Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Oct 1.
Published in final edited form as: Cytokine Growth Factor Rev. 2016 May 9;31:1–15. doi: 10.1016/j.cytogfr.2016.05.001

STAT3 signaling in immunity

Emily J Hillmer 1,*, Huiyuan Zhang 1,*, Haiyan S Li 1,*, Stephanie S Watowich 1,2
PMCID: PMC5050093  NIHMSID: NIHMS786860  PMID: 27185365

Abstract

The transcriptional regulator STAT3 has key roles in vertebrate development and mature tissue function including control of inflammation and immunity. Mutations in human STAT3 associate with diseases such as immunodeficiency autoimmunity and cancer. Strikingly, however, either hyperactivation or inactivation of STAT3 results in human disease, indicating tightly regulated STAT3 function is central to health. Here, we attempt to summarize information on the numerous and distinct biological actions of STAT3, and highlight recent discoveries, with a specific focus on STAT3 function in the immune and hematopoietic systems. Our goal is to spur investigation on mechanisms by which aberrant STAT3 function drives human disease and novel approaches that might be used to modulate disease outcome.

Keywords: STAT3, cytokines, immune system, inflammation

Graphical Abstract

graphic file with name nihms786860f6.jpg

Introduction: STAT3 discovery, structure and transcriptional function

STAT3 was discovered over 20 years ago as a component of the interleukin-6- (IL-6) activated acute phase response factor (APRF) complex [13], which has a crucial role in stimulating expression of innate immune mediators in liver. This discovery led to rapid identification of STAT3 as a member of the STAT (Signal Transducers and Activators of Transcription) family, based on size similarity, antigenic and structural relatedness, as well as comparable DNA binding activity, to the interferon (IFN)-responsive STAT proteins. Subsequent work identified 7 members of the STAT protein family in mammals [36]. Activation of STAT3 is elicited by numerous cytokines and growth factors, including cytokines utilizing the IL-6 signal-transducing receptor chain gp130 (e.g., IL-6, oncostatin M, interleukin-11) or homodimeric cytokine receptors (e.g., granulocyte colony-stimulating factor, G-CSF), as well as growth factors that act through protein tyrosine kinase receptors (e.g., epidermal growth factor) [2, 3, 7, 8]. Moreover, STAT3 mediates important signal transduction cascades elicited by intracellular proteins such as activated Ras or tyrosine kinase oncoproteins (e.g., Src) [914]. Many early studies foreshadowed the multiple and distinct biological roles for STAT3 that are appreciated today. Accordingly, interest in STAT3 has risen substantially since its discovery, as judged by a survey of STAT3-immune system-related publications (Figure 1).

Figure 1. STAT3-immune publication numbers.

Figure 1

The number of publications listed in PubMed with the query “STAT3 immune” is shown for each year between 1994 and 2015.

The primary amino acid sequence of STAT3 revealed a conserved Src homology 2 (SH2) domain and a C-terminal tyrosine residue (Y705 in mice) that becomes phosphorylated by Jak kinases upon cytokine stimulation, protein tyrosine kinase receptor signaling or intracellular protein tyrosine kinase activation [5, 9]. STAT3 forms homodimers by reciprocal SH2 domain-phosphotyrosine interactions between 2 monomers; this was identified as a key activating mechanism leading to stimulation of STAT3 transcriptional function. STAT3 also undergoes serine phosphorylation at position 727 (S727), a modification that enhances transcriptional activity [1518]. STAT3 DNA association is mediated by a central DNA-binding region (Figure 2), while protein:protein association domains located at the STAT3 N- and C-terminal regions are also involved in transcriptional regulation. Numerous approaches including sequence comparisons, mutational analyses, biochemical and structural studies of STAT3 and other family members led to these important discoveries [1931]. Further posttranslational modifications such as acetylation and methylation have been implicated more recently in STAT3 transcriptional function [3235]. Moreover, STAT3 can be activated constitutively by engineered introduction of cysteine residues, which drive cytokine-independent dimerization, rendering oncogenic activity [36]. Several excellent reviews summarize the discovery of STATs and the intense work to characterize their signaling mechanisms and functions in the early days of the field [3742].

Figure 2. Schematic illustration of a STAT3 dimer bound to DNA.

Figure 2

A representation of the major STAT3 domains resolved in the X-ray crystal structure of the STAT3-DNA binding complex is shown. Examples of STAT3 mutations associated with STAT3 AD-HIES (LOF) or autoimmunity (GOF) are indicated.

More recently, unphosphorylated STAT3 (uSTAT3) has been recognized as an important transcriptional regulator [4345]. In the unphosphorylated state, uSTAT3 binds similar DNA sites as tyrosine-phosphorylated and dimerized STAT3 (e.g., IFN-γ-activated sequence (GAS) elements), yetuSTAT3 works in collaboration with transcriptional regulators such as NF-κB to control a cadre of genes not normally affected by tyrosine-phosphorylated STAT3 [34, 44, 46]. STAT3 also induces its own gene expression via a STAT3- Stat3 positive autoregulatory loop [47]. Thus, STAT3 homodimers activated by cytokine or growth factor receptors, as well as intracellular protein tyrosine kinases, have potential to boost total STAT3 protein amounts, increasing availability of uSTAT3 and STAT3 as a tyrosine kinase substrate. Accordingly, STAT3 autoregulation provides a mechanism to shape gene expression temporally throughout the duration of a cytokine response. Further work is needed to understand the global transcriptional impact of uSTAT3 and the tyrosine-phosphorylated form in the immune response as well as induction or maintenance of immunological diseases and cancer.

Non-transcriptional activities of STAT3

STAT3 also has important activities in cellular respiration, metabolism, autophagy and cancer that are separate from transcriptional roles mediated by the tyrosine-phosphorylated or unphosphorylated isoforms. In 2009, the intriguing discovery was made that STAT3 associates with mitochondria and therein regulates electron transport chain function as well as glycolytic and oxidative phosphorylation [13, 48]. Mitochondrial STAT3 is also implicated in control of the gamma-glutamyl cycle, production of glutathione and regulation of reactive oxygen species (ROS) [49]. Furthermore, in embryonic stem (ES) cells, STAT3 controls mitochondrial gene expression and respiration, mechanisms that optimize ES cell proliferation and maintenance [50]. By contrast, genetic screens identified STAT3 as a negative regulator of autophagy via inhibition of type III phosphoinositol 3-kinase (PI3K) or association with the eukaryotic translation initiation factor 2a (eIF2a) kinase 2/protein kinase R (EIF2AK2/PKR), respectively [5153].

