Sunteți pe pagina 1din 12

Immunobiology 220 (2015) 215226

Contents lists available at ScienceDirect

Immunobiology
journal homepage: www.elsevier.com/locate/imbio

Review

Insights how monocytes and dendritic cells contribute and regulate


immune defense against microbial pathogens
Kristin Bieber, Stella E. Autenrieth
Department of Internal Medicine II, University of Tbingen, Germany

a r t i c l e i n f o a b s t r a c t

Article history: The immune system protects from infections primarily by detecting and eliminating invading pathogens.
Received 1 May 2014 Beside neutrophils, monocytes and dendritic cells (DCs) have been recently identied as important sen-
Received in revised form 17 October 2014 tinels and effectors in combating microbial pathogens. In the steady state mononuclear phagocytes like
Accepted 23 October 2014
monocytes and DCs patrol the blood and the tissues. Mammalian monocytes contribute to antimicro-
Available online 31 October 2014
bial defense by supplying tissues with macrophage and DC precursors. DCs recognize pathogens and are
essential in presenting antigens to initiate antigen-specic adaptive immune responses, thereby bridging
Keywords:
the innate and adaptive immune systems. Both, monocytes and DCs play distinct roles in the shaping of
Monocytes
Dendritic cells
immune response. In this review we will focus on the contributions of monocytes and lymphoid organ
Microbial infection DCs to immune defense against microbial pathogens in the mouse and their dynamic regulation from
Inammatory DCs steady state to infection.
Inammatory monocytes 2014 Elsevier GmbH. All rights reserved.
Tip-DCs
Innate immunity

Contents

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 216
Development of monocytes and DCs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 216
Classical DCs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 217
DCs as inducers of adaptive immunity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 218
DCs as tolerogenic cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 218
Murine monocyte subsets . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 218
Role of classical DCs during microbial infection in vivo . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 218
Bacterial transport/dissemination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 218
Priming of NK cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 219
Antimicrobial activity of DCs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 219
Priming of T-cell responses via maturation and cytokine secretion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 219
Evasion strategies of bacterial pathogens . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 220
Roles of CD115+ Ly6Chi inammatory monocytes/DCs and Tip-DCs during microbial infection in vivo . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 220
Listeria monocytogenes (Lm) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 221
Mobilization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 221

Corresponding author at: Department of Internal Medicine II, University of Tbingen, Otfried-Mller-Str. 10, D-72076 Tbingen, Germany. Tel.: +49 7071 2983156;
fax: +49 7071 294822.
E-mail address: Stella.Autenrieth@med.uni-tuebingen.de (S.E. Autenrieth).

http://dx.doi.org/10.1016/j.imbio.2014.10.025
0171-2985/ 2014 Elsevier GmbH. All rights reserved.
216 K. Bieber, S.E. Autenrieth / Immunobiology 220 (2015) 215226

Antimicrobial activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 221


Monopoiesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 221
Other infections . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 221
Infection induced myelopoiesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 222
Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 222
Conict of interests . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223

Introduction (Banchereau and Steinman, 1998). DCs present tissue-derived anti-


gens to CD8+ T cells (cross-presentation) and CD4+ T cells (direct
The mammalian immune system acts against common and presentation). This leads to the induction of peripheral tolerance in
rare microbial pathogens and prevents their invasion by consti- the steady state or to effector immunity by priming of CD4+ T cells
tutive innate immune barriers in mucosal and epithelial tissues. to either T helper 1 (Th1) cells, Th2, Th17 cells, or regulatory T cells
Highly virulent pathogens, which are generally less prevalent, have (Adams et al., 2005; Mellman and Steinman, 2001; Steinman et al.,
evolved mechanisms to circumvent constitutive immune defense 2002; Steinman and Banchereau, 2007).
mechanisms. Infection with these organisms leads to the induc- Monocytes are located at the site of antigen entry and of T-
tion of auxiliary innate defenses like the activation of the cellular cell priming. They are equipped with a large range of pattern
mediators neutrophils, macrophages, and dendritic cells (DCs) to recognition receptors such as toll-like receptors (TLRs), thereby
combat the pathogen. These phagocytes internalize and digest recognizing e.g., microorganisms and subsequently producing
bacteria, infected or dying cells and produce inammatory medi- inammatory molecules. One important function of monocytes
ators thereby killing pathogens and contributing to the activation is their contribution to the renewal of DCs and some tis-
of other cell types. Thus, while granulocytes have long been known sue macrophages predominantly under inammatory conditions
as important components of anti-microbial host defense, DCs and (Geissmann et al., 2003; Randolph and Furie, 1996; Serbina and
circulating monocytes are more and more implicated as essential Pamer, 2006; Varol et al., 2007). In addition to their functions in
players in defense against a range of microbial pathogens. innate immunity data have suggested that monocytes can con-
The developmental pathway of most cellular components of tribute to the polarization and expansion of lymphocytes (Auffray
the mammalian immune system including monocytes and DCs et al., 2007; Geissmann et al., 2003; Krutzik et al., 2005; Le Borgne
starts with pluripotent stem cells in the bone marrow. They et al., 2006; Len et al., 2007; Randolph et al., 1999; Serbina and
give rise to progenitors that follow a variety of differentiation Pamer, 2006; Serbina et al., 2003).
pathways to differentiate into mature cells with dened effec- Both, monocytes and DCs may play distinct roles in the shaping
tor functions. Mammalian monocytes contribute to antimicrobial of immune responses. In this review we will focus on the contrib-
defense by supplying tissues with macrophage and DC precursors utions of monocyte and lymphoid organ DCs to immune defense
(Gordon and Taylor, 2005). In terms of infection with a viru- against microbial pathogens in the mouse and their dynamic regu-
lent pathogen, however, the normal, homeostatic differentiation lation from steady state to infection.
pathway of monocytes is temporarily modied leading to the devel-
opment of a variety of effector cells with distinct antimicrobial Development of monocytes and DCs
activities from bone marrow and blood monocytes.
DCs are a heterogeneous population consisting of classical DCs Similar to all other leukocytes, monocytes and DCs originate
and plasmacytoid DCs (pDCs) with different abilities to respond to from multipotent self-renewing hematopoietic stem cells (HSCs)
external stimuli and activate distinct effector lymphocytes (Heath residing in the bone marrow (BM) (Liu and Nussenzweig, 2010).
and Carbone, 2009; Reizis et al., 2011). They recognize and ingest During differentiation these cells give rise to multipotential pro-
pathogens and are essential in presenting antigens to initiate anti- genitor cells (MPPs), loosing sequentially self-renewal capacity
gen specic adaptive immune responses, thereby bridging the whereas their differentiation capacity becomes more restricted.
innate and adaptive immune systems (Banchereau and Steinman, MPPs further differentiate into oligopotent progenitor cells such as
1998). the common myeloid progenitor cells (CMPs) (Akashi et al., 2000)
DCs in their immature state are sentinels of the immune system giving rise to monocytes, macrophages, granulocytes, megakary-
as they patrol in the periphery and continuously take up different ocytes, and erythrocytes; and the common lymphoid progenitors
kinds of self and foreign antigens, e.g., commensals, food antigens, (CLP) forming T, B and natural killer cells (Kondo et al., 1997).
or exogenous antigens and migrate in a CCR7-dependent man- The origin of DCs in this representation of myelopoiesis and
ner to the draining lymph nodes. Following uptake, antigens are lymhpopoiesis is controversially discussed. In vitro and in vivo
processed and loaded in form of peptides on major histocompati- differentiation assays showed that CLPs are more potent at pro-
bility complexes (MHC) (Mellman and Steinman, 2001). Activation ducing thymic DCs whereas CMPs are more efcient at generation
of DCs is induced, e.g., by microorganisms, infected cells as well as of splenic and lymph node cDCs (Izon et al., 2001; Liu and
by apoptotic bodies from dying cells. After stimulation immature Nussenzweig, 2010; Manz et al., 2001; Wu et al., 2001). However,
DCs transform into mature DCs, which comprise exceptional capac- as the steady state abundance of CMPs is 10-fold higher than that of
ity for T-cell stimulation. This activation-induced transformation is CLPs, most splenic DCs and a substantial fraction of thymic DCs orig-
accompanied by a dramatic cytoplasmic reorganization leading to inate from CMPs (K. Liu and Nussenzweig, 2010). Thus, DCs share
increased expression of surface MHC class II and co-stimulatory in part the developmental pathway with monocytes, macrophages
molecules. The role of DCs is absolutely critical for the induc- and neutrophils (Fig. 1). Further studies have identied a monocyte
tion and regulation of T-cell responses (Banchereau and Steinman, and DC progenitor (MDP) (Fogg, 2006; Varol et al., 2007) giving
1998). For example T-cell activation in a mixed lymphocyte reac- rise to either monocytes or to common DC progenitors (CDPs) (Liu
tion is normally carried out with equal numbers of stimulators and et al., 2009; Onai et al., 2007b) as committed DC progenitor in the
responders, but only one DC is necessary to turn on 1003000 T cells steady state. Very recently clonal studies on a single cell level argue
K. Bieber, S.E. Autenrieth / Immunobiology 220 (2015) 215226 217

Fig. 1. Schematic overview of the developmental pathway of DCs and monocytes in the steady state. DCs and monocytes develop from MPPs via CMP to MDP. MDPs give rise
to both monocytes and DCs via CDPs and pre-DCs. Preformed monocytes and pre-DCs migrate to the periphery where they differentiate into macrophages or DCs, respectively.
These processes are regulated by various growth factors, cytokines and transcription factors (gray). MPP: multipotential progenitor cells; CMP: common myeloid progenitor;
MDP: monocyte & DC progenitor; pre-DC: precursor of DCs, pDC: plasmacytoid DC, mono: monocyte, MF: macrophage.

