic CD169+ macrophages selectively together, the implication is that B cells,
allophilic macrophages (MMM) in the express the ubiquitin-specific protease but not antibody, are indispensable for
spleen capture the virus (Ciavarra et al., Usp18, which inhibits interferon ab re- promoting replication of VSV in SCS
2005). This initial line of cellular defense is ceptor (IFNAR) signaling. This inhibition macrophages leading to IFN production
vital to protection as indicated by the fact specifically in the CD169+ macrophages and protective immunity.
that depletion of splenic macrophages provides a more hospitable environment These intriguing data raise a number
results in virus dissemination and lethality for VSV replication. In IFNAR-deficient of important questions especially when
(Honke et al., 2012). B cell-derived LTab mice, all other splenic macrophage considered in light of earlier work indi-
is also required for splenic CD169+ macro- subsets allow VSV replication whereas in cating that splenic CD169+ macro-
phage organization (Mebius et al., 2004), normal mice, only the CD169+ macro- phages allow VSV replication through
further establishing an antibody-indepen- phages support virus growth. Moreover, mechanisms that inhibit IFNAR signaling
dent role for B cells in antiviral immunity. deletion of Usp18 results in lower splenic (Figure 1). Thus, although CD169+ macro-
More recently, the subcapsular sinus virus titers but rapid dissemination of the phages in both spleen and LN prefer-
(SCS) macrophages located in lymph virus to the central nervous system. entially allow VSV replication, whether
nodes (LN) have been shown to be essen- Honke et al. (2012) suggest that uncon- Usp18 mediates downmodulation of
tial for protection against subcutaneous trolled virus spread is caused by limited IFNAR signaling in LN SCS macrophages
VSV infection. Deletion of the SCS macro- neutralizing antibody production resulting needs to be examined to complete the
phages in the popliteal LN via injection of from poor initial virus replication in circuit. In addition, the possibility that
clodronate-loaded liposomes results in CD169+ macrophages, which is neces- LTa1b2 regulates Usp18 gene expression
loss of early protection against footpad sary to promote adaptive immunity. and thus virus replication in CD169+

316 Immunity 36, March 23, 2012 ª2012 Elsevier Inc.


Immunity

Previews

macrophages warrants analysis. Mose- prevent entry of pathogens upon sec- Akira, S., Vicari, A., et al. (2005). J. Immunol. 175,
6723–6732.
man et al. (2012) go on to show that B ondary encounter. In fact, recent findings
cell-derived LTa1b2 is critical for inducing indicate that a specialized subset of Edelson, B.T., Bradstreet, T.R., Hildner, K.,
the protective phenotype of the SCS dendritic cells are permissive for the initial Carrero, J.A., Frederick, K.E., Kc, W., Belizaire,
R., Aoshi, T., Schreiber, R.D., Miller, M.J., et al.
macrophages. When B cells are the only replication of Listeria monocytogenes (2011). Immunity 35, 236–248.
cell type that cannot produce LTa1b2, that leads to the induction of a rapid
Garcı́a-Sastre, A., and Biron, C.A. (2006). Science
macrophages are able to capture lymph- innate response leading to the generation
312, 879–882.
borne viral particles but the virus does of a protective T cell response (Edelson
not replicate and type I IFN secretion is et al., 2011). Additional research will be Honke, N., Shaabani, N., Cadeddu, G., Sorg, U.R.,
Zhang, D.E., Trilling, M., Klingel, K., Sauter, M.,
compromised. An outstanding question needed to determine whether events Kandolf, R., Gailus, N., et al. (2012). Nat. Immunol.
is what factors allow virus replication to such as these are more commonplace 13, 51–57.
proceed—can the effects all be attributed than previously appreciated.
Iannacone, M., Moseman, E.A., Tonti, E., Bosurgi,
to the inability of SCS macrophages to L., Junt, T., Henrickson, S.E., Whelan, S.P.,
respond to autocrine as well as perhaps Guidotti, L.G., and von Andrian, U.H. (2010).
REFERENCES Nature 465, 1079–1083.
paracrine type I interferon? The study by
Moseman et al. (2012) along with other Mebius, R.E., Nolte, M.A., and Kraal, G. (2004).
Benedict, C.A., Banks, T.A., Senderowicz, L., Ko, Crit. Rev. Immunol. 24, 449–464.
available data suggest the fascinating M., Britt, W.J., Angulo, A., Ghazal, P., and Ware,
possibility that the immune system has C.F. (2001). Immunity 15, 617–626. Moseman, E.A., Iannacone, M., Bosurgi, L., Tonti,
evolved to allow a certain proscribed E., Chevrier, N., Tumanov, A., Fu, Y.-X., Hacohen,
Ciavarra, R.P., Taylor, L., Greene, A.R., Yousefieh, N., and von Andrian, U.H. (2012). Immunity 36, this
degree of microbial growth to ‘‘prime the N., Horeth, D., van Rooijen, N., Steel, C., Gregory, issue, 415–426.
pump’’ in order to promote both rapid B., Birkenbach, M., and Sekellick, M. (2005).
innate immunity and provide sufficient Virology 342, 177–189. Schneider, K., Loewendorf, A., De Trez, C., Fulton,
J., Rhode, A., Shumway, H., Ha, S., Patterson, G.,
antigenic material and inflammatory Delale, T., Paquin, A., Asselin-Paturel, C., Dalod, Pfeffer, K., Nedospasov, S.A., et al. (2008). Cell
signals to promote adaptive immunity to M., Brizard, G., Bates, E.E., Kastner, P., Chan, S., Host Microbe 3, 67–76.

Natural ‘‘Helper’’ Cells in the Lung:


Good or Bad Help?
Shigeo Koyasu1,2,3,* and Kazuyo Moro1,2,4
1Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
2Laboratoryfor Immune Cell System, RIKEN Research Center for Allergy and Immunology (RCAI), Kanagawa 230-0045, Japan
3Research Center for Science Systems, Japan Society for the Promotion of Science (JSPS), Tokyo 102-8472, Japan
4Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo 102-0076,

Japan
*Correspondence: koyasu@z3.keio.jp
DOI 10.1016/j.immuni.2012.03.001

The natural helper (NH) cell comprises a newly identified Th2 cell-type innate lymphocyte population. In this
issue of Immunity, Halim et al. (2012) provide evidence that NH cells reside in the lung and play a critical role in
protease allergen-induced airway inflammation.

Proteases are important components of occurs via high protease activities and Th2 cell-type inflammation. Polymor-
many allergens (Reed and Kita, 2004). helminth infection induces rapid activa- phisms of IL-33 and its receptor ST2 are
For example, papain is a well-known tion of Th2 cell-type inflammatory re- associated with allergic diseases,
protease that causes occupational sponses. Proteases are thought to disrupt including asthma in humans, demon-
asthma, and native papain but not heat- mucosal integrity by digesting cell adhe- strating the importance of epithelial cell-
inactivated papain induces lung inflam- sion molecules and further act on derived cytokines for the onset of Th2
mation in mice. House dust mites also protease-activated receptors to activate cell-type immune responses.
produce a well-known allergen whose airway epithelial cells. The cytokines Papain causes asthma-like symptoms
allergy-inducing ability is largely depen- thymic stroma lymphopoietin (TSLP), associated with Th2 cell-type reactions
dent on its protease activity. In addition, IL-25, and IL-33 derived from epithelial such as eosinophilia and goblet cell
invasion of host tissues by helminthes cells activated in this way induce rapid hyperplasia in Rag-deficient mice. These

Immunity 36, March 23, 2012 ª2012 Elsevier Inc. 317

You might also like