Significantly, the mitochondrial function of STAT3 is critical for Ras-mediated oncogenic transformation [13,14]. Ras proteins (e.g., KRas, NRas and HRas) belong to a family of small GTPases; gain-of-function (GOF) mutations in Ras homologs were the first oncogenic mutations identified in human cancers and are associated with a variety of malignancies [5456]. Ras GOF mutations stimulate MEK-Erk signaling and STAT3 S727 phosphorylation, which is essential for Ras-mediated malignant transformation [14]. Recent studies using KRas-driven pancreatic and myeloproliferative tumor models demonstrated a requirement for STAT3 in oncogenesis in vivo, yet conditional Stat3 ablation concomitant with KRas activation in lung epithelial cells had the unexpected result of enhancing Kras-driven lung tumor progression [5759]. These disparate findings suggest additional complexity in the oncogenic activity of mitochondrial-associated STAT3, which may relate to tissue-specific roles. Thus, further studies using conditional Stat3 ablation in spontaneous Ras-driven tumor models as well as investigation of mitochondrial STAT3 function in human tumor specimens are needed to clarify the function of mitochondrial STAT3 in cancer.

STAT3 mutations in human immune disorders

STAT3 inactivation and HIES

Autosomal dominant STAT3 inactivating mutations in the human immunodeficiency condition termed Hyper immunoglobulin E syndrome (HIES) reveal a causal role for STAT3 loss-of-function (LOF) in human immune disease. HIES can be triggered by mutation of several immune regulatory proteins (e.g., Tyk2, DOCK8) [6062]; however, a comparison of symptoms in a subgroup of HIES patients with the roles for STAT3 in the immune system led 2 independent groups to identify missense and point mutations in STAT3 that abrogate STAT3 transcriptional function and subsequent biological responses [63, 64]. The mutations include disruptions in STAT3 DNA-binding, SH2 and transactivation domains (Figure 2; not shown) [63, 64]. Characteristic features of HIES associated with STAT3 mutation (STAT3 AD-HIES) comprise recurring bacterial infections of skin and lungs, enhanced oral fungal infection, elevated innate immune pro-inflammatory responses, bone and connective tissue abnormalities and increased circulating immunoglobulin E (IgE) [6568]. Key evidence implicating STAT3 in AD-HIES included upregulation of mRNAs encoding host defense, IFN-inducible and signal transduction components, collectively suggesting altered cytokine responses. In addition, a complex phenotype marked by elevated pro-inflammatory cytokine production upon LPS stimulation, yet defective IL-6-responsiveness, mimics aspects of conditional Stat3-deficient mouse models [63, 69, 70].

An important advance in understanding the select organ manifestation of STAT3 AD-HIES (e.g., impaired skin and lung immunity) was made upon observations that human keratinocytes and bronchial epithelial cells require the combined activity of cytokines from STAT3-dependent, IL-17-producing CD4+ T lymphocytes (Th17 cells) and pro-inflammatory cytokines to generate antibacterial mediators and neutrophil chemoattractants [71]. By contrast, other cell types activate antimicrobial responses in response to pro-inflammatory cytokines alone. Thus, deficiency in the STAT3-dependent Th17 lineage plays a significant role in the tissue-restricted phenotype of STAT3 AD-HIES.

Evidence of immune system dysfunction in STAT3 AD-HIES might suggest hematopoietic stem cell transplantation as an effective therapy to treat the disease. Recently, bacterial artificial chromosome (BAC) engineering was used to generate a transgenic mouse model of STAT3 AD-HIES [72]. These animals overexpress a DNA-binding-defective STAT3 isoform (Stat3 ΔV463), which recapitulates several features of the human disease [72]. By testing bone marrow transplantation, Steward-Tharp et al. demonstrated, however, that effective host defense responses require functional STAT3 in both hematopoietic and non-hematopoietic compartments. In other words, appropriate inflammatory and immune responses were only partially restored by transplantation of Stat3-sufficient hematopoietic cells into mice carrying the mutant allele [72]. These data underscore the powerful utility of the STAT3 AD-HIES mouse as a model to examine molecular and cellular events, as well as communication between immune and non-immune components, that regulate immune competency [72].

STAT3 hyperactivation, autoimmunity and immunodeficiency

Early observations implicating a role for STAT3 in human autoimmunity centered on association of STAT3 gene variations (e.g., single nucleotide polymorphisms, SNPs) with increased predisposition to psoriasis and multiple sclerosis; STAT3 SNPs are also linked with inflammatory bowel disease [7376]. Recently, de novo activating point mutations in STAT3 (i.e., GOF) were identified in individuals with juvenile-onset autoimmunity and lymphoproliferation. Disease manifestations include type I diabetes in infancy, lymphadenopathy, autoimmune cytopenias, primary hypothyroidism, solid organ autoimmunity and short stature [7780]. The STAT3 point mutations in these individuals affect conserved residues in the DNA binding, SH2 or transactivation domains, which are different from those mutated in STAT3 AD-HIES, as well as a residue located in the STAT3 N-terminal coiled:coil domain (Figure 2; not shown) [7779]. STAT3 GOF mutations linked with autoimmunity induce elevated basal and cytokine-responsive STAT3 transcriptional activity, yet constitutive STAT3 tyrosine phosphorylation is not detected [7779]. Biochemical and molecular modeling experiments suggest autoimmunity-associated GOF mutations enhance STAT3 DNA binding activity, which presumably elevates STAT3-mediated transcriptional responses [77]. Furthermore, the activity of other STATs is altered upon hyperactivation of STAT3, as both STAT1 and STAT5 signaling are suppressed in cells expressing autoimmunity-associated STAT3 mutants. Effects on other STATs may explain certain disease features such as short stature or aberrant immune competency, processes that are regulated by growth hormone (GF)-STAT5 or IFN-STAT1 signaling, respectively [7779].

Individuals carrying STAT3 GOF mutations show reduced regulatory T lymphocytes (Tregs), consistent with roles for STAT3 in restraining FoxP3 expression and Treg development [8183], as well as the importance of Tregs in suppressing autoimmunity [8488]. Surprisingly, however, other immune subsets that were previously reported to be STAT3-dependent were also reduced in some individuals with STAT3 GOF mutations. For example, fewer plasmactyoid dendritic cells (pDCs), natural killer (NK) cells and Th17 cells were detected in certain cases, relative to amounts in healthy controls [78]. Moreover, reduced amounts of class-switched memory B lymphocytes and lower circulating IgG amounts were found, implying coexisting immune defects. One individual carrying an activating STAT3 mutation developed disseminated mycobacterial disease, which often associates with impaired DC/antigen-presenting populations, and may reflect a more global DC deficiency [78]. A second individual developed T-cell large granular lymphocytic leukemia, indicating the malignant potential of de novo STAT3 GOF mutations [78]. Overall, a complex phenotype of autoimmunity and specific immune deficiencies presents with STAT3 GOF mutation [80]. While this disease has yet to be modeled in the murine system for pre-clinical studies, there is encouraging evidence that IL-6 inhibition or bone marrow transplantation may provide treatment options [79].

Conditional deletion of Stat3 in mice

Stat3 gene deletion in mice is used as a principal approach to evaluate systemic, cellular and molecular roles of STAT3. Since germline Stat3 deletion leads to embryonic lethality [89], conditional Stat3 ablation using the cre-LoxP system is necessary to study developmental and mature organ functions. Four separate Stat3 floxed alleles (Stat3f) and corresponding Stat3flox/flox (Stat3f/f) mouse strains have been generated and analyzed [70, 9093]. Each Stat3f allele targets distinct regions of Stat3 for removal, yet in all cases these were designed to eliminate STAT3 domains necessary for canonical, tyrosine-phosphorylation-mediated transcriptional activity [70, 90, 92, 93]. Here, we provide brief descriptions of the distinct Stat3f alleles, which have been used in numerous reports on STAT3 immune functions.