against the MDP as a homogenous progenitor of both monocytes DCs (Auffray et al., 2007; Geissmann et al., 2003; Greter et al.,
and DCs (Sathe et al., 2014). Therefore, it is highly debated by the 2012; Krutzik et al., 2005; Le Borgne et al., 2006; Len et al., 2007;
community whether MDPs are an intermediate progenitor of DCs. Randolph et al., 1999; Serbina et al., 2003; Serbina and Pamer,
CDPs have lost their potential to differentiate into monocytes or 2006), but leaves the homeostasis of steady-state DCs unaltered.
macrophages and exclusively give rise to pDCs and to committed However, recent ndings using Csf2rb/ Csf2rb2/ mice argue
precursor of cDCs, the pre-cDCs (Gordon and Taylor, 2005; Liu et al., against an involvement of GM-CSF in the differentiation of inam-
2009; Naik et al., 2007; Onai et al., 2007a). While MDPs and CDPs matory DCs, instead this study revealed M-CSF al the master driver
are restricted to the BM, pre-cDCs migrate from the BM through for inammatory DC development in different infection models
the blood to peripheral lymphoid and non-lymphoid organs, where (Greter et al., 2012).
they further differentiate into DCs (Ginhoux et al., 2009; Heath Blood monocyte development is dependent on the growth factor
and Carbone, 2009; Liu et al., 2009; Reizis et al., 2011). Human M-CSF (Csf-1) and its receptor (Cecchini et al., 1994; Dai et al., 2002;
monocytes cultured with GM-CSF and IL-4 differentiate into DCs Liu and Nussenzweig, 2010), whereas GM-CSF and Flt3L appear to
in vitro (Banchereau and Steinman, 1998; Palucka, 1998; Sallusto be dispensable for monocyte development.
and Lanzavecchia, 1994). In humans, however, it has never been Furthermore, commitment and differentiation to DCs and
shown that monocytes can give rise to DCs in situ. monocytes of BM progenitors is tightly orchestrated by the expres-
This led to the hypothesis that monocytes are the immediate sion and balance of different transcription factors (Akashi et al.,
upstream precursors of DCs. However, further in vivo studies in 2000; Belz and Nutt, 2012; Miller et al., 2012). A number of
the mouse revealed that monocytes give mainly rise to DCs under transcription factors have been shown to be involved in the devel-
inammatory and infectious conditions leading to the adopted opment of DCs and DC subsets like PU.1, ID2, Zbtb46, BatF3,
concept of monocytes as precursors of inammatory DCs such interferon regulatory factors (IRFs) and STATs (see review (Kondo
as monocyte-derived DCs (mo-DCs) (Len et al., 2007; Mellman et al., 1997; Merad et al., 2013), whereas transcription factors like
and Steinman, 2001; Randolph et al., 1999; Varol et al., 2007) and PU.1, MafB, c-Maf and KLF-4 are required for monocyte differenti-
Tip-DCs (TNF--iNOS-producing DCs) (Banchereau and Steinman, ation (Auffray et al., 2009b; Izon et al., 2001; Liu and Nussenzweig,
1998; Naik et al., 2006; Serbina et al., 2003). It should be stressed 2010; Manz et al., 2001; Wu et al., 2001). By antagonistic interaction
that Tip-DCs neither constitute a separate DC lineage nor a DC with different transcription factors PU.1 controls several cell fate
subtype, but simply represent an activation status. Every type of decisions along the myeloid pathway. Thus, the relative expression
myeloid DC can be activated for the expression of inducible NO levels and the balance of transcription factors are important. For
synthase (iNOS) and TNF- by microbial and cytokine stimuli. example, the expression of PU.1 in MDPs is essential for DC differen-
DC differentiation in the steady state is mostly driven by FMS- tiation, whereas ectopically expressed MafB induces differentiation
related tyrosine kinase 3 ligand (Flt3L) with contribution of other into monocytes and macrophages thereby inhibiting DC commit-
growth factors like GM-CSF. Flt3L is required for the DC commit- ment (Bakri, 2005; Carotta et al., 2010; Liu and Nussenzweig, 2010).
ment of early hematopoietic progenitors in the BM (Banchereau
and Steinman, 1998; Mackarehtschian et al., 1995) and likewise Classical DCs
for the nal steps of homeostatic DC differentiation in periph-
eral organs in vivo (Adams et al., 2005; Mellman and Steinman, Classical DCs (here referred to as DCs) are the main DC subset
2001; Steinman, 2002; Steinman and Banchereau, 2007; Waskow in the lymphoid organs of mice and can be further differenti-
et al., 2008). GM-CSF (Csf-2) seemed to be involved in the develop- ated by their surface marker expression and location. As recently
ment of inammatory DCs (Geissmann et al., 2003; Randolph and reviewed by Merad et al. DCs are located in lymphoid tissues
Furie, 1996; Schmid et al., 2010; Serbina and Pamer, 2006; Varol where they are termed CD8+ DCs and phenotypically described as
et al., 2007) and the regulation of non-lymphoid tissue-resident CD11c+ CD11b CD8+ MHCII+ B220 or CD11b+ DCs, which express
218 K. Bieber, S.E. Autenrieth / Immunobiology 220 (2015) 215226

CD11c+ CD11b+ CD8 MHCII+ B220 on their cell surface (Fogg, subsets with a specialized Treg inducing function or by the action
2006; Merad et al., 2013; Varol et al., 2007; Vremec et al., 2000). In of the mentioned mediators present in local tissue microenviron-
non-lymphoid tissues DCs are divided by their distinct expression ment, which act on DCs and drive them to behave as tolerogenic DCs
of the surface markers CD11b and CD103 into CD103+ CD11b DCs, and induce Treg differentiation. The clonal deletion of self-reactive
CD103+ CD11b+ intestinal DCs and CD103 CD11b+ DCs (Liu et al., T cells is mediated by the presentation of tissue-derived antigens
2009; Merad et al., 2013; Onai et al., 2007b). Beside these tissue DCs, on MHC molecules expressed by predominantly immature DCs in
Langerhans Cells (LC), which are located in the epidermal layer of the steady state (Kushwah and Hu, 2011; Liu et al., 2002; Qiu et al.,
the skin and are characterized by the high level expression of the 2009).
C-type lectin Langerin (CD207) form another subset of DCs (Merad
et al., 2008; Sathe et al., 2014). Murine monocyte subsets

DCs as inducers of adaptive immunity Murine blood monocytes can be divided in two subsets. Both
subsets express CD115 (M-CSFR), CD11b, and low levels of F4/80.
In the steady state DCs are enriched at the borderlines of the One subset expresses in addition Ly6C, the chemokine receptor
body with the environment like the gut, lung, skin, the marginal CCR2, the adhesion molecule L-Selectin (CD62L) and the 7/4 anti-
zone in the spleen or the subcapsular sinus in the lymph nodes (LN) gen (Henderson et al., 2003; Taylor et al., 2003). This subset of
and are functionally specialized to sense pathogens (Helft et al., CD115+ Ly6Chi cells is named inammatory monocytes due to their
2010; Idoyaga et al., 2009). DCs express a specic set of pattern selective recruitment to inamed tissues and lymph nodes where
recognition receptors to sense bacteria, viruses, fungi, parasites as they differentiate into inammatory DCs (Geissmann et al., 2003;
well as dead cells and necrotic bodies (Cambi et al., 2003; Davey Palframan et al., 2001; Serbina et al., 2003; Sunderktter et al.,
et al., 2010; Edwards et al., 2003; Sancho et al., 2009; Yarovinsky, 2004).
2005). Activation of DCs drives immature cells to migrate in The second subset of monocytes is characterized by high expres-
response to chemokines into lymph nodes or the spleen where they sion of the chemokine receptor CX3CR1, LFA-1 and CD43 but low
develop into a mature phenotype. The maturation process is accom- expression of Ly6C and no CCR2 expression (Geissmann et al., 2003;
panied by the disappearance of the antigen-capturing apparatus Sunderktter et al., 2004). They are smaller in size, have a longer
and the up-regulation of essential co-stimulatory molecules such as half-life, and trafc into peripheral tissues under non-inammatory
CD80, CD86 and CD54. In addition maturation leads to an increased conditions (Geissmann et al., 2003). They were termed resident
display of MHC molecules with antigenic peptides on the cell sur- monocytes because they reconstitute tissue macrophages and DCs.
face (Banchereau and Steinman, 1998). Antigen presentation by More recent studies revealed that the CD115+ Ly6C monocytes are
DCs displays a critical process, as T cells will ignore unhandled anti- specialized in crawling in the blood vessels for extended periods of
gens. CD8+ DCs are critical for the induction of anti-viral CD8+ time thereby patrolling the endothelium (Auffray et al., 2007) for
T-cell responses in vivo due to their cross-presentation potential scavenging dead cells or potential pathogens. For this CD115+ Ly6C
as well as production of IL-12 and IL-15, two cytokines critical for monocytes were termed patrolling monocytes. Infection or tissue
cytotoxic CD8+ T-cell differentiation. In contrast, CD11b+ DCs are damage leads to the rapid extravasation of CD115+ Ly6C mono-
more efcient in MHC II-mediated CD4+ T-cell activation in the cytes and invasion of the inamed tissue, which is hardly observed
steady state and upon bacterial infections [reviewed by (Merad in the steady state (Auffray et al., 2007).
et al., 2013; Villadangos and Schnorrer, 2007)]. This is supported
by results showing that the preferential priming of CD8+ and CD4+
T cells by CD8+ and CD8 DCs, respectively, correlates with the Role of classical DCs during microbial infection in vivo
differential capacity of these DC subsets to process and present anti-
gens in association with MHC class I and MHC class II molecules As already mentioned DCs hallmark outstanding functions
(Dudziak et al., 2007). related to defense mechanisms against invading pathogens. There-
Furthermore, DCs are able to promote B-cell proliferation into fore pathogens have evolved many strategies to evade such host
plasma cells (Dubois et al., 1998, 1997) mediated via soluble fac- immune responses as provided by DCs. In this section we want
tors released by activated DCs or by direct cellcell interaction to dissect on the one hand the role of DCs during systemic infec-
(Obayashi et al., 2006; Wan et al., 2008). tions and on the other hand the evasion-strategies of the invading
pathogen. To elucidate the role of DCs during systemic infections
DCs as tolerogenic cells in more detail and in a more physiological way in vivo studies
in mouse experimental models in combination with DC depletion
Beside their important role during infections DCs are involved in using diphtheria toxin mouse models are a common approach.
the induction of tolerance. Central tolerance in the thymus is medi- Table 1 summarizes the ndings of DC depletion in various mouse
ated by negative selection of developing thymocytes or induction infection models discussed in the following section (Table 1).
of T regulatory cells (Tregs) [reviewed in (Hsieh et al., 2012; Klein
et al., 2009)]. Developing T cells that recognize self-peptides, which Bacterial transport/dissemination
are presented by thymic DCs or epithelial cells, with a sufciently
high afnity are either deleted through negative selection or dif- Recently, subpopulations of CD11c+ DCs have been implicated
ferentiate into Tregs. Injection of antigen-bearing DCs directly into at various stages of Listeria immunity, including the transport of
the developing thymus results in deletion of the reactive T cells blood-borne Listeria monocytogenes (Lm) to the spleen by CD8+
(Brocker et al., 1997). Similarly, CD8lo Sirp+ DCs in the thymus, DCs (Neuenhahn et al., 2006; Verschoor et al., 2011) and the early
which are phenotypically more mature than their Sirp coun- activation of CD8+ T cells (Jung et al., 2002). Lm rapidly associated
terparts, can induce antigen-specic Tregs (Proietto et al., 2008). with platelets in the bloodstream, which are specically taken up
Peripheral tolerance on the other hand is mediated by DCs through by splenic CD8+ DCs and thereby target Lm to these cells. Platelet
generation of Tregs and clonal deletion of self-reactive T cells. DC- tagging was also observed with other Gram-positive bacteria (S.
induced generation of Tregs is mainly mediated by IL-27, TGF-, aureus, E. fecalis, B. subtilis, S. pneumoniae (encapsulated and non-
IL-10, retinoic acid, indoleamine-2,3-dioxy-genase and vitamin D. capsulated)) suggesting a broadly active shuttling mechanism for
Treg generation is either mediated by tissue resident specic DC systemic bacteria (Verschoor et al., 2011). In the rst study of the
K. Bieber, S.E. Autenrieth / Immunobiology 220 (2015) 215226 219

Table 1
Summary of the outcome of DC depletion in infectious disease models.

Functional category of DCs Benecial effects of DC depletion Detrimental effects of DC depletion Refs.

Pathogen transport/dissemination Lm Neuenhahn


Lm, S. aureus, E. fecalis, B. subtilis, S. pneumoniae Verschoor
S. pyogenes Loof
NK cell priming Leishmania infantum Schleicher
Lm, vaccinia, LCMV Lucas
HSV-1 Kassim
Antimicrobial activity Y. enterocolitica Autenrieth
E. coli Tittel

T cell priming:
Cytokine production S. aureus Schindler
Th1 S. thyphimurium Salazar-Gonzales
M. tuberculosis Tian, Samstein
Th2 Schistosoma mansonii Phythian-Adams
CD8+ T cells Inuenza Belz
VSV Ciavarra
LCMV, MHV Probst, Birnberg
Lm Jung
Plasmodium yoelii Jung

group from Bush and co-workers DC-depletion seemed to be ben- model (Lucas et al., 2007), whereas in the visceral Leishmania model
ecial for Lm systemic infection due to reduced spreading of the TLR9 signaling by DCs and their subsequent production of IL-12 led
bacteria (Neuenhahn et al., 2006), but it was only clear after the to NK cell priming (Schleicher et al., 2007). These studies demon-
second study that targeting Lm and other bacteria to CD8+ DCs strate that depletion of DCs is detrimental for NK cells priming
by platelets results in the induction of anti-bacterial immunity and point out the importance of DC/NK cell interactions and the
(Verschoor et al., 2011) and is thereby benecial for the host. help of DCs to induce antimicrobial NK cell responses in secondary
In addition, DCs are shown to be important host cells for the dis- lymphoid organs.
semination of various pathogens. For example in a mouse model of
subcutaneous Streptococcus pyogenes infection ablation of DCs led Antimicrobial activity of DCs
to increased bacterial dissemination into draining lymph nodes and
systemic organs (Loof et al., 2007). Experiments with Salmonella Antimicrobial DC functions can be replaced or taken over by
dublin addressed the efcient internalization, survival and repli- other immune cells. Two independent studies showed, that the
cation within puried DCs. Similar to the experiments discussed depletion of DCs leads to the inux of phagocytes, namely neu-
above DCs are the main subset that harbor Salmonella (Marriott trophils and monocytes, to the spleen or the kidney (similar to
et al., 1999). Infection experiments with Mycobacterium bovis bacil- Kang et al. (Kang et al., 2008)). Infection of these mice with Yersinia
lus CalmetteGuerin (BCG) revealed DCs as a reservoir for bacteria enterocolitica results in an increased survival rate and signicantly
as BCG bacilli survive but do not proliferate during the rst two lower bacterial load due to the increased numbers of phagocytes in
weeks of an infection (Jiao et al., 2002). It is still not completely the spleen as well as their enhanced anti-bacterial killing capacity
understood how these bacteria avoid intracellular killing. dedicated to additional ROS (reactive oxygen species) production
(Autenrieth et al., 2012). Similar observations were made by Tit-
Priming of NK cells tel et al. showing that DC-depletion induced neutrophilia leading
to reduced bacterial load in the kidney in a pyelonephritis model
DCs are important cytokine producers in the early stages of (Tittel et al., 2012). In case of antimicrobial activity as rst line of
infections. In murine models of Lm, Staphylococcus aureus, and defense against invading pathogens DC depletion is benecial as it
Toxoplasma gondii infections DCs were the outstanding source of leads to the accumulation of more effective phagocytes at the site
IL-12 as an antimicrobial molecule (Liu et al., 2006; Schindler et al., of infection.
2012; Tam and Wick, 2006). IL-12 produced upon Listeria infec-
tion stimulates natural killer (NK)-cell activity, including IFN- Priming of T-cell responses via maturation and cytokine secretion
production, which in turn further activates macrophages, DCs and
neutrophils (Tripp et al., 1993). Activation of splenic DCs by Listeria Most bacterial infections induce the maturation of DCs, which
induced rapid clustering of innate cells, including granulocytes, NK is a prerequisite for T-cell activation (Autenrieth et al., 2010;
cells, and monocytes, to sites of bacterial localization (Kang et al., De Trez et al., 2005; Flohe, 2005; Sponaas, 2006; Sundquist and
2008). Herein, DC-depletion completely abrogated the infection- Wick, 2005; Tam and Wick, 2006, 2004). In Salmonella-infected
induced clustering of these incoming innate immune cells and the mice, maturation is induced directly in DCs associated with intra-
production of IFN- by NK cells, which in turn prevented the dif- cellular Salmonella and indirectly via Salmonella-induced TNF
ferentiation of Tip-DCs from monocytes. Thus, spatial organization production (Sundquist and Wick, 2005). Infection experiments
of the early immune response by DCs ensures functional activation with the extracellular Gram-negative bacterium Yersinia entero-
of innate cells. In line with these studies ablation of DCs revealed colitica highlighted that pathogens differentially affect subsets
that the in vivo priming of NK cell responses to TLR ligands, viruses of DCs. In contrast to Salmonella infection, Y. enterocolitica dif-
like vaccinia, LCMV, and HSV-1, the bacterial pathogen Lm and the ferently affect the maturation of splenic DC subpopulations and
parasite Leishmania infantum required the presence of classical DCs injection of pathogenicity factors into splenic DCs, predominantly
for optimal IFN- production (Hochweller et al., 2008; Kassim et al., into CD8+ DCs, diminished the up-regulation of MHC II and co-
2009; Lucas et al., 2007; Schleicher et al., 2007). IL-15 produced by stimulatory molecules. Indeed, only a small fraction of DCs is
DCs, that is induced by type I IFNs after TLR stimulation, was neces- infected in vivo. It will be interesting to identify the factors responsi-
sary and sufcient for the priming of NK cells in the Listeria infection ble for this cell tropism. These can be receptors especially expressed
220 K. Bieber, S.E. Autenrieth / Immunobiology 220 (2015) 215226

by CD8+ DCs, e.g., CD205, or the localization in the tissues. Y. with Y. enterocolitica the numbers of splenic CD4+ and CD8+ DCs
enterocolitica impaired the antigen uptake and degradation capac- are reduced partly due to TLR4-dependent induction of cell death
ity as well as cytokine production of CD8+ DCs but not CD11b+ (Autenrieth et al., 2010). Similarly, infection of mice with E. coli or
DCs (Autenrieth et al., 2010). Consequently, a reduced CD4+ T- treatment with LPS led to markedly reduced numbers of CD4+ and
cell proliferation was observed in Yersinia-infected mice, and a CD8+ DCs, a result of TLR4-TRIF signaling induced apoptosis (De
proper CD4+ T-cell response is necessary for the eradication of Smedt et al., 1996; De Trez et al., 2005). Furthermore, oral infection
the pathogen (Autenrieth et al., 1992, 2010), demonstrating that of mice with Salmonella typhimurium specically induces death of
microbial pathogens have successfully evolved strategies to com- CD8+ DCs, but not of CD8 DCs, via MyD88 and TNFR1 signaling
bat DC functions. Ex vivo studies revealed, that CD8+ DCs from (Sundquist and Wick, 2009). A signicant loss of DCs during a
infected mice were mostly impaired in their T-cell activation capac- systemic infection was also shown in a model of cecal ligation and
ity, suggesting that direct infection of DCs is necessary for effective puncture (CLP)-induced polymicrobial sepsis, which was depend-
modulation of DC functions (Autenrieth et al., 2010). Studies from ent on TLR2 and TLR4 signaling (Efron et al., 2004; Pne et al.,
a polymicrobial sepsis model (CLP) revealed that splenic DCs from 2009). Similar ndings were observed in the human system: in
septic mice released high levels of IL-10 compared to DCs from sepsis patients the number of splenic DCs is dramatically reduced,
control mice, whereas they were unable to secrete IL-12, even upon whereas the number of macrophages is not affected (Hotchkiss
stimulation with TLR ligands. These DC revealed a reduced capacity et al., 2002). These ndings resemble them from the conditional DC
for allogeneic T-cell activation that was associated with decreased depletion studies described above and it is tempting to speculate
IL-2 production (Flohe, 2005). It would be interesting to know as to that the more pronounced the reduction of DCs at the onset of an
whether DCs acquire a tolerogenic phenotype by IL-10 production infection is and the longer the DC numbers are reduced the worse
leading to the induction of Tregs in this model thereby modulating the course of infection will be. As a number of bacterial infections
T-cell activation. lead to reduced DC numbers the question arises whether the host
In contrast to these studies, fully functional DCs are rapidly has developed alternative strategies to overcome this evasion
recruited into infected tissue in a systemic infection model with mechanism.
S. aureus. DC-depletion revealed a benecial effect afforded by DCs
during S. aureus infection, which was not mediated by their con-
tribution to direct bacterial killing, nor by increased neutrophil Roles of CD115+ Ly6Chi inammatory monocytes/DCs and
recruitment, but rather by their IL-12 production (Schindler et al., Tip-DCs during microbial infection in vivo
2012), thereby most likely promoting T- and NK cell priming. In this
direction, ablation of DCs abolished IL-12 production in a Strepto- Describing the role of monocytes during microbial infections
coccus pyogenes model, which is required for effective control of we are going to focus on the CD115+ Ly6Chi inammatory mono-
infection (Loof et al., 2007). cyte subset. This is due to the fact that there is barely any
Using DC depletion it was shown that DCs have an outstand- report describing an important function of CD115+ Ly6C/low res-
ing ability to induce T-cell priming in various infection disease ident monocytes upon infection. At least Auffray et al. showed
models. As already mentioned, DCs are essential to induce an effec- that resident CD115+ Ly6Clow monocytes can be recruited to
tive long term CD8+ T cell mediated protective immunity upon sites of infection and thereby contribute to the initial inam-
infection with living but not killed Lm (Jung et al., 2002; Muraille matory response, e.g., by the release of TNF (Auffray et al.,
et al., 2005) and in various virus infection models (Belz et al., 2007).
2007; Birnberg et al., 2008; Ciavarra et al., 2005; Probst and van Upon infection monocytes are rapidly recruited to the site
den Broek, 2005). In addition to CD8+ T-cell response, DCs are of inammation (Geissmann et al., 2003; Randolph et al., 1999;
essential for CD4+ T-cell priming. For example, Salazar-Gonzales Sunderktter et al., 2004) and in such situations they can out-
and colleagues showed that a specic CCR6+ DC subset mediates number DCs and macrophages. Ly6Chi monocyte-derived cells have
pathogen-specic activation of CD4+ T cells in Peyers Patches upon been classied on the basis of a set of restricted but non-exclusive
oral infection with Salmonella typhimurium (Salazar-Gonzalez et al., functional properties as monocyte-derived DCs or inammatory
2006). Using a murine Mycobacterium tuberculosis model it was DCs, monocyte-derived macrophages or myeloid-derived suppres-
shown that even though monocyte-derived DCs deliver M. tuber- sor cells (MDSCs). However, it lacks suitable methods to exactly
culosis from the lung to lymph nodes (Samstein et al., 2013) and discriminate between different populations of these monocyte-
acquire a DC-like phenotype, classical DCs induce proliferation of derived cells. Due to this, it was recently suggested to term these
M. tuberculosis-specic CD4+ T cells (Samstein et al., 2013; Tian cells monocyte-derived cells or short MCs with distinct prop-
et al., 2005). DCs were also shown to be critical for the induction erties, e.g., iNOS+ MCs for Tip-DCs (Guilliams et al., 2014). In
and development of Th2 response in a mouse infection model with the following section we refer to the MCs and their function
the parasitic helminth Schistosoma mansoni (Phythian-Adams et al., upon microbial infection like there were described in the original
2010). papers.
All these studies clearly provide evidence for the importance Monocytes are able to take up antigens in peripheral tis-
of DCs in T-cell activation in various infection models and that DC sues and in the course of inammation, transport them to
depletion is detrimental for the outcome of infections. Although DC the draining lymph nodes. In addition, they produce cytokines
depletion is at least in the beginning of an infection benecial for and inammatory or antiinammatory mediators, and dif-
the innate anti-microbial response against bacterial pathogens by ferentiate into antigen-presenting cells (inammatory DCs)
modulating phagocyte homeostasis and activity, all the data on NK (Geissmann et al., 2003; Randolph et al., 1999). Inamma-
cell- and T-cell priming argue for an essential role of DCs for the tory DCs are dened by their expression of CD11c+ CD11b+ MHC
induction of adaptive immunity, which is shown to be necessary to II+ F4/80+ Ly6C+ CD206+ CD115+ CD64+ FcRI+ , their absence in the
overcome systemic infections. steady state, their migration from tissues to lymph nodes and their
ability to activate T cells (Segura and Amigorena, 2013). The best
Evasion strategies of bacterial pathogens markers used to discriminate macrophages from inammatory DCs
are CD64 and FcRI. Tip-DCs are a subset of inammatory DCs
One prominent immune evasion strategy of microbial initially identied in a Lm mouse infection model due to their pro-
pathogens is the induction of cell death of DCs. Following infection duction of TNF- and iNOS.
K. Bieber, S.E. Autenrieth / Immunobiology 220 (2015) 215226 221