Takeda, Akira and colleagues generated a Stat3f allele in which portions of exons 21 and 22 are flanked by loxP sites [70]. Upon cre-mediated deletion, this Stat3f allele encodes an internally deleted STAT3 protein missing 29 amino acids, including Y705 and S727. As expected, this mutant protein fails to undergo cytokine-responsive tyrosine phosphorylation and is predicted to be deficient in serine phosphorylation [70]. A very low level of a truncated STAT3 protein can be observed in cre-expressing hematopoietic and immune cells containing 1 or 2 copies of this Stat3f allele. Based on the protein coding regions targeted and cross-reactivity with C-terminal-specific antibodies, the truncated STAT3 protein appears to result from an in-frame internal deletion due to the location of the loxP sites [70, 94]. While it is formally possible that a low amount of the internal deletion STAT3 mutant has certain unknown biological function, this Stat3f strain recapitulates data obtained with distinct Stat3f alleles [69, 92, 93], indicating that it permits full ablation of canonically activated STAT3.

Raz, Lee, Levy and colleagues developed a Stat3f allele with loxP sites bracketing exons 16–21 [90, 91]. Cre-mediated excision of these floxed exons removes sequences encoding the STAT3 SH2 domain and Y705, and renders complete absence of tyrosine-phosphorylated as well as full-length STAT3 protein [90, 91]. Although the STAT3 S727- or TAD-encoding sequences were not removed through this approach, the targeting construct does not generate an in-frame internal STAT3 deletion and thus effectively eliminates all STAT3 sequences C-terminalto exon 21 [90,91].

Welte et al generated a Stat3f allele in which exons 18–20 are flanked by loxP sequences [92]. Removal of exons 18–20 upon cre expression leads to deletion of the STAT3 SH2 domain, which is expected to eliminate transcriptional activity dependent on STAT3 Y705 phosphorylation and STAT3 homodimerization. STAT3 protein was not observed in immunoblotting assays with antibodies that recognize the C-terminus of STAT3 [92], implying a lack of in-frame sequences beyond exon 20 (e.g., encoding Y705, S727 and TAD). These data indicate efficient depletion of canonically activated STAT3 protein [92].

Alzoni, Poli and colleagues developed a Stat3f allele with loxP sites flanking exons 12 to 14, which encode the DNA-binding domain of STAT3 [93]. This Stat3f allele generates a truncated and frameshifted mRNA, predicted to encode a STAT3 protein deficient in DNA-binding. Thus, this approach should ablate both canonically activated (tyrosine-phosphorylated) as well as uSTAT3 transcriptional functions. STAT3 protein levels produced from this Stat3f allele upon cre expression were greatly reduced and Stat3-deficient animals recapitulated phenotypes observed with other mutant Stat3 strains, indicating effective deletion of STAT3 [93].

Since homozygosity (Stat3f/f) of each of the 4 Stat3f alleles renders severe reduction or non-detectable amounts of tyrosine-phosphorylated, full-length STAT3 in cre expressing cells [70, 9093, 95], we refer to these strains collectively as Stat3f/f or Stat3-deficient. Nonetheless, it is important to understand the Stat3f allele utilized in various literature reports due to new information on roles for uSTAT3, mitochondrial STAT3 and potential for residual canonical or non-canonical function in the absence of complete protein depletion.

STAT3 in innate immunity

Emergency and steady state granulopoiesis

STAT3 regulates critical steps during emergency granulopoiesis, a key innate response elicited upon bacterial or fungal invasion that provides a rapid increase in the supply of circulating neutrophils to help contain infection. Emergency granulopoiesis involves both the swift release of mature neutrophils from the bone marrow reserve as well as the induction of granulopoiesis de novo to sustain elevated neutrophil output [96]. Elegant studies indicate that initial pathogen encounter with endothelial cells (ECs) stimulates ECs to synthesize G-CSF, the major neutrophil growth factor, which then induces granulocytic progenitor proliferation and neutrophil mobilization responses in bone marrow [9799]. Importantly, emergency granulopoiesis can be mimicked by delivery of supraphysiologic amounts of recombinant G-CSF, a situation also encountered clinically during G-CSF treatment of immunocompromised individuals [100].

STAT3 is the primary STAT protein activated upon G-CSF engagement with G-CSFR [101]. Initial evidence for the involvement of STAT3 in emergency granulopoiesis was obtained by study of the Tg(Tek-cre)12Flv Stat3f/f mice (Tek-cre Stat3f/f, also known as Tie2 cre Stat3f/f), in which cre expression is driven by the Tek/Tie2 promoter. These animals have been used as a model to understand hematopoietic STAT3 function [94,102106]. Upon systemic administration of G-CSF or delivery of the chemokine MIP-2, an agonist for the neutrophil chemoattractant receptor CXCR2, Tek-cre Stat3f/f mice fail to mobilize mature neutrophils from the bone marrow effectively, relative to Stat3-sufficient controls. Furthermore, FACS-purifiedStat3-deficient neutrophils from Tek-cre Stat3f/f mice are impaired in their ability to undergo chemotaxis toward CXCR2 ligands (i.e., MIP-2, KC) ex vivo [94,104]. These data indicate STAT3 regulates neutrophil release to the circulation and CXCR2-mediated chemotaxis needed to access infected tissues, which are key steps in emergency granulopoiesis. Tek-cre Stat3f/f mice also fail to upregulate immature granulocyte amounts in bone marrow or peripheral blood. This defective response reflects a vital role for STAT3 in G-CSF-driven proliferation of granulocytic progenitor cells, which undergo rapid expansion during emergency granulopoiesis [94,103]. It is critical to recognize, however, that Tek-cre Stat3f/f animals also have endothelial Stat3-deficiency due to the Tie2 expression pattern. While this has potential to contribute to observed phenotypes, data from ex vivo proliferation and chemotaxis assays, as well as the identification of STAT3 target genes mediating emergency granulopoiesis, imply key hematopoietic cell-intrinsic roles for STAT3 [94,103].

During the proliferative response to G-CSF in emergency granulopoiesis, STAT3 stimulates expression of C/EBPβ and c-Myc in bone marrow granulocytic progenitors; these regulators have critical functions in driving enhanced G1/S phase progression and induction of the emergency response (Figure 3) [103,107,108]. Molecular studies indicate STAT3 mediates transcriptional activation of Cebpb and c-myc directly [103]. Moreover, STAT3-dependentupregulation of C/EBPβ also promotes C/EBPβ association at the c-myc promoter. The concurrent association of STAT3 and C/EBPβ at the c-myc promoter displaces the negative regulator C/EBPα, resulting in enhancement of c-myc transcription [103,107]. STAT3 also stimulates expression of CXCR2/Il8rb and MIP-2/Cxcl2 by direct promoter binding upon G-CSF stimulation to augment neutrophil migratory capability [104,109].