Listeria monocytogenes (Lm) CX3CR1 is only weakly expressed by Ly6Chi monocytes deciency
of CX3CR1 impaired the recruitment of these Tip-DC precursors
Several studies have characterized the role of monocytes to the spleen during acute Lm infection and decreased the ef-
in vivo following microbial infection of mice with the intracellu- ciency of bacterial clearance (Auffray et al., 2009a). Recently, IL-23
lar bacterium Lm (Serbina et al., 2003; Serbina and Pamer, 2006; was shown to be required for the optimal recruitment of Tip-
Sunderktter et al., 2004). Lm infection leads to a massive CCR2- DCs. Lm-infected IL-23p19 KO mice revealed reduced levels of the
dependent egress of Ly6Chi monocytes from bone marrow to the monocyte-recruiting chemokines CCL2 and CCL7 and in conse-
bloodstream. The maximum recruitment of monocytes occurs 72 quence fewer monocytes in the spleen. Additionally, the overall
to 96 h following infection and thus is delayed relative to gran- concentrations of TNF- and nitric oxide as well as numbers of
ulocyte recruitment (North, 1970). At the site of infection Ly6Chi Tip-DCs were reduced in IL-23p19 KO mice leading to increased
monocytes differentiate in a MyD88-dependent manner into called bacterial burdens in the spleen upon Lm infection (Indramohan
Tip-DCs producing TNF- and iNOS combined with up-regulation et al., 2012). Thus, Ly6Chi monocytes/Tip-DCs are essential in medi-
of MHC II, CD80, CD86, and CD11c on the cell surface (Serbina et al., ating innate anti-microbial host defense upon Lm infection in vivo.
2003; Serbina and Pamer, 2006; Tsou et al., 2007). TLR-signaling
is required for protection during Lm infection, and mice decient
Monopoiesis
in the TLR adaptor molecule, MyD88, are highly susceptible to
infection due to diminished IL-12, IFN-, TNF- and NO responses
In addition to monocyte egress and differentiation of monocytes
(Edelson, 2002; Seki et al., 2002).
into Tip-DCs Lm infection specically and selectively promotes
monopoiesis accompanied by marked proliferation of bone mar-
Mobilization
row monocyte precursors and the depletion of myeloid progenitors
already 2 days post infection. The replenishment of monocytes
CCR2-decient mice have reduced numbers of Tip-DCs, which
depends on MyD88/Trif signaling whereas it is dispensable for early
is associated with impaired control and clearance of Lm following
emigration of the mature monocytes from the bone marrow. Thus,
infection (Serbina et al., 2003). Lm infection leads to the produc-
TLR-mediated signals are essential for the maintenance of mono-
tion of MCP-1 (CCL2) with detectable levels in spleen 6 h after
cyte homeostasis during systemic bacterial infection (Serbina et al.,
bacterial inoculation (Kurihara et al., 1997; Serbina et al., 2003),
2009).
Following Lm infection MCP-1 is produced in two phases. The
Infection of mice with very high bacterial dose has been used to
rst TLR/MyD88 signaling-dependent phase is rapid and induces
investigate the pathogenesis of Lm meningitis. In these settings, Lm
low-levels of MCP-1, whereas the second IFNR-dependent phase
is found predominantly in circulating Ly6C+ inammatory mono-
promotes prolonged and higher levels of MCP-1 secretion (Jia et al.,
cytes in the bloodstream and bone marrow (Drevets, 1999; Drevets
2009). Recently, at least one source of MCP-1 was identied. Low
et al., 2004; Join-Lambert et al., 2004; Sunderktter et al., 2004). Fol-
concentrations of TLR ligands in the bloodstream induce the pro-
lowing infection Lm-infected Ly6C+ monocytes are recruited into
duction of MCP-1 by bone marrow mesenchymal stem cells (MSCs)
the brain parenchyma independently of bacterial invasion of the
and their progeny, including CXC chemokine ligand (CXCL)12-
CNS. This recruitment is triggered by proinammatory cytokines,
abundant reticular (CAR) cells leading to the CCR2-dependent
in particular IFN-, produced by the innate immune response to
emigration of monocytes from the bone marrow (Shi et al., 2011). In
intracellular infection in peripheral organs (Drevets et al., 2010a,
addition to TLR- and IFNAR-signaling MPYS, a four-transmembrane
2008). These data indicate that despite the abundant presence of
protein essential for Lm induced type I IFN production in vitro
IFN- and TNF- BM monocytes developing during lethal infec-
(Ishikawa and Barber, 2008) was identied as player in the induc-
tion fail to acquire bactericidal activity. Further studies revealed
tion of monocyte emigration from the bone marrow following Lm
that BM monocytes phagocytose Lm only in lethal infection yet
infection. MPYS mediates type I IFN production by sensing cytosolic
they have increased antiinammatory signaling pathways, mak-
bacterial DNA (Ishikawa et al., 2009). Infection of MPYS decient
ing these cells unresponsive to proinammatory stimuli that could
mice with Lm lead to increased bacterial burden in the liver, which
trigger bactericidal activity (Drevets et al., 2010b).
correlated with the diminished production of MCP-1 and reduced
Ly6C+ monocyte frequencies in blood and liver (Jin et al., 2013).
Other infections
Antimicrobial activity
The importance of inammatory Ly6Chi monocytes/Tip-DCs
Tip-DCs were not directly infected with Lm and therefore are have also been reported in other infectious models in vivo and in
probably not involved in bacterial transport to the spleen (Serbina which they contribute to the control of pathogen growth during
et al., 2003). Mice with a genetic deciency in iNOS, gp91, or infections with bacteria, such as Mycobacterium tuberculosis (Peters
p47phox or mice treated with the iNOS inhibitor aminoguani- et al., 2001; Scott and Flynn, 2002), Brucella melitensis (Copin et al.,
dine are more susceptible to Lm infection (Beckerman et al., 1993; 2007, 2012), Burkholderia mallei (Goodyear et al., 2010), or with
Dinauer et al., 1997; Endres et al., 1997), implicating an inu- parasites, such as Plasmodium chabaudi (Sponaas et al., 2009), Toxo-
ence of these mechanisms in bacterial clearance. Tip-DCs are not plasma (Courret, 2006; Robben, 2005) and Leishmania major (De
essential for T-cell priming in vivo, although they are able to ef- Trez et al., 2009; Len et al., 2007).
ciently stimulate mixed lymphocyte reactions in vitro, as CD8 and In contrast to these benecial effects of inammatory Ly6Chi
CD4 T-cell responses to Lm are preserved in CCR2-decient mice monocytes/Tip-DCs there are also microbial infections in which
(Serbina et al., 2003). These ndings suggest that the main role these cells are suppressed or even pathological. During a model
of these cells is to combat the pathogen by killing rather by reg- of septic peritonitis neutrophils suppress peritoneal functions of
ulating T-cell responses although they may inuence the latter Tip-DCs through the production of IL-10 (Ocuin et al., 2011). More-
by their extraordinary production of the inammatory mediator over, Trypanosoma brucei infection led to the accumulation of
TNF-. Mice with a selective depletion of TNF- in monocytes Tip-DCs in the spleen, liver, and lymph nodes, which contribute
and macrophages are highly susceptible to Lm infection, suggest- actively to tissue damage during the chronic stage of T. brucei infec-
ing that these cell populations are the relevant source of TNF- in tion due to their sustained production of high amounts of TNF
defense against Lm infection (Grivennikov et al., 2005). Although and iNOS (Guilliams et al., 2009). This infection-induced Tip-DC
222 K. Bieber, S.E. Autenrieth / Immunobiology 220 (2015) 215226

Fig. 2. Modulation of myelopoiesis by infection. The inuence of the various infection agents on the developmental pathway of DCs and monocytes are summarized in here.
Infections induce TLR-dependent (i) DC depletion in the periphery, (ii) biased TLR- and IFN--dependent promotion of monopoiesis, and (iii) differentiation of monocytes to
Tip-DCs.

development is regulated by IL-10 and dependent on CCL2/CCR2, nonhematopoietic cell compartment in pathogen sensing, which
IFN- and MyD88 signaling (Bosschaerts et al., 2010; Guilliams is subsequently translated into cytokine release for enhanced,
et al., 2009). Similarly, in a systemic Candida infection model demand-adapted myeloid cell production (Boettcher et al., 2012).
type I interferons were shown to promote fatal immunopathol- Several studies demonstrate the requirement of innate immune
ogy by regulating inammatory monocytes and neutrophils sensing for myelopoiesis. TLR2-MyD88 signaling is essential for
(Majer et al., 2012). The high presence and activity of Tip-DC the production of myeloid cells in mice infected with Candida
s and neutrophils in the kidney during the early phase of Can- albicans (Ynez et al., 2011, 2009), whereas the maintenance of
dida infection promotes a secondary strong inux of neutrophils monocytes during Lm infection (Serbina et al., 2009) requires
culminating in lethal immunopathology. MyD88 signaling. Thus, the host responds to a wide variety of
Recently cells similar to the Tip-DCs were described that provide pathogens by the infection-induced modication of hematopoiesis
protection against a secondary Lm infection, suggesting that these via TLR- and MyD88-mediated signaling. Alternatively, induction
cells may be important for primary and secondary protective of monopoiesis may result from circulating bacterial products or
immunity (Narni-Mancinelli et al., 2007). from mediators produced by other cells in response to TLR activa-
In addition to the ndings that NO produced by Tip-DCs directly tion. Recently, it became obvious that IFN- mediated signals are
control their pathogen burden a study by Olekhnovitch et al. also essential for functional myelopoiesis upon infection with var-
argues for a collective nitric oxide production by numerous phago- ious pathogens, e.g., Ehrlichia, lymphocytic choriomeningitis virus
cytes providing tissue-wide immunity during Leishmania infection (LCMV), and Plasmodium chabaudi (Belyaev et al., 2013; de Bruin
proposing the model of a cooperative mechanism that generates et al., 2012; Goldszmid et al., 2012; MacNamara et al., 2011).
an antimicrobial milieu providing the basis for pathogen control at
the tissue level (Olekhnovitch et al., 2014).
Concluding remarks

Infection induced myelopoiesis The immune system protects from infections primarily by
detecting and eliminating the invading pathogens. Beside neu-
Several studies described the induction of monopoiesis by trophils, monocytes and DCs have been recently identied
microbial pathogens or pathogen derived molecules (Baldridge as important sentinels and effectors in combating microbial
et al., 2011; Belyaev et al., 2013; de Bruin et al., 2012; MacNamara pathogens. In the steady state mononuclear phagocytes like mono-
et al., 2011; Murray et al., 1998; Nagai et al., 2006; Serbina cytes and DCs patrol the blood and the tissues. During microbial
et al., 2009; Ynez et al., 2011) (Fig. 2). Pathogen induced alter- infections there is a need of monocytes and DCs with specic
ation of hematopoiesis may involve direct action on hematopoietic functions to elicit the appropriate immune response against the
stem and progenitor cells, as they express TLRs and myeloid cells invading pathogens. The variety of diverse subtypes of monocytes
could be generated from HSPCs by direct stimulation with LPS and DCs with different functions during the steady state and upon
via TLR4- and MyD88-dependent signaling in vitro (Nagai et al., infections is amazing and it possibly reects the variety of differ-
2006; Schmid et al., 2011). This is in contrast to ndings by ent microbial pathogens, different compartments and tissues, as
Boettcher et al. showing a key and nonredundant role for the well as different biological cell features like life span. The optimal
K. Bieber, S.E. Autenrieth / Immunobiology 220 (2015) 215226 223