Figure 3. Regulation and effects of STAT3 signaling in myelopoiesis.

Figure 3

Activation of STAT3 by the G-CSFR during emergency granulopoiesis drives expression of C/EBPβ and c-Myc in granulocytic progenitors, inducing their proliferation and increasing neutrophil production. IFN-γ stimulates SOCS3 expression, which blocks G-CSFR-STAT3 signaling, and promotes PU.1 and IRF8 induction to drive monocyte generation.

The aforementioned responses are involved in driving emergency granulopoiesis, however, STAT3 is correspondingly required to restrain neutrophil production and limit inflammatory responses [91,110]. A key negative regulatory mechanism is directed by STAT3-dependent induction of SOCS3, an important signaling intermediate that suppresses the activity of the G-CSFR receptor (G-CSFR) as well as a subset of other cytokine receptors [91,111113]. STAT3 stimulates Socs3 transcription rapidly upon cytokine engagement, from nearly undetectable amounts in basal conditions to a greater than 10-fold increase in mRNA and protein expression [114]. Accumulated SOCS3 protein in the cytoplasm interacts with receptor/Jak complexes through two distinct interfaces simultaneously, resulting in inhibition of receptor-mediated signal transduction. The central SH2 domain of SOCS3 binds to the activated (tyrosine-phosphorylated) receptor intracellular region via a classic SH2-phosphotyrosine interaction. In parallel, the kinase-inhibitory region (KIR) of SOCS3 located near the N-terminus associates with the receptor-associated Jak protein, binding within the Jak-substrate interaction groove, which inhibits the Jak kinase from association with other substrates [115,116]. These SOCS3-mediated mechanisms have been established by study of IL-6 receptor signaling, and are thought to extend to G-CSFR as well as specific other cytokine receptors effectively blocked by SOCS3 [112,113,115117].

Collectively, therefore, information from numerous murine models indicates STAT3 orchestrates key proliferative and neutrophil migratory functions required for emergency granulopoiesis, and also limits the duration of this response to prevent destructive inflammation. Furthermore, Tek-cre Stat3f/f animals have defective clearance of Listeria monocytogenes, while Stat3-deficient neutrophils and macrophages show impaired bactericidal activity, indicating critical STAT3 antibacterial functions [104,118]. These activities of STAT3 maybe conserved, since STAT3 AD-HIES individuals are susceptible to certain bacteria and isolated neutrophils show migration impairments [94,104,119122]. Further investigation is needed to examine roles for STAT3 in mediating human neutrophil responses.

In steady state, G-CSF and G-CSFR also serve as the major ligand-receptor pair controlling neutrophil development [98,101,123126]. Thus it was expected STAT3 would be required to mediate neutrophil production in homeostasis. Surprisingly, however, animals with Stat3 deletion in hematopoietic cells (e.g., Tek-cre Stat3f/f mice or global Mx-cre-mediated Stat3 deletion) show elevated amounts of circulating neutrophil numbers [91, 92, 94]. The key mechanism thought to underlie this excessive neutrophil accumulation centers on STAT3-dependent control of SOCS3, since Stat3-deficiency as well Socs3-deficiency in the hematopoietic system leads to neutrophilia [91, 111]. STAT3-mediated regulation of neutrophil chemotactic factors may also prevent effective neutrophil margination in tissues [94,104,109,127], contributing to neutrophil accumulation in the circulation, yet this hypothesis must be further examined.

STAT3-responsive pathways in developing granulocytes are affected by other cytokine cues, with significant impact upon granulopoiesis. Type IIIFN (IFN-γ), which is produced by activated CD4+ and CD8+ T lymphocytes as well as NK and NKT cells, preferentially stimulates monocyte generation from the granulocyte-monocyte progenitor (GMP) population, at the expense of granulocytic cell production [128,129]. The action of IFN-γ on monocytes and macrophages is critical for enhancing immunity toward intracellular bacteria [130], thus IFN-γ-driven monocyte generation can be viewed as a feedforward mechanism to promote a dedicated immune response for this specific pathogen class. IFN-γ potently inhibits STAT3 activation in myeloid progenitor cells while simultaneously inducing genes required for monocyte differentiation (e.g., PU.1, IRF8). IFN-γ-dependent STAT3 inhibition is thought to be regulated via IFN-γ-responsive SOCS3 induction and subsequent inhibition of G-CSFR-STAT3-mediated proliferation within the GMP and common myeloid progenitor (CMP) populations [129]. Thus, cytokine-driven stimulation or inhibition of STAT3 activity in myeloid progenitors may be key to tailoring hematopoietic output, to meet unique demands upon the immune system during different infection or emergency conditions (Figure 3) [103,129,131].

Dendritic cell (DC) development and function

DCs comprise several distinct populations that are identified by unique cell surface marker expression, sites of anatomical residence and functional responses to damage- and pathogen-associated molecular patterns (DAMPs and PAMPs, respectively) elicited in tissue injury and infection [132,133]. The classic division in DC lineages is drawn between the plasmacytoid DCs (pDCs), professional type IIFN producing cells, and conventional DCs, which reside in lymphoid and non-lymphoid tissues and have potent phagocytic and antigen presenting activities [132,133].

Initial work with Tek-cre Stat3f/f mice indicated a critical role for STAT3 in development of pDC and cDC lineages [102]. This function was particularly evident in DC generation driven by administration of exogenous Fms-related tyrosine kinase 3 (Flt3) ligand (Flt3L), the major DC growth factor in vivo [102,134]. These data agree with selective STAT3 activation (versus other STATs) by Flt3L, as well as the critical role for STAT3 in stimulating Flt3L-responsive DC progenitor proliferation [106,135]. Recently, prostaglandin E2 (PGE2) has been implicated in optimal Flt3L-mediated DC generation via a STAT3-dependent pathway [136], indicating non-cytokine signals can converge on DC progenitors to drive STAT3 activation and DC production.

Studies employing overexpression of STAT3 or Flt3 (the receptor for Flt3L) within Flt3-negative hematopoietic progenitors, which normally do not have the ability to produce DCs, implicated key instructive functions for these factors in DC development [137,138]. For example, constitutive Flt3 expression promotes induction of genes involved in DC development (Stat3, Spi1/PU.1) and enhances pDC and cDC generation [138]. Similarly, STAT3 overexpression stimulates Flt3 expression and DC development [138]. Additional work implicated STAT3 in driving Spi1/PU.1 expression [127,139]. Furthermore, PU.1 is critical for Flt3 expression and DC development in vivo [140]. These data collectively suggest the existence of a positive, feedforward molecular pathway mediated by Flt3L-Flt3-STAT3 signaling and PU.1 that promotes DC development [138,140,141].

More recent work with Tek-cre Stat3f/f or Tg(Itgax-cre)1-1Reiz Stat3f/f (Itgax-cre, also known as CD11c cre) animals confirms the crucial role for STAT3 in pDC generation, yet indicates steady state production of lymphoid organ CD8α+ cDCs and nonlymphoid resident CD103+ DCs is STAT3-independent [106,142]. Since Stat3 deletion is restricted to CD11c+ populations in Itgax-cre Stat3f/f mice, it is possible cDCs require STAT3 only before reaching a CD11c-expressing, DC-committed precursor stage. By contrast, a likely explanation for the differences obtained in separate studies of Tek-cre Stat3f/f mice may center on the severe inflammatory disease that develops in these animals, which causes early lethality (e.g., 6–8 weeks) in specific pathogen-free housing conditions [92, 94]. Systemic inflammation may have deleterious consequences on hematopoietic stem and progenitor populations, indirectly affecting DC output. Further studies to explore cell autonomous roles for STAT3 in homeostatic DC generation are needed to examine this issue more carefully.

In addition to STAT3, pDC development is critically dependent upon the basic helix-loop-helix (bHLH) transcriptional regulator E2-2 [143]. Conversely, pDC generation and pDC-mediated type IIFN production are suppressed by inhibitor of differentiation 2 (Id2), a member of the Id protein family that blocks the DNA binding activity of bHLH-containing transcription factors including E2-2 [144,145]. These findings raised the question of whether STAT3 and/or other STATs control DC lineage-regulatory factors. Experiments using Tek-cre Stat3f/f mice demonstrated a critical role for STAT3 in mediating Flt3L-responsive Tcf4/E2-2 expression in common DC progenitors (CDPs). Moreover, molecular assays indicate STAT3 directly stimulates Tcf4/E2-2 transcription. Taken together, these results suggest the Flt3L-Flt3-STAT3 pathway promotes pDC generation from CDPs via direct induction of Tcf4 [106,146]. By contrast, GM-CSF and STAT5 upregulate Id2 in CDPs and inhibit pDC development [106]. These studies reveal cytokine-STAT pathways that influence commitment to specific DC lineages, yet they are likely to reflect only a small fraction of STAT3- (or other STAT-) driven molecular responses. Global STAT3 transcriptional profiling and STAT3 chromatin association studies from defined hematopoietic progenitors are needed to fully appreciate STAT3 mechanisms in DC development.

Numerous reports indicate STAT3 suppresses DC maturation and activation, and promotes tolerogenic function [147151]. This response is attributed to inhibition of MHC class II and co-stimulatory molecule expression; upregulation of myeloid-related protein SA100A9, which suppresses DC function; induction of inhibitory programmed death ligand-1 (PD-L1) on DCs; and STAT3-mediated restraint of TLR-induced pro-inflammatory mediators [152155] [142]. Repression of DC maturation/function can be achieved via IL-6-STAT3 or IL-10-STAT3-mediated signaling directly, or indirectly through inhibitory molecules that induce IL-6 [152,156,157]. Blockade of STAT3 reverses many of these immunosuppressive responses, which may have particularly important consequences in rewiring tolerogenic tumor microenvironments for improved tumor clearance [151,158160].

STAT3 anti-inflammatory signaling in phagocytes

STAT3 has a key role in suppressing signal transduction mediated by Toll-like receptors (TLRs), most notably TRL4, TLR2 and TLR9, in mature phagocytic cells [69,142]. For example, Stat3-deficient macrophages, neutrophils and DCs produce elevated amounts of pro-inflammatory cytokines upon TLR4 activation, including TNF-α, IL-6, IL-12, and IFN-γ. While these populations also produce excessive amounts of IL-10, they lose responsiveness to this anti-inflammatory cytokine, which inhibits TLR4-dependent pro-inflammatory cytokine production [69,142]. Mice with hematopoietic-wide Stat3-deficiency (Tek-cre Stat3f/f), DC-restricted Stat3-deficiency (Itgax-cre Stat3f/f), or Stat3-deficiency in macrophages and neutrophils (Lyz2tm1(cre)Ifo Stat3f/f, also known as Lyz2-cre Stat3f/f or LysM cre Stat3f/f) have increased amounts of circulating pro-inflammatory cytokines and develop mild to severe enterocolitis in early adulthood [69, 92, 93,142]. Intestinal inflammation in these Stat3-deficient strains is accompanied by enhanced activity of IL-12-dependent T helper 1 (Th1) cells, as evidenced by elevated Th1-mediated IFN-γ production [69, 142].

Significantly, enterocolitis was improved in Lyz2 Stat3f/f mice upon concomitant deletion of Tlr4 or Il12b (encoding IL-12p40), but not Stat1 or Tnfa [161]. Treatment with IL-12p40 neutralizing antibodies or simultaneous Rag1-deficiency also suppresses intestinal inflammation [93,162]. These results indicate critical roles for IL-12 as well as innate and adaptive immune populations in mediating inflammatory disease. Furthermore, Il10-deficient animals develop a chronic intestinal inflammatory disease similar to that observed in the hematopoietic-wide, DC- or myeloid-restrictedStat3-deficientanimals [163]. Taken together, these data underscore the key protective nature of IL-10 and STAT3 signaling in the intestine, and imply persistent stimulation from the intestinal microbiota induces excessive cytokine production in intestinal phagocytes via unchecked TLR4 signaling, resulting in an IL-12-dependent Th1-mediated inflammatory disease [93,161,162]. This model is consistent with studies demonstrating a critical role for phagocytic-specific TLR-MyD88 signaling in driving intestinal inflammation when unopposed by IL-10 [164]. Interestingly, aged Tlr4-null Lyz2 Stat3f/f mice show evidence of intestinal inflammatory disease, suggesting excessive signaling via other TLRs (e.g., TLR9) contributes to pathology [161].

The critical role for STAT3 in mediating the anti-inflammatory effects of IL-10 was firmly established by several groups [165168], yet the cell intrinsic mechanism whereby STAT3 restrains pro-inflammatory gene expression has been elusive for nearly a decade. Early studies indicated regulation at the transcriptional level [169]. Pro-inflammatory genes are targets of NF-κB and MAPK signaling cascades and, in some cases, also regulated by IRF3/7; however, there is little evidence for direct STAT3 interference at the wide array of pro-inflammatory gene promoters. By contrast, STAT3 was found to induce expression of transcriptional repressors and co-repressors that inhibit NF-κB gene reporters [170], suggesting an indirect mechanism by which STAT3 restrains pro-inflammatory gene transcription. Other potential anti-inflammatory effectors have been identified [171177]. Nonetheless, the impact of these pathways on the broad STAT3 anti-inflammatory effect remains to be tested with genetically-modified mouse models and in vivo studies.

Key insight into the cell intrinsic STAT3 anti-inflammatory mechanism was obtained recently by studies in Tek-cre Stat3f/f mice [178]. In addition to developing severe intestinal inflammation, these animals have decreased calcified bone and elevated amounts of osteoclasts. Furthermore, Stat3-deficientbone marrow-derived macrophages exhibit an enhanced propensity to develop osteoclasts upon Receptor Activator of NF-κB (RANK) stimulation ex vivo. These data collectively suggest Stat3-deficient macrophages/osteoclast precursors have increased RANK responsiveness. Since RANK and TLR4 utilize similar signal transduction cascades, culminating in NF-κB and MAPK activation, this prompted investigation into core signaling factors in Stat3-deficient macrophages.