immune response is determined by the balance between efcient cells is compatible with normal development and T cell homeosta-
sis, but causes myeloid proliferative syndrome. Immunity 29, 986997,
pathogen clearance and an acceptable level of immunopathology
http://dx.doi.org/10.1016/j.immuni.2008.10.012.
elicited by the immune response. Further insights into the com- Boettcher, S., Ziegler, P., Schmid, M.A., Takizawa, H., van Rooijen, N., Kopf, M.,
plex mutual regulation of microbial pathogens and the immune Heikenwalder, M., Manz, M.G., 2012. Cutting edge: LPS-induced emergency
system are needed. While the initiation of inammatory processes myelopoiesis depends on TLR4-expressing nonhematopoietic cells. J. Immunol.
188, 58245828, http://dx.doi.org/10.4049/jimmunol.1103253.
and myeloid cell development has been studied in detail in the Bosschaerts, T., Guilliams, M., Stijlemans, B., Morias, Y., Engel, D., Tacke, F.,
past years, the mechanisms that down-regulate these events are Hrin, M., De Baetselier, P., Beschin, A., 2010. Tip-DC development dur-
less well understood. ing parasitic infection is regulated by IL-10 and requires CCL2/CCR2,
IFN- and MyD88 signaling. PLoS Pathog. 6, e1001045, http://dx.doi.org/
10.1371/journal.ppat.1001045.t001.
Conict of interests Brocker, T., Riedinger, M., Karjalainen, K., 1997. Targeted expression of major his-
tocompatibility complex (MHC) class II molecules demonstrates that dendritic
cells can induce negative but not positive selection of thymocytes in vivo. J. Exp.
The authors declare no conicts of interests.
Med. 185, 541550.
Cambi, A., Gijzen, K., de Vries, I.J.M., Torensma, R., Joosten, B., Adema, G.J., Netea,
Acknowledgement M.G., Kullberg, B.-J., Romani, L., Figdor, C.G., 2003. The C-type lectin DC-SIGN
(CD209) is an antigen-uptake receptor for Candida albicans on dendritic cells.
Eur. J. Immunol. 33, 532538, http://dx.doi.org/10.1002/immu.200310029.
The authors would like to thank Ingo B. Autenrieth and Man- Carotta, S., Dakic, A., Amico, A.D., Pang, S.H.M., Greig, K.T., Nutt, S.L., Wu, L., 2010. The
ina Gnter for critical reading of the manuscript. The work was transcription factor PU.1 controls dendritic cell development and Flt3 cytokine
receptor expression in a dose-dependent manner. Immunity 32, 628641,
supported by the Deutsche Forschungsgemeinschaft (AU338/3-1,
http://dx.doi.org/10.1016/j.immuni.2010.05.005.
SFB 685 (TP-A3), SFB654 (TP-C7)) and the European Social Fund in Cecchini, M.G., Dominguez, M.G., Mocci, S., Wetterwald, A., Felix, R., Fleisch, H.,
Baden-Wrttemberg. Chisholm, O., Hofstetter, W., Pollard, J.W., Stanley, E.R., 1994. Role of colony
stimulating factor-1 in the establishment and regulation of tissue macrophages
during postnatal development of the mouse. Development 120, 13571372.
References Ciavarra, R.P., Taylor, L., Greene, A.R., Youseeh, N., Horeth, D., van Rooijen,
N., Steel, C., Gregory, B., Birkenbach, M., Sekellick, M., 2005. Impact of
Adams, S., ONeill, D.W., Bhardwaj, N., 2005. Recent advances in dendritic cell biol- macrophage and dendritic cell subset elimination on antiviral immunity,
ogy. J. Clin. Immunol. 25, 8798, http://dx.doi.org/10.1007/s10875-005-2814-2. viral clearance and production of type 1 interferon. Virology 342, 177189,
Akashi, K., Traver, D., Miyamoto, T., Weissman, I.L., 2000. A clonogenic common http://dx.doi.org/10.1016/j.virol.2005.07.031.
myeloid progenitor that gives rise to all myeloid lineages. Nature 404, 193197, Copin, R., De Baetselier, P., Carlier, Y., Letesson, J.-J., Muraille, E., 2007. MyD88-
http://dx.doi.org/10.1038/35004599. dependent activation of B220-CD11b + LY-6C+ dendritic cells during Brucella
Auffray, C., Fogg, D., Garfa, M., Elain, G., Join-Lambert, O., Kayal, S., Sarnacki, S., melitensis infection. J. Immunol. 178, 51825191.
Cumano, A., Lauvau, G., Geissmann, F., 2007. Monitoring of blood vessels and Copin, R., Vitry, M.-A., Hanot Mambres, D., Machelart, A., De Trez, C., Vanderwinden,
tissues by a population of monocytes with patrolling behavior. Science 317, J.-M., Magez, S., Akira, S., Ryffel, B., Carlier, Y., Letesson, J.-J., Muraille, E., 2012.
666670, http://dx.doi.org/10.1126/science.1142883. In situ microscopy analysis reveals local innate immune response developed
Auffray, C., Fogg, D.K., Narni-Mancinelli, E., Senechal, B., Trouillet, C., Saederup, N., around brucella infected cells in resistant and susceptible mice. PLoS Pathog. 8,
Leemput, J., Bigot, K., Campisi, L., Abitbol, M., Molina, T., Charo, I., Hume, D.A., e1002575, http://dx.doi.org/10.1371/journal.ppat.1002575.g009.
Cumano, A., Lauvau, G., Geissmann, F., 2009a. CX3CR1+CD115+CD135+ common Courret, N., 2006. CD11c- and CD11b-expressing mouse leukocytes transport
macrophage/DC precursors and the role of CX3CR1 in their response to inam- single Toxoplasma gondii tachyzoites to the brain. Blood 107, 309316,
mation. J. Exp. Med. 206, 595606, http://dx.doi.org/10.1084/jem.20081385. http://dx.doi.org/10.1182/blood-2005-02-0666.
Auffray, C., Sieweke, M.H., Geissmann, F., 2009b. Blood monocytes: development, Dai, X.-M., Ryan, G.R., Hapel, A.J., Dominguez, M.G., Russell, R.G., Kapp, S., Sylvestre,
heterogeneity, and relationship with dendritic cells. Annu. Rev. Immunol. 27, V., Stanley, E.R., 2002. Targeted disruption of the mouse colony-stimulating fac-
669692, http://dx.doi.org/10.1146/annurev.immunol.021908.132557. tor 1 receptor gene results in osteopetrosis, mononuclear phagocyte deciency,
Autenrieth, I.B., Tingle, A., Reske-Kunz, A., Heesemann, J., 1992. T lymphocytes medi- increased primitive progenitor cell frequencies, and reproductive defects. Blood
ate protection against Yersinia enterocolitica in mice: characterization of murine 99, 111120.
T-cell clones specic for Y. enterocolitica. Infect. Immun. 60, 11401149. Davey, G.M., Wojtasiak, M., Proietto, A.I., Carbone, F.R., Heath, W.R., Bedoui, S.,
Autenrieth, S.E., Linzer, T.-R., Hiller, C., Keller, B., Warnke, P., Kberle, M., Bohn, 2010. Cutting edge: priming of CD8 T cell immunity to herpes simplex virus
E., Biedermann, T., Bhring, H.-J., Hmmerling, G.J., Rammensee, H.-G., Aut- type 1 requires cognate TLR3 expression in vivo. J. Immunol. 184, 22432246,
enrieth, I.B., 2010. Immune evasion by Yersinia enterocolitica: differential http://dx.doi.org/10.4049/jimmunol.0903013.
targeting of dendritic cell subpopulations in vivo. PLoS Pathog. 6, e1001212, de Bruin, A.M., Libregts, S.F., Valkhof, M., Boon, L., Touw, I.P., Nolte, M.A., 2012. IFN
http://dx.doi.org/10.1371/journal.ppat.1001212.g008. induces monopoiesis and inhibits neutrophil development during inamma-
Autenrieth, S.E., Warnke, P., Wabnitz, G.H., Lucero Estrada, C., Pasquevich, tion. Blood 119, 15431554, http://dx.doi.org/10.1182/blood-2011-07-367706.
K.A., Drechsler, D., Gnter, M., Hochweller, K., Novakovic, A., Beer- De Smedt, T., Pajak, B., Muraille, E., Lespagnard, L., Heinen, E., De Baetselier, P., Urbain,
Hammer, S., Samstag, Y., Hmmerling, G.J., Garbi, N., Autenrieth, I.B., 2012. J., Leo, O., Moser, M., 1996. Regulation of dendritic cell numbers and maturation
Depletion of dendritic cells enhances innate anti-bacterial host defense by lipopolysaccharide in vivo. J. Exp. Med. 184, 14131424.
through modulation of phagocyte homeostasis. PLoS Pathog. 8, e1002552, De Trez, C., Magez, S., Akira, S., Ryffel, B., Carlier, Y., Muraille, E., 2009. iNOS-producing
http://dx.doi.org/10.1371/journal.ppat.1002552. inammatory dendritic cells constitute the major infected cell type during the
Bakri, Y., 2005. Balance of MafB and PU.1 species alternative macrophage chronic Leishmania major infection phase of C57BL/6 resistant mice. PLoS Pathog.
or dendritic cell fate. Blood 105, 27072716, http://dx.doi.org/10.1182/ 5, e1000494, http://dx.doi.org/10.1371/journal.ppat.1000494.s007.
blood-2004-04-1448. De Trez, C., Pajak, B., Brait, M., Glaichenhaus, N., Urbain, J., Moser, M., Lauvau,
Baldridge, M.T., King, K.Y., Goodell, M.A., 2011. Inammatory signals regu- G., Muraille, E., 2005. TLR4 and Toll-IL-1 receptor domain-containing adapter-
late hematopoietic stem cells. Trends Immunol. 32, 5765, http://dx.doi.org/ inducing IFN-beta, but not MyD88, regulate Escherichia coli-induced dendritic
10.1016/j.it.2010.12.003. cell maturation and apoptosis in vivo. J. Immunol. 175, 839846.
Banchereau, J., Steinman, R.M., 1998. Dendritic cells and the control of immunity. Dinauer, M.C., Deck, M.B., Unanue, E.R., 1997. Mice lacking reduced nicotinamide
Nature 392, 245252, http://dx.doi.org/10.1038/32588. adenine dinucleotide phosphate oxidase activity show increased susceptibility
Beckerman, K.P., Rogers, H.W., Corbett, J.A., Schreiber, R.D., McDaniel, M.L., Unanue, to early infection with Listeria monocytogenes. J. Immunol. 158, 55815583.
E.R., 1993. Release of nitric oxide during the T cell-independent pathway Drevets, D.A., 1999. Dissemination of Listeria monocytogenes by infected phagocytes.
of macrophage activation. Its role in resistance to Listeria monocytogenes. J. Infect. Immun. 67, 35123517.
Immunol. 150, 888895. Drevets, D.A., Dillon, M.J., Schawang, J.E., Stoner, J.A., Leenen, P.J.M., 2010a. IFN-
Belyaev, N.N., Bir, J., Langhorne, J., Potocnik, A.J., 2013. Extramedullary gamma triggers CCR2-independent monocyte entry into the brain during
myelopoiesis in malaria depends on mobilization of myeloid-restricted systemic infection by virulent Listeria monocytogenes. Brain Behav. Immun. 24,
progenitors by IFN- induced chemokines. PLoS Pathog. 9, e1003406, 919929, http://dx.doi.org/10.1016/j.bbi.2010.02.011.
http://dx.doi.org/10.1371/journal.ppat.1003406.s009. Drevets, D.A., Dillon, M.J., Schawang, J.S., van Rooijen, N., Ehrchen, J., Sunderktter, C.,
Belz, G.T., Nutt, S.L., 2012. Transcriptional programming of the dendritic cell net- Leenen, P.J.M., 2004. The Ly-6Chigh monocyte subpopulation transports Listeria
work. Nat. Rev. Immunol. 12, 101113, http://dx.doi.org/10.1038/nri3149. monocytogenes into the brain during systemic infection of mice. J. Immunol. 172,
Belz, G.T., Zhang, L., Lay, M.D.H., Kupresanin, F., Davenport, M.P., 2007. Killer 44184424.
T cells regulate antigen presentation for early expansion of memory, but Drevets, D.A., Schawang, J.E., Dillon, M.J., Lerner, M.R., Bronze, M.S., Brackett,
not naive, CD8+T cell. Proc. Natl. Acad. Sci. U. S. A. 104, 63416346, D.J., 2008. Innate responses to systemic infection by intracellular bacteria
http://dx.doi.org/10.1073/pnas.0609990104. trigger recruitment of Ly-6Chigh monocytes to the brain. J. Immunol. 181,
Birnberg, T., Bar-On, L., Sapoznikov, A., Caton, M.L., Cervantes-Barragn, L., 529536.
Makia, D., Krauthgamer, R., Brenner, O., Ludewig, B., Brockschnieder, D., Drevets, D.A., Schawang, J.E., Mandava, V.K., Dillon, M.J., Leenen, P.J.M., 2010b.
Riethmacher, D., Reizis, B., Jung, S., 2008. Lack of conventional dendritic Severe Listeria monocytogenes infection induces development of monocytes
224 K. Bieber, S.E. Autenrieth / Immunobiology 220 (2015) 215226

with distinct phenotypic and functional features. J. Immunol. 185, 24322441, Indramohan, M., Sieve, A.N., Break, T.J., Berg, R.E., 2012. Inammatory monocyte
http://dx.doi.org/10.4049/jimmunol.1000486. recruitment is regulated by interleukin-23 during systemic bacterial infection.
Dubois, B., Massacrier, C., Vanbervliet, B., Fayette, J., Briere, F., Banchereau, J., Caux, Infect. Immun. 80, 40994105, http://dx.doi.org/10.1128/IAI.00589-12.
C., 1998. Critical role of IL-12 in dendritic cell-induced differentiation of naive B Ishikawa, H., Barber, G.N., 2008. STING is an endoplasmic reticulum adap-
lymphocytes. J. Immunol. 161, 22232231. tor that facilitates innate immune signalling. Nature 455, 674678,
Dubois, B., Vanbervliet, B., Fayette, J., Massacrier, C., Van Kooten, C., Briere, F., http://dx.doi.org/10.1038/nature07317.
Banchereau, J., Caux, C., 1997. Dendritic cells enhance growth and differentiation Ishikawa, H., Ma, Z., Barber, G.N., 2009. STING regulates intracellular DNA-
of CD40-activated B lymphocytes. J. Immunol. 185, 941951. mediated, type I interferon-dependent innate immunity. Nature 461, 788792,
Dudziak, D., Kamphorst, A.O., Heidkamp, G.F., Buchholz, V.R., Trumpfheller, C., http://dx.doi.org/10.1038/nature08476.
Yamazaki, S., Cheong, C., Liu, K., Lee, H.W., Park, C.G., Steinman, R.M., Nussen- Izon, D., Rudd, K., DeMuth, W., Pear, W.S., Clendenin, C., Lindsley, R.C., Allman, D.,
zweig, M.C., 2007. Differential antigen processing by dendritic cell subsets in 2001. A common pathway for dendritic cell and early B cell development. J.
vivo. Science 315, 107111, http://dx.doi.org/10.1126/science.1136080. Immunol. 167, 13871392.
Edwards, A.D., Chaussabel, D., Tomlinson, S., Schulz, O., Sher, A., Reis e Sousa, C., 2003. Jia, T., Leiner, I., Dorothee, G., Brandl, K., Pamer, E.G., 2009. MyD88 and type
Relationships among murine CD11c(high) dendritic cell subsets as revealed by I interferon receptor-mediated chemokine induction and monocyte recruit-
baseline gene expression patterns. J. Immunol. 171, 4760. ment during Listeria monocytogenes infection. J. Immunol. 183, 12711278,
Efron, P.A., Martins, A., Minnich, D., Tinsley, K., Ungaro, R., Bahjat, F.R., Hotchkiss, R., http://dx.doi.org/10.4049/jimmunol.0900460.
Clare-Salzler, M., Moldawer, L.L., 2004. Characterization of the systemic loss of Jiao, X., Lo-Man, R., Guermonprez, P., Fiette, L., Deriaud, E., Burgaud, S., Gicquel, B.,
dendritic cells in murine lymph nodes during polymicrobial sepsis. J. Immunol. Winter, N., Leclerc, C., 2002. Dendritic cells are host cells for mycobacteria in vivo
173, 30353043. that trigger innate and acquired immunity. J. Immunol. 168, 12941301.
Endres, R., Luz, A., Schulze, H., Neubauer, H., Ftterer, A., Holland, S.M., Wagner, Jin, L., Getahun, A., Knowles, H.M., Mogan, J., Akerlund, L.J., Packard, T.A., Per-
H., Pfeffer, K., 1997. Listeriosis in p47(phox/ ) and TRp55/ mice: protection raud, A.-L., Cambier, J.C., 2013. STING/MPYS mediates host defense against
despite absence of ROI and susceptibility despite presence of RNI. Immunity 7, Listeria monocytogenes infection by regulating Ly6C(hi) monocyte migration. J.
419432. Immunol. 190, 28352843, http://dx.doi.org/10.4049/jimmunol.1201788.
Flohe, S.B., 2005. Dendritic cells during polymicrobial sepsis rapidly mature but fail Join-Lambert, O.F., Ezine, S., Le Monnier, A., Jaubert, F., Okabe, M., Berche, P., Kayal,
to initiate a protective Th1-type immune response. J. Leukoc. Biol. 79, 473481, S., 2004. Listeria monocytogenes-infected bone marrow myeloid cells promote
http://dx.doi.org/10.1189/jlb.0705413. bacterial invasion of the central nervous system. Cell. Microbiol. 7, 167180,
Fogg, D.K., 2006. A clonogenic bone marrow progenitor specic for macrophages http://dx.doi.org/10.1111/j.1462-5822.2004.00444.x.
and dendritic cells. Science 311, 8387, http://dx.doi.org/10.1126/science. Jung, S., Unutmaz, D., Wong, P., Sano, G.-I., De los Santos, K., Sparwasser, T., Wu,
1117729. S., Vuthoori, S., Ko, K., Zavala, F., Pamer, E.G., Littman, D.R., Lang, R.A., 2002.
Geissmann, F., Jung, S., Littman, D.R., 2003. Blood monocytes consist of two principal In vivo depletion of CD11c+ dendritic cells abrogates priming of CD8+ T cells by
subsets with distinct migratory properties. Immunity 19, 7182. exogenous cell-associated antigens. Immunity 17, 211220.
Ginhoux, F., Liu, K., Helft, J., Bogunovic, M., Greter, M., Hashimoto, D., Price, J., Yin, Kang, S.-J., Liang, H.-E., Reizis, B., Locksley, R.M., 2008. Regulation of hierarchical
N., Bromberg, J., Lira, S.A., Stanley, E.R., Nussenzweig, M., Merad, M., 2009. The clustering and activation of innate immune cells by dendritic cells. Immunity
origin and development of nonlymphoid tissue CD103+ DCs. J. Exp. Med. 206, 29, 819833, http://dx.doi.org/10.1016/j.immuni.2008.09.017.
31153130, http://dx.doi.org/10.1084/jem.20091756. Kassim, S.H., Rajasagi, N.K., Ritz, B.W., Pruett, S.B., Gardner, E.M., Chervenak, R., Jen-
Goldszmid, R.S., Caspar, P., Rivollier, A., White, S., Dzutsev, A., Hieny, S., nings, S.R., 2009. Dendritic cells are required for optimal activation of natural
Kelsall, B., Trinchieri, G., Sher, A., 2012. NK cell-derived interferon-g orches- killer functions following primary infection with herpes simplex virus type 1. J.
trates cellular dynamics and the differentiation of monocytes into dendritic Virol. 83, 31753186, http://dx.doi.org/10.1128/JVI.01907-08.
cells at the site of infection. Immunity 36, 10471059, http://dx.doi.org/ Klein, L., Hinterberger, M., Wirnsberger, G., Kyewski, B., 2009. Antigen presentation
10.1016/j.immuni.2012.03.026. in the thymus for positive selection and central tolerance induction. Nat. Rev.
Goodyear, A., Jones, A., Troyer, R., Bielefeldt-Ohmann, H., Dow, S., 2010. Critical pro- Immunol. 9, 833844, http://dx.doi.org/10.1038/nri2669.
tective role for MCP-1 in pneumonic Burkholderia mallei infection. J. Immunol. Kondo, M., Weissman, I.L., Akashi, K., 1997. Identication of clonogenic common
184, 14451454, http://dx.doi.org/10.4049/jimmunol.0900411. lymphoid progenitors in mouse bone marrow. Cell 91, 661672.
Gordon, S., Taylor, P.R., 2005. Monocyte and macrophage heterogeneity. Nat. Rev. Krutzik, S.R., Tan, B., Li, H., Ochoa, M.T., Liu, P.T., Sharfstein, S.E., Graeber, T.G., Sieling,
Immunol. 5, 953964, http://dx.doi.org/10.1038/nri1733. P.A., Liu, Y.-J., Rea, T.H., Bloom, B.R., Modlin, R.L., 2005. TLR activation triggers
Greter, M., Helft, J., Chow, A., Hashimoto, D., Mortha, A., Agudo-Cantero, J., Bogunovic, the rapid differentiation of monocytes into macrophages and dendritic cells.
M., Gautier, E.L., Miller, J., Leboeuf, M., Lu, G., Aloman, C., Brown, B.D., Pollard, Nat. Med. 11, 653660, http://dx.doi.org/10.1038/nm1246.
J.W., Xiong, H., Randolph, G.J., Chipuk, J.E., Frenette, P.S., Merad, M., 2012. GM- Kurihara, T., Warr, G., Loy, J., Bravo, R., 1997. Defects in macrophage recruitment
CSF controls nonlymphoid tissue dendritic cell homeostasis but is dispensable and host defense in mice lacking the CCR2 chemokine receptor. J Exp Med 186,
for the differentiation of inammatory dendritic cells. Immunity 36, 10311046, 17571762.
http://dx.doi.org/10.1016/j.immuni.2012.03.027. Kushwah, R., Hu, J., 2011. Role of dendritic cells in the induction of regulatory T cells.
Grivennikov, S.I., Tumanov, A.V., Liepinsh, D.J., Kruglov, A.A., Marakusha, B.I., Cell Biosci. 1, 20, http://dx.doi.org/10.1186/2045-3701-1-20.
Shakhov, A.N., Murakami, T., Drutskaya, L.N., Frster, I., Clausen, B.E., Tessarollo, Le Borgne, M., Etchart, N., Goubier, A., Lira, S.A., Sirard, J.C., van Rooijen,
L., Ryffel, B., Kuprash, D.V., Nedospasov, S.A., 2005. Distinct and nonredundant N., Caux, C., At-Yahia, S., Vicari, A., Kaiserlian, D., Dubois, B., 2006. Den-
in vivo functions of TNF produced by T cells and macrophages/neutrophils. dritic cells rapidly recruited into epithelial tissues via CCR6/CCL20 are
Immunity 22, 93104, http://dx.doi.org/10.1016/j.immuni.2004.11.016. responsible for CD8+ T cell crosspriming in vivo. Immunity 24, 191201,
Guilliams, M., Ginhoux, F., Jakubzick, C., Naik, S.H., Onai, N., Schraml, B.U., Segura, http://dx.doi.org/10.1016/j.immuni.2006.01.005.
E., Tussiwand, R., Yona, S., 2014. Perspectives. Nat. Rev. Immunol. 14, 571578, Len, B., Lpez-Bravo, M., Ardavn, C., 2007. Monocyte-derived dendritic cells
http://dx.doi.org/10.1038/nri3712. formed at the infection site control the induction of protective T helper
Guilliams, M., Movahedi, K., Bosschaerts, T., VandenDriessche, T., Chuah, M.K., 1 responses against Leishmania. Immunity 26, 519531, http://dx.doi.org/
Hrin, M., Acosta-Sanchez, A., Ma, L., Moser, M., Van Ginderachter, J.A., Brys, 10.1016/j.immuni.2007.01.017.
L., De Baetselier, P., Beschin, A., 2009. IL-10 dampens TNF/inducible nitric Liu, C.H., Fan, Y., Dias, A., Esper, L., Corn, R.A., 2006. Cutting edge: dendritic cells
oxide synthase-producing dendritic cell-mediated pathogenicity during para- are essential for in vivo IL-12 production and development of resistance against
sitic infection. J. Immunol. 182, 11071118. Toxoplasma gondii infection in mice. J. Immunol. 177, 3135.
Heath, W.R., Carbone, F.R., 2009. Dendritic cell subsets in primary and sec- Liu, K., Iyoda, T., Saternus, M., Kimura, Y., Inaba, K., Steinman, R.M., 2002. Immune
ondary T cell responses at body surfaces. Nat. Immunol. 10, 12371244, tolerance after delivery of dying cells to dendritic cells in situ. J. Exp. Med. 196,
http://dx.doi.org/10.1038/ni.1822. 10911097.
Helft, J., Ginhoux, F., Bogunovic, M., Merad, M., 2010. Origin and functional het- Liu, K., Nussenzweig, M.C., 2010. Development and homeostasis of dendritic cells.
erogeneity of non-lymphoid tissue dendritic cells in mice. Immunol. Rev. 234, Eur. J. Immunol. 40, 20992102, http://dx.doi.org/10.1002/eji.201040501.
5575, http://dx.doi.org/10.1111/j.0105-2896.2009.00885.x. Liu, K., Victora, G.D., Schwickert, T.A., Guermonprez, P., Meredith, M.M.,
Henderson, R.B., Hobbs, J.A.R., Mathies, M., Hogg, N., 2003. Rapid recruitment of Yao, K., Chu, F.F., Randolph, G.J., Rudensky, A.Y., Nussenzweig, M., 2009.
inammatory monocytes is independent of neutrophil migration. Blood 102, In vivo analysis of dendritic cell development and homeostasis. Science,
328335, http://dx.doi.org/10.1182/blood-2002-10-3228. http://dx.doi.org/10.1126/science.1170540.
Hochweller, K., Striegler, J., Hmmerling, G.J., Garbi, N., 2008. A novel CD11c.DTR Loof, T.G., Rohde, M., Chhatwal, G.S., Jung, S., Medina, E., 2007. The contribution of
transgenic mouse for depletion of dendritic cells reveals their requirement for dendritic cells to host defenses against Streptococcus pyogenes. J. Infect. Dis. 196,
homeostatic proliferation of natural killer cells. Eur. J. Immunol. 38, 27762783, 17941803, http://dx.doi.org/10.1086/523647.
http://dx.doi.org/10.1002/eji.200838659. Lucas, M., Schachterle, W., Oberle, K., Aichele, P., Diefenbach, A., 2007. Dendritic
Hotchkiss, R.S., Tinsley, K.W., Swanson, P.E., Grayson, M.H., Osborne, D.F., Wagner, cells prime natural killer cells by trans-presenting interleukin 15. Immunity 26,
T.H., Cobb, J.P., Coopersmith, C., Karl, I.E., 2002. Depletion of dendritic cells, but 503517, http://dx.doi.org/10.1016/j.immuni.2007.03.006.
not macrophages, in patients with sepsis. J. Immunol. 168, 24932500. Mackarehtschian, K., Hardin, J.D., Moore, K.A., Boast, S., Goff, S.P., Lemischka, I.R.,
Hsieh, C.-S., Lee, H.-M., Lio, C.-W.J., 2012. Selection of regulatory T cells in the thymus. 1995. Targeted disruption of the k2/t3 gene leads to deciencies in primitive
Nat. Rev. Immunol. 12, 157167, http://dx.doi.org/10.1038/nri3155. hematopoietic progenitors. Immunity 3, 147161.
Idoyaga, J., Suda, N., Suda, K., Park, C.G., Steinman, R.M., 2009. Antibody to lan- MacNamara, K.C., Oduro, K., Martin, O., Jones, D.D., McLaughlin, M., Choi, K.,
gerin/CD207 localizes large numbers of CD8+ dendritic cells to the marginal zone Borjesson, D.L., Winslow, G.M., 2011. Infection-induced myelopoiesis during
of mouse spleen. Proc. Natl. Acad. Sci. U. S. A. 106, 15241529, http://dx.doi.org/ intracellular bacterial infection is critically dependent upon IFN-signaling. J.
10.1073/pnas.0812247106. Immunol. 186, 10321043, http://dx.doi.org/10.4049/jimmunol.1001893.
K. Bieber, S.E. Autenrieth / Immunobiology 220 (2015) 215226 225