STAT3 was found to inhibit expression of a key E2 ubiquitin-conjugating enzyme, Ubc13, required for RANK and TLR4 signaling. STAT3 controls Ubc13 expression by direct transcriptional repression of Ube2n, the gene encoding Ubc13 [178]. Significantly, the accumulation of Ubc13 in Stat3-deficient macrophages has a central, non-redundant role in mediating enhanced RANK- and TLR4 signaling in the absence of STAT3. Therefore, taken together, these data indicate STAT3 exerts a broad suppressive function upon NF-κB and MAPK activity in macrophages by restraining Ubc13 abundance through Ube2n transcriptional inhibition (Figure 4) [178]. These results are consistent with the global repression of macrophage proinflammatory gene expression mediated by STAT3 [169]. Furthermore, they suggest excessive STAT3 activity may aberrantly induce anti-inflammatory responses. In agreement, IL-6 was shown to stimulate an atypical anti-inflammatory response in Socs3-deficient cells, which show unrestrained and prolonged STAT3 activation [179,180]. While the discovery of the STAT3-Ubc13 pathway reveals a key cell intrinsic anti-inflammatory mechanism in macrophages, additional work is needed to examine the role of this pathway in cytokine-mediated anti-inflammatory signaling in vivo.

Figure 4. Mechanism of the STAT3 anti-inflammatory response.

Figure 4

Activation of STAT3 by IL-6R or IL-10R (not shown) inhibits expression of Ube2n/Ubc13, thereby restraining TLR4 signal transduction via NF-κB and MAP kinase (not shown) cascades.

Significantly, STAT3 anti-inflammatory function appears to be conserved between mice and humans. Elevated basal and TLR4-responsive expression of pro-inflammatory cytokines was found in peripheral blood neutrophils and mononuclear cells from individuals with STAT3 AD-HIES [63], suggesting human STAT3 restrains pro-inflammatory gene expression. Moreover, the STAT3 binding site within the Ube2n/UBE2N promoter is highly conserved, which suggests potential for a STAT3-Ubc13 anti-inflammatory mechanism in humans [178]. Disordered inflammation is a prominent characteristic of STAT3 AD-HIES, consistent with elevated pro-inflammatory signaling and severely impaired IL-10 responses [181,182]. In addition, individuals with STAT3 AD-HIES exhibit skeletal abnormalities and propensity for bone fractures with mild trauma, suggesting STAT3 may regulate bone homeostasis in humans via Ubc13 restraint, similar to mice [67,178,183]. Nonetheless, whether the STAT3-Ubc13 pathway is key to human anti-inflammatory responses requires further examination.

STAT3 anti-inflammatory signaling in non-immune populations

A handful of evidence implicates anti-inflammatory roles for STAT3 in non-immune populations, suggesting this function of STAT3 may operate in numerous tissues in homeostasis and/or disease. For example, STAT3 signaling within endothelial cells, tumor cells (e.g., melanoma) or fibroblasts suppresses production of proinflammatory factors [148,184,185]. Whether this inhibition occurs via Ubc13 restraint remains to be established, although Ubc13 is broadly expressed and further increased in Stat3-deficient fibroblasts, suggesting the potential for a parallel STAT3-Ubc13 anti-inflammatory mechanism in non-immune cells [178]. Moreover, STAT3 mediates an additional protective role in the intestine by regulating epithelial cell homeostasis, mucosal wound healing and mucus production during experimental colitis. This mechanism involves secretion of IL-22 by innate immune cells and subsequent IL-22-mediated STAT3 signaling within intestinal epithelial cells [186188], indicating beneficial crosstalk between immune and non-immune cells involving STAT3.

STAT3 regulation of adaptive immunity

B lymphocytes

B cells serve multiple important roles in immunity, including immunoglobulin production, antigen presentation and T lymphocyte helper functions. Deletion of Stat3 affects numerous B cell activities. Stat3 removal using the interferon-inducible Mx-cre transgene led to fewer B cells in bone marrow and peripheral tissues versus control animals. These studies showed STAT3 is required for developmental transition of the pre-pro-B cell progenitor to subsequent precursor populations, as well as precursor survival (Figure 5) [189]. This phenotype may reflect the dependence of early B cell development on Flt3L, which induces STAT3 signaling in Flt3+ hematopoietic progenitor cells including common lymphocyte progenitors (CLPs) [134,135]. Moreover, IL-7-responsive progenitors were reduced in the absence of STAT3 without effects upon IL-7 receptor (IL-7R) signaling [189], consistent with an upstream defect in progenitor development (e.g., via a Flt3L-STAT3-dependent stage) and the primary use of STAT5 by the IL-7R.

Figure 5. STAT3 roles in adaptive immunity.

Figure 5

Key functions for STAT3 in precursor B cell generation from CLPs, production of IgG-secreting B cells, and generation of Th17, Tfh and CD8+ memory T cells are indicated by orange arrows. Also shown, STAT3 inhibitory activity on Treg generation (blunt-ended orange line). Non-STAT3 functions are indicated by thin black lines.

By contrast, STAT3 deletion from later stage B lineage-committed CD19+ precursors using Cd19-cre Stat3f/f mice (i.e., deleted at pro-B, pre-B and subsequent developmental stages) demonstrated a critical role for STAT3 in the differentiation of immunoglobulin G (IgG) secreting plasma cells [190]. This process requires T lymphocytes producing IL-21, which elicits STAT3 (and STAT1) signaling in B cells (Figure 5). IL-21-induced STAT3 stimulates expression of the B cell maturation factor Blimp-1 and thereby drives IgG-producing plasma cell generation [191194]. Molecular studies indicate STAT3 binds the Prdm1/Blimp1 locus in B cells, displacing the negative regulator BCL6; moreover, this transcriptional mechanism is enhanced upon CD40L stimulation [193]. STAT3 may also have a role in IL-35-mediated induction of IL-10- and IL-35-secreting B regulatory cells (Bregs), due to its responsiveness to IL-35 receptor signaling (i.e., via IL-12Rβ2 and IL-27Rα subunits) [195]. Bregs are implicated in suppression of host defense, autoimmunity and anti-tumor responses [195]. Thus, data to date collectively imply functions for STAT3 in both activation and repression of B cell-mediated adaptive immunity, mirroring stimulatory and inhibitory roles for STAT3 in innate immunity.

Significantly, studies of STAT3 AD-HIES show STAT3 regulates human B effector and memory populations, including IL-21-dependent plasma cells [196, 197]. The humoral response in STAT3 AD-HIES maybe further compromised by defective T follicular helper (Tfh) cell generation, as Tfh production also requires STAT3 (discussed further below) [198,199]. It is critical now to understand how STAT3 control of IL-35 responses and Breg functions factor into the STAT3 AD-HIES disease phenotype. In addition, the unexpected finding that STAT3 GOF mutations lead to fewer class-switched memory B lymphocytes and reduced circulating IgG in individuals with associated autoimmunity implies as yetunrevealed immunological mechanisms, which must be explored further.