Majer, O., Bourgeois, C., Zwolanek, F., Lassnig, C., Kerjaschki, D., Mack, M., Mller, M., 2010. CD11c depletion severely disrupts Th2 induction and development in vivo.
Kuchler, K., 2012. Type I interferons promote fatal immunopathology by regu- J. Exp. Med. 207, 20892096, http://dx.doi.org/10.1084/jem.20100734.
lating inammatory monocytes and neutrophils during Candida infections. PLoS Probst, H.-C., van den Broek, M., 2005. Priming of CTLs by lymphocytic choriomenin-
Pathog. 8, e1002811, http://dx.doi.org/10.1371/journal.ppat.1002811.s005. gitis virus depends on dendritic cells. J. Immunol. 174, 39203924.
Manz, M.G., Traver, D., Miyamoto, T., Weissman, I.L., Akashi, K., 2001. Den- Proietto, A.I., van Dommelen, S., Zhou, P., Rizzitelli, A., DAmico, A., Steptoe, R.J., Naik,
dritic cell potentials of early lymphoid and myeloid progenitors. Blood 97, S.H., Lahoud, M.H., Liu, Y., Zheng, P., Shortman, K., Wu, L., 2008. Dendritic cells
33333341. in the thymus contribute to T-regulatory cell induction. Proc. Natl. Acad. Sci. U.
Marriott, I., Hammond, T.G., Thomas, E.K., et al., 1999. Salmonella efciently enter and S. A. 105, 1986919874, http://dx.doi.org/10.1073/pnas.0810268105.
survive within cultured CD11c+ dendritic cells initiating cytokine expression. Qiu, C.H., Miyake, Y., Kaise, H., Kitamura, H., Ohara, O., Tanaka, M., 2009. Novel
Eur. J. Immunol. 29, 11071125. Subset of CD8+ dendritic cells localized in the marginal zone is responsi-
Mellman, I., Steinman, R.M., 2001. Dendritic cells: specialized and regulated antigen ble for tolerance to cell-associated antigens. J. Immunol. 182, 41274136,
processing machines. Cell 106, 255258. http://dx.doi.org/10.4049/jimmunol.0803364.
Merad, M., Ginhoux, F., Collin, M., 2008. Origin, homeostasis and function of Langer- Randolph, G.J., Furie, M.B., 1996. Mononuclear phagocytes egress from an in vitro
hans cells and other Langerin-expressing dendritic cells. Nat. Rev. Immunol. 8, model of the vascular wall by migrating across endothelium in the basal to
935947, http://dx.doi.org/10.1038/nri2455. apical direction: role of intercellular adhesion molecule 1 and the CD11/CD18
Merad, M., Sathe, P., Helft, J., Miller, J., Mortha, A., 2013. The dendritic cell integrins. J. Exp. Med. 183, 451462.
lineage: ontogeny and function of dendritic cells and their subsets in the Randolph, G.J., Inaba, K., Robbiani, D.F., Steinman, R.M., Muller, W.A., 1999. Dif-
steady state and the inamed setting. Annu. Rev. Immunol. 31, 563604, ferentiation of phagocytic monocytes into lymph node dendritic cells in vivo.
http://dx.doi.org/10.1146/annurev-immunol-020711-074950. Immunity 11, 753761.
Miller, J.C., Brown, B.D., Shay, T., Gautier, E.L., Jojic, V., Cohain, A., Pandey, G., Leboeuf, Reizis, B., Bunin, A., Ghosh, H.S., Lewis, K.L., Sisirak, V., 2011. Plasmacytoid dendritic
M., Elpek, K.G., Helft, J., Hashimoto, D., Chow, A., Price, J., Greter, M., Bogunovic, cells: recent progress and open questions. Annu. Rev. Immunol. 29, 163183,
M., Bellemare-Pelletier, A., Frenette, P.S., Randolph, G.J., Turley, S.J., Merad, M., http://dx.doi.org/10.1146/annurev-immunol-031210-101345.
2012. Deciphering the transcriptional network of the dendritic cell lineage. Nat. Robben, P.M., 2005. Recruitment of Gr-1+ monocytes is essential for con-
Immunol. 13, 114, http://dx.doi.org/10.1038/ni.2370. trol of acute toxoplasmosis. J. Exp. Med. 201, 17611769, http://dx.doi.org/
Muraille, E., Giannino, R., Guirnalda, P., Leiner, I., Jung, S., Pamer, E.G., 10.1084/jem.20050054.
Lauvau, G., 2005. Distinct in vivo dendritic cell activation by live Salazar-Gonzalez, R.M., Niess, J.H., Zammit, D.J., Ravindran, R., Srinivasan, A.,
versus killed Listeria monocytogenes. Eur. J. Immunol. 35, 14631471, Maxwell, J.R., Stoklasek, T., Yadav, R., Williams, I.R., Gu, X., 2006. CCR6 mediated
http://dx.doi.org/10.1002/eji.200526024. dendritic cell activation of pathogen-specic T cells in Peyers Patches. Immunity
Murray, P.J., Young, R.A., Daley, G.Q., 1998. Hematopoietic remodeling in interferon- 24, 623632, http://dx.doi.org/10.1016/j.immuni.2006.02.015.
decient mice infected with mycobacteria. Blood 91, 112. Sallusto, F., Lanzavecchia, A., 1994. Efcient presentation of soluble antigen by
Nagai, Y., Garrett, K.P., Ohta, S., Bahrun, U., Kouro, T., Akira, S., Takatsu, K., cultured human dendritic cells is maintained by granulocyte/macrophage
Kincade, P.W., 2006. Toll-like receptors on hematopoietic progenitor cells colony-stimulating factor plus interleukin 4 and downregulated by tumor necro-
stimulate innate immune system replenishment. Immunity 24, 801812, sis factor alpha. J. Exp. Med. 179, 11091118.
http://dx.doi.org/10.1016/j.immuni.2006.04.008. Samstein, M., Schreiber, H.A., Leiner, I.M., Susac, B., Glickman, M.S., Pamer,
Naik, S.H., Metcalf, D., van Nieuwenhuijze, A., Wicks, I., Wu, L., OKeeffe, M., Short- E.G., 2013. Essential yet limited role for CCR2+ inammatory monocytes
man, K., 2006. Intrasplenic steady-state dendritic cell precursors that are distinct during Mycobacterium tuberculosis-specic T cell priming. eLife 2, e01086,
from monocytes. Nat. Immunol. 7, 663671, http://dx.doi.org/10.1038/ni1340. http://dx.doi.org/10.7554/eLife.01086.
Naik, S.H., Sathe, P., Park, H.-Y., Metcalf, D., Proietto, A.I., Dakic, A., Carotta, S., Sancho, D., Joffre, O.P., Keller, A.M., Rogers, N.C., Martnez, D., Hernanz-Falcn,
OKeeffe, M., Bahlo, M., Papenfuss, A., Kwak, J.-Y., Wu, L., Shortman, K., 2007. P., Rosewell, I., Sousa, C.R.E., 2009. Identication of a dendritic cell recep-
Development of plasmacytoid and conventional dendritic cell subtypes from tor that couples sensing of necrosis to immunity. Nature 458, 899903,
single precursor cells derived in vitro and in vivo. Nat. Immunol. 8, 12171226, http://dx.doi.org/10.1038/nature07750.
http://dx.doi.org/10.1038/ni1522. Sathe, P., Metcalf, D., Vremec, D., Naik, S.H., Langdon, W.Y., Huntington,
Narni-Mancinelli, E., Campisi, L., Bassand, D., Cazareth, J., Gounon, P., Glaichen- N.D., Wu, L., Shortman, K., 2014. Lymphoid tissue and plasmacytoid
haus, N., Lauvau, G., 2007. Memory CD8+ T cells mediate antibacterial immunity dendritic cells and macrophages do not share a common macrophage-
via CCL3 activation of TNF/ROI+ phagocytes. J. Exp. Med. 204, 20752087, dendritic cell-restricted progenitor. Immunity 41, 104115, http://dx.doi.org/
http://dx.doi.org/10.1084/jem.20070204. 10.1016/j.immuni.2014.05.020.
Neuenhahn, M., Kerksiek, K.M., Nauerth, M., Suhre, M.H., Schiemann, M., Gebhardt, Schindler, D., Gutierrez, M.G., Beineke, A., Rauter, Y., Rohde, M., Foster, S., Gold-
F.E., Stemberger, C., Panthel, K., Schrder, S., Chakraborty, T., Jung, S., Hochrein, mann, O., Medina, E., 2012. Dendritic cells are central coordinators of the host
H., Rssmann, H., Brocker, T., Busch, D.H., 2006. CD8+ dendritic cells are immune response to Staphylococcus aureus bloodstream infection. Am. J. Pathol.
required for efcient entry of Listeria monocytogenes into the Spleen. Immunity 181, 13271337, http://dx.doi.org/10.1016/j.ajpath.2012.06.039.
25, 619630, http://dx.doi.org/10.1016/j.immuni.2006.07.017. Schleicher, U., Liese, J., Knippertz, I., Kurzmann, C., Hesse, A., Heit, A., Fischer,
North, R.J., 1970. The relative importance of blood monocytes and xed macrophages J.A.A., Weiss, S., Kalinke, U., Kunz, S., Bogdan, C., 2007. NK cell acti-
to the expression of cell-mediated immunity to infection. J. Exp. Med. 132, vation in visceral leishmaniasis requires TLR9, myeloid DCs, and IL-12,
521534. but is independent of plasmacytoid DCs. J. Exp. Med. 204, 893906,
Obayashi, K., Doi, T., Koyasu, S., 2006. Dendritic cells suppress IgE production in B http://dx.doi.org/10.1084/jem.20061293.
cells. Int. Immunol. 19, 217226, http://dx.doi.org/10.1093/intimm/dxl138. Schmid, M.A., Kingston, D., Boddupalli, S., Manz, M.G., 2010. Instructive cytokine
Ocuin, L.M., Bamboat, Z.M., et al., 2011. Neutrophil IL-10 suppresses periton- signals in dendritic cell lineage commitment. Immunol. Rev. 234, 3244,
eal inammatory monocytes during polymicrobial sepsis. J. Immunol. 83, http://dx.doi.org/10.1111/j.0105-2896.2009.00877.x.
423432. Schmid, M.A., Takizawa, H., Baumjohann, D.R., Saito, Y., Manz, M.G., 2011. Bone
Olekhnovitch, R., Ryffel, B., Mller, A.J., Bousso, P., 2014. Collective nitric oxide marrow dendritic cell progenitors sense pathogens via Toll-like receptors
production provides tissue-wide immunity during Leishmania infection. J. Clin. and subsequently migrate to inamed lymph nodes. Blood 118, 48294840,
Invest. 124, 17111722, http://dx.doi.org/10.1172/JCI72058DS1. http://dx.doi.org/10.1182/blood-2011-03-344960.
Onai, N., Obata-Onai, A., Schmid, M.A., Manz, M.G., 2007a. Flt3 in regulation of type Scott, H.M., Flynn, J.L., 2002. Mycobacterium tuberculosis in chemokine receptor 2-
I interferon-producing cell and dendritic cell development. Ann. N.Y. Acad. Sci. decient mice: inuence of dose on disease progression. Infect. Immun. 70,
1106, 253261, http://dx.doi.org/10.1196/annals.1392.015. 59465954.
Onai, N., Obata-Onai, A., Schmid, M.A., Ohteki, T., Jarrossay, D., Manz, M.G., 2007b. Segura, E., Amigorena, S., 2013. Inammatory dendritic cells in mice and humans.
Identication of clonogenic common Flt3 + M-CSFR+ plasmacytoid and con- Trends Immunol. 34, 16, http://dx.doi.org/10.1016/j.it.2013.06.001.
ventional dendritic cell progenitors in mouse bone marrow. Nat. Immunol. 8, Serbina, N.V., Hohl, T.M., Cherny, M., Pamer, E.G., 2009. Selective expansion of the
12071216, http://dx.doi.org/10.1038/ni1518. monocytic lineage directed by bacterial infection. J. Immunol. 183, 19001910,
Palframan, R.T., Jung, S., Cheng, G., Weninger, W., Luo, Y., Dorf, M., Littman, D.R., http://dx.doi.org/10.4049/jimmunol.0900612.
Rollins, B.J., Zweerink, H., Rot, A., Von Andrian, U.H., 2001. Inammatory Serbina, N.V., Pamer, E.G., 2006. Monocyte emigration from bone marrow during
chemokine transport and presentation in HEV: a remote control mechanism bacterial infection requires signals mediated by chemokine receptor CCR2. Nat.
for monocyte recruitment to lymph nodes in inamed tissues. J Exp Med 194, Immunol. 7, 311317, http://dx.doi.org/10.1038/ni1309.
13611373. Serbina, N.V., Salazar-Mather, T.P., Biron, C.A., Kuziel, W.A., Pamer, E.G., 2003.
Palucka, A.K., 1998. Dendritic cells as the terminal stage of monocyte differentiation. TNF/iNOS-producing dendritic cells mediate innate immune defense against
J. Immunol. 160, 45874595. bacterial infection. Immunity 19, 5970.
Peters, W., Scott, H.M., Chambers, H.F., Flynn, J.L., Charo, I.F., Ernst, J.D., 2001. Shi, C., Jia, T., Mendez-Ferrer, S., Hohl, T.M., Serbina, N.V., Lipuma, L.,
Chemokine receptor 2 serves an early and essential role in resistance to Leiner, I., Li, M.O., Frenette, P.S., Pamer, E.G., 2011. Bone marrow mes-
Mycobacterium tuberculosis. Proc. Natl. Acad. Sci. U. S. A. 98, 79587963, enchymal stem and progenitor cells induce monocyte emigration in
http://dx.doi.org/10.1073/pnas.131207398. response to circulating toll-like receptor ligands. Immunity 34, 590601,
Pne, F., Courtine, E., Ouaaz, F., Zuber, B., Sauneuf, B., Sirgo, G., Rousseau, C., Toubiana, http://dx.doi.org/10.1016/j.immuni.2011.02.016.
J., Balloy, V., Chignard, M., Mira, J.P., Chiche, J.D., 2009. Toll-like receptors 2 and 4 Sponaas, A.-M., Freitas do Rosario, A.P., Voisine, C., Mastelic, B., Thompson, J.,
contribute to sepsis-induced depletion of spleen dendritic cells. Infect. Immun. Koernig, S., Jarra, W., Renia, L., Mauduit, M., Potocnik, A.J., Langhorne, J.,
77, 56515658, http://dx.doi.org/10.1128/IAI.00238-09. 2009. Migrating monocytes recruited to the spleen play an important role
Phythian-Adams, A.T., Cook, P.C., Lundie, R.J., Jones, L.H., Smith, K.A., Barr, T.A., in control of blood stage malaria. Blood 114, 55225531, http://dx.doi.org/
Hochweller, K., Anderton, S.M., Hammerling, G.J., Maizels, R.M., MacDonald, A.S., 10.1182/blood-2009-04-217489.
226 K. Bieber, S.E. Autenrieth / Immunobiology 220 (2015) 215226

Sponaas, A.M., 2006. Malaria infection changes the ability of splenic dendritic cell Tsou, C.-L., Peters, W., Si, Y., Slaymaker, S., Aslanian, A.M., Weisberg, S.P., Mack, M.,
populations to stimulate antigen-specic T cells. J. Exp. Med. 203, 14271433, Charo, I.F., 2007. Critical roles for CCR2 and MCP-3 in monocyte mobilization
http://dx.doi.org/10.1084/jem.20052450. from bone marrow and recruitment to inammatory sites. J. Clin. Invest. 117,
Steinman, R.M., et al., 2002. Avoiding horror autotoxicus: the importance of dendritic 902909, http://dx.doi.org/10.1172/JCI29919.
cells in peripheral T cell tolerance. Proc. Natl. Acad. Sci. U. S. A. 99, 351358. Varol, C., Landsman, L., Fogg, D.K., Greenshtein, L., Gildor, B., Margalit, R.,
Steinman, R.M., Banchereau, J., 2007. Taking dendritic cells into medicine. Nature Kalchenko, V., Geissmann, F., Jung, S., 2007. Monocytes give rise to mucosal,
449, 419426, http://dx.doi.org/10.1038/nature06175. but not splenic, conventional dendritic cells. J. Exp. Med. 204, 171180,
Sunderktter, C., Nikolic, T., Dillon, M.J., van Rooijen, N., Stehling, M., Drevets, D.A., http://dx.doi.org/10.1084/jem.20061011.
Leenen, P.J.M., 2004. Subpopulations of mouse blood monocytes differ in matu- Verschoor, A., Neuenhahn, M., Navarini, A.A., Graef, P., Plaumann, A., Seidlmeier,
ration stage and inammatory response. J. Immunol. 172, 44104417. A., Nieswandt, B., Massberg, S., Zinkernagel, R.M., Hengartner, H., Busch, D.H.,
Sundquist, M., Wick, M.J., 2005. TNF-alpha-dependent and -independent maturation 2011. A platelet-mediated system for shuttling blood-borne bacteria to CD8+
of dendritic cells and recruited CD11c(int) CD11b+ cells during oral Salmonella dendritic cells depends on glycoprotein GPIb and complement C3. Nat. Immunol.
infection. J. Immunol. 175, 32873298. 12, 11941201, http://dx.doi.org/10.1038/ni.2140.
Sundquist, M., Wick, M.J., 2009. Salmonella induces death of CD8+ dendritic cells but Villadangos, J.A., Schnorrer, P., 2007. Intrinsic and cooperative antigen-presenting
not CD11cint CD11b+ inammatory cells in vivo via MyD88 and TNFR1. J. Leukoc. functions of dendritic-cell subsets in vivo. Nat. Rev. Immunol. 7, 543555,
Biol. 85, 225234, http://dx.doi.org/10.1189/jlb.0708413. http://dx.doi.org/10.1038/nri2103.
Tam, M.A., Wick, M.J., 2004. Dendritic cells and immunity to Listeria: Vremec, D., Pooley, J., Hochrein, H., Wu, L., Shortman, K., 2000. CD4 and CD8 expres-
TipDCs are a new recruit. Trends Immunol. 25, 335339, http://dx.doi.org/ sion by dendritic cell subtypes in mouse thymus and spleen. J. Immunol. 164,
10.1016/j.it.2004.05.004. 29782986.
Tam, M.A., Wick, M.J., 2006. Differential expansion, activation and effector func- Wan, S., Zhou, Z., Duan, B., Morel, L., 2008. Direct B cell stimulation by den-
tions of conventional and plasmacytoid dendritic cells in mouse tissues dritic cells in a mouse model of lupus. Arthr. Rheum. 58, 17411750,
transiently infected with Listeria monocytogenes. Cell. Microbiol. 8, 11721187, http://dx.doi.org/10.1002/art.23515.
http://dx.doi.org/10.1111/j.1462-5822.2006.00700.x. Waskow, C., Liu, K., Darrasse-Jze, G., Guermonprez, P., Ginhoux, F., Merad, M., Shen-
Taylor, P.R., Brown, G.D., Geldhof, A.B., Martinez-Pomares, L., Gordon, S., 2003. gelia, T., Yao, K., Nussenzweig, M., 2008. The receptor tyrosine kinase Flt3 is
Pattern recognition receptors and differentiation antigens dene murine required for dendritic cell development in peripheral lymphoid tissues. Nat.
myeloid cell heterogeneity ex vivo. Eur. J. Immunol. 33, 20902097, Immunol. 9, 676683, http://dx.doi.org/10.1038/ni.1615.
http://dx.doi.org/10.1002/eji.200324003. Wu, L., DAmico, A., Hochrein, H., OKeeffe, M., Shortman, K., Lucas, K., 2001.
Tian, T., Woodworth, J., Skld, M., Behar, S.M., 2005. In vivo depletion of CD11c+ cells Development of thymic and splenic dendritic cell populations from different
delays the CD4+ T cell response to Mycobacterium tuberculosis and exacerbates hemopoietic precursors. Blood 98, 33763382.
the outcome of infection. J. Immunol. 175, 32683272. Yarovinsky, F., 2005. TLR11 activation of dendritic cells by a protozoan prolin-like
Tittel, A.E.P., Heuser, C., Ohliger, C., Llanto, C., Yona, S., mmerling, G.U.N.J.H.A., protein. Science 308, 16261629, http://dx.doi.org/10.1126/science.1109893.
Engel, D.R., Garbi, N., Kurts, C., 2012. Functionally relevant neutrophilia Ynez, A., Megas, J., OConnor, J.-E., Gozalbo, D., Gil, M.L., 2011. Candida
in CD11c diphtheria toxin receptor transgenic mice. Nat. Methods, 17, albicans induces selective development of macrophages and monocyte derived
http://dx.doi.org/10.1038/nmeth.1905. dendritic cells by a TLR2 dependent signalling. PLoS ONE 6, e24761,
Tripp, C.S., Wolf, S.F., Unanue, E.R., 1993. Interleukin 12 and tumor necrosis http://dx.doi.org/10.1371/journal.pone.0024761.g009.
factor alpha are costimulators of interferon gamma production by natural Ynez, A., Murciano, C., OConnor, J.-E., Gozalbo, D., Gil, M.L., 2009. Candida
killer cells in severe combined immunodeciency mice with listeriosis, and albicans triggers proliferation and differentiation of hematopoietic stem and
interleukin 10 is a physiologic antagonist. Proc. Natl. Acad. Sci. U. S. A. 90, progenitor cells by a MyD88-dependent signaling. Microb. Infect. 11, 531535,
37253729. http://dx.doi.org/10.1016/j.micinf.2009.01.011.

S-ar putea să vă placă și