CD4+ T lymphocytes

In response to antigen stimulation via the T cell receptor (TCR) and specific cytokine cues, naïve CD4+ T cells develop into distinct effector subsets with unique immune functions including CD8+ T cell activation, stimulation of innate immune cells and induction of B cell responses [200]. Early evidence from mice with T cell-specific Stat3 deletion (Lck-cre Stat3f/f) indicated a crucial function for STAT3 in IL-6-mediated T cell survival, independent of Bcl2 regulation [70, 201]. Subsequently, STAT3 was found to be essential for induction of IL-17-producing (Th17) cells from naïve CD4+ precursors (Figure 5). Th17 generation is driven by IL-6- and TGF-β, as well as an autocrine IL-21 signaling cascade, upon TCR activation [202205]. STAT3 is specifically required for responses to IL-6 and IL-21. At the molecular level, STAT3 stimulates expression of the Th17 lineage-specifying factors retinoic acid receptor-related orphan receptors gamma and alpha (RORγ and RORα), which are required for Th17 development. STAT3 also upregulates IL-23 receptor (IL-23R) and IL-17 expression. IL-23R signaling enhances Th17 development in the presence of IL-23, while secreted IL-17 executes canonical effector functions of the Th17 subset [203, 205209]. Th17 cells are critical for host defense to extracellular and intracellular bacteria, as well as fungi, yet are also involved in numerous inflammatory and autoimmune diseases. These functions are mediated primarily by IL-17, which stimulates production of immune effectors such as anti-microbial peptides, chemokines and granulopoietic cytokines [208, 209]. Thus, STAT3 mediates neutrophil-driven inflammatory responses by direct effects on granulopoiesis and via regulation of the Th17 lineage.

STAT3 also controls development of Tfh cells. This population is characterized by expression of the CXCR5 chemokine receptor, localization to the B cell follicle within germinal centers of secondary lymphoid organs and IL-21 secretion. IL-21 production from Tfh cells has a key role in mediating B cell “help” in germinal centers by stimulating proliferation and antibody affinity maturation, as discussed above. Furthermore, Tfh cell generation is dependent upon IL-6- or IL-21-responsive STAT3 signaling (Figure 5), which upregulates expression of BCL6, a lineage-specifying transcriptional regulator [210, 211]. STAT3 also mediates IL-27 signaling within developing Tfh cells to stimulate IL-21 production, T cell survival and expression of Tfh phenotypic markers [212]. Therefore, STAT3 has critical roles within both T and B cell compartments that culminate in production of plasma cells and IgG secretion.

By contrast to these lineage-inducing events, STAT3 potently inhibits generation of CD4+ T regulatory cells (Treg) from naïve CD4+ precursors and suppresses expression of the Treg-specifying transcription factor Foxp3 in mature Tregs (Figure 5) [83, 213]. In the setting of graft-versus-host disease (GVHD), Stat3-deficiency promotes inducible Treg generation, restrains GVHD and improves survival [83]. These data suggest STAT3 blockade in CD4+ T cells may be useful in treating GVHD. Moreover, STAT3 and Foxp3 appear to co-regulate specific genes in differentiated Tregs, including Il10, as indicated by expression studies in Foxp3-cre Stat3f/f versus wild type Tregs [214]. In this context, STAT3 has a key antiinflammatory role by maintaining the ability of Foxp3+ Tregs to inhibit inflammatory Th17 cells [214216].

Interestingly, STAT3 and STAT5 have mutually antagonistic activity in IL-2-induced Treg and IL-6-induced Th17 generation, respectively [202]. These results are explained at least in part by unique patterns of STAT binding and activity at loci encoding the Treg lineage-specifying factor Foxp3 (Foxp3) and the Th17-specific cytokine IL-17 (Il17). For example, Foxp3 expression is stimulated directly by IL-2-activated STAT5, while inhibited by IL-6-activated STAT3 [81, 213, 217]. On the contrary, Il17 expression is driven by IL-6-stimulated STAT3, which binds multiple sites in the Il17 locus. Upon IL-2 stimulation, however, STAT5 associates with common/overlapping sites, resulting in decreased STAT3 association. STAT5 binding in the Il17 locus is accompanied by a reduction in chromatin marks that reflect active transcription and lower Il17 mRNA expression [217]. These data imply divergent roles for STAT3 and STAT5 in transcriptional activation or repression of Il17, respectively. Collectively, the opposing functions for STAT3 and STAT5 at the transcriptional and epigenetic levels induce distinct developmental outcomes from naïve CD4+ T cells, contributing essentially to the diversity of CD4+ T cell subsets [200].

Importantly, STAT3 has key conserved functions in regulating human Th17 and Tfh cells; loss of these activities in STA T3 AD-HIES contributes significantly to the disease phenotype [199, 218]. Yet, unexpectedly, STAT3 LOF also reduces inducible Tregs in humans. This phenotype may be due to impaired IL-10 signaling in DCs [218]. Moreover, as discussed above, early studies of humans with STAT3 GOF-mediated diseases have revealed surprising deficits in cells that exhibit STAT3-dependency in mice [78, 80]. Since many immune responses depend on interaction of multiple cell types, often mediated by cytokines, it is imperative to dissect primary and secondary responses in STAT3 LOF and GOF-associated disease.

CD8+ T lymphocytes

Cytotoxic CD8+ T cells (CTLs) are critical for clearing cells infected with intracellular pathogens, typically viruses, as well as cells expressing aberrant host factors such as oncoproteins. The direct cytotoxic activity of CD8+ T cells in tumors is frequently associated with better prognosis and improved tumor clearance, generating significant interest in understanding how to further promote this response via conventional cancer therapy and/or immunotherapy. Naïve CD8+ T cells differentiate into potent effector cells, which in turn generate long-lived memory cells. While both CD8+ T cell effector and memory responses are regulated transcriptionally, STAT3 has critical roles in generating stable, long-lived memory cells (Figure 5) [219, 220]. STAT3 controls expression of the CD8+ T cell transcriptional regulators Eomes, BCL-6 and Blimp-1, as well as the cytokine signaling inhibitor SOCS3, as indicated by their reduction in Stat3-deficient memory CD8+ T cells. Moreover, the absence of SOCS3 is associated with CD8+ T cell hyperresponsiveness to IL-12. These data suggest STAT3-SOCS3 signaling may protect CD8+ T memory precursors from cytokine cues that regulate CD8+ T effector differentiation [219]. Upstream, the cytokines IL-10 and IL-21 play critical roles in activating STAT3 to drive formation of stable memory CD8+ T cells [219]. Recent data indicate the memory T response is enhanced by IL-10 secretion during the resolution phase of infection [221]. Importantly, individuals with STAT3 AD-HIES also demonstrate reduced amounts of central memory CD8+ T cells, as well as fewer memory CD4+ T cells, relative to healthy controls [222]. This likely contributes significantly to the impaired ability of individuals with STAT3 AD-HIES to manage certain chronic infections [222]. Furthermore, similar to their murine counterparts, human naïve T cells with STAT3-deficiency exhibit impaired proliferation and have reduced expression of BCL6 and SOCS3 [222, 223], implying highly conserved pathways in memory T cell generation.

Conclusions and future perspectives

While STAT3 functions are broadly recognized in the immune system, we lack insight into whether and how STAT3 regulates newly discovered or less abundant populations, such as innate immune lymphocytes or other granulocytic subsets. Recently, STAT3 was linked with mast cell degranulation and protection from allergic disease [224], implying additional activities to be discovered. These will be important to uncover in light of the development of STAT3 inhibitors for clinical use.

Moreover, STAT3-mediated responses in innate and adaptive immune subsets appear to have key roles in tumorigenesis, yet critical insight into cell type-specific activities is still needed. Significantly, many tumor types produce STAT3-stimulatory cytokines such as IL-6, G-CSF or vascular endothelial growth factor.

Enhanced STAT3 activation in bone marrow progenitors by the combined actions of IL-6 and G-CSF induce neutrophil generation with elevated pro-tumor responses and concomitant suppression of neutrophil functions associated with anti-tumor activity [225], indicating critical tumor-immune crosstalk. Lastly, it is important to highlight results that indicate persistently activated STAT3 within cancer cells is associated with malignancy. Chronic STAT3 activity is induced via several mechanisms, including tumor-specific STAT3 GOF mutations, hyperactivation due to kinase activities (e.g. KRAS) and local cytokine production. Therefore, modulating STAT3 activity to a level that supports immunity but prevents disease-promoting mechanisms may prove to be essential to the success of STAT3 inhibitors in the clinic.

Highlights.

  • STAT3 discovery, structure and transcriptional function

  • Non-transcriptional activities of STAT3

  • STAT3 mutations associate with human immune disorders

  • Conditional Stat3 mouse models reveal important immune functions

  • Roles for STAT3 in innate immunity

  • Roles for STAT3 in adaptive immunity

  • Conclusions and future perspectives

Acknowledgments

The authors are supported by grants from the NIH NIAID (R01AI109294, SSW) and the MD Anderson Center for Inflammation and Cancer (SSW, HL, HZ).

Abbreviations

APRF

acute phase response factor

BAC

bacterial artificial chromosome

bHLH

basic helix-loop-helix

Breg

B regulatory cells

CDPs

common DC progenitors

CLPs

common lymphocyte progenitors

CMP

common myeloid progenitor

CTLs

cytotoxic CD8+ T lymphocytes

DAMPs

damage-associated molecular patterns

eIF2a

eukaryotic translation initiation factor 2a

EIF2AK2/PKR

eIF2a kinase 2/protein kinase R

ES cells

embryonic stem cells

DC

dendritic cell

Flt3

Fms-related tyrosine kinase 3

Flt3L

Flt3 ligand

G-CSF

granulocyte colony-stimulating factor

G-CSFR

G-CSFR receptor

GAS

IFN-γ-activated sequence element

GOF

gain-of-function

GMP

granulocyte-monocyte progenitor

GVHD

graft-versus-host disease

HIES

hyper IgE syndrome

IFN

interferon

IL

interleukin

IL-6

interleukin-6

IL-7

interleukin-7

IL-7R

IL-7 receptor

IL-23

interleukin-23

IL-23R

IL-23 receptor

IgE

immunoglobulin E

IgG

immunoglobulin G

Id2

inhibitor of differentiation 2

KIR

kinase-inhibitory region

LOF

loss-of-function

MAPK

mitogen-activated protein kinase

NF-κB

nuclear factor kappa-light-chain-enhancer of activated B cells

NK

natural killer cell

PAMPs

pathogen-associated molecular patterns

pDCs

plasmactyoid dendritic cells

PD-L1

programmed death ligand-1

PGE2

prostaglandin E2

PI3K

phosphoinositol 3-kinase

RANK

Receptor Activator of NF-κB

RORa

retinoic acid receptor-related orphan receptor alpha

RORy

retinoic acid receptor-related orphan receptor gamma

ROS

reactive oxygen species

SH2

Src homology 2

SNPs

single nucleotide polymorphisms

STAT

Signal Transducers and Activators of Transcription

STAT3 AD-HIES

HIES associated with STAT3 mutation

Stat3f/f

Stat3flox/flow

TCR

T cell receptor

Tfh

T follicular helper

Th1

CD4+ T helper 1 lymphocytes

Th17

IL-17-producing CD4+ T lymphocytes

TLRs

Toll-like receptors

Tregs

regulatory T lymphocytes

uSTAT3

unphosphorylated STAT3

Biographies

Emily J. Hillmer received her B.A. in Biology from Carleton College in 2015. She is conducting a research internship with Dr. Watowich in the Department of Immunology at MD Anderson Cancer Center. Her studies are investigating the molecular mechanisms by which STAT3 protects hematopoietic stem cells from damaging pro-inflammatory signals.

Huiyuan Zhang received her M.D. from Beijing University of Chinese Medicine in 2000 and her Ph.D. in Immunology from the Chinese Academy of Medical Sciences & Peking Union Medical College in 2005. Dr. Zhang is an Instructor in the Department of Immunology at MD Anderson Cancer Center. Her research focuses on cytokine and STAT-mediated control of hematopoietic stem cells and myeloid lineages in homeostasis, inflammatory disease and cancer.

Haiyan S. Li received her M.D. from Beijing Medical University in 1998, her M.P.H. degree from Hadassah Medical School at The Hebrew University in 2000, and her Ph.D. in Immunology from Memorial University of Newfoundland in 2007. Dr. Li is an Instructor in the Department of Immunology at MD Anderson Cancer Center. Her research investigates the molecular regulation of dendritic cells and innate immunity. Her current studies include a focus on improving dendritic cell-based tumor vaccines.

graphic file with name nihms786860b1.gif

Stephanie S. Watowich received her B.A. in Biology from Carleton College in 1983. She performed cancer research at the University of Chicago from 1983–1985 and obtained her Ph.D. from Northwestern University in 1990. Dr. Watowich conducted postdoctoral studies at the Whitehead Institute of Biomedical Research with Dr. Harvey F. Lodish from 1990–1995, where she discovered the critical importance of erythropoietin receptor dimerization, providing a paradigm for cytokine receptor signal transduction. Dr. Watowich is Professor in Immunology and Co-Director of the Center for Inflammation and Cancer at MD Anderson. Her laboratory investigates the molecular regulation of immunity by cytokines and transcription factors, with a specific focus on the cytokine-activated STATs. Current projects are investigating STAT-mediated regulation of hematopoietic stem cells and mechanisms to enhance anti-tumor immunity.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflicts of interest: None

References

  • 1.Lutticken C, Wegenka UM, Yuan J, Buschmann J, Schindler C, Ziemiecki A, et al. Association of transcription factor APRF and protein kinase Jak1 with the interleukin-6 signal transducer gp130. Science. 1994;263:89–92. doi: 10.1126/science.8272872. [DOI] [PubMed] [Google Scholar]
  • 2.Standke GJ, Meier VS, Groner B. Mammary gland factor activated by prolactin on mammary epithelial cells and acute-phase response factor activated by interleukin-6 in liver cells share DNA binding and transactivation potential. Mol Endocrinol. 1994;8:469–477. doi: 10.1210/mend.8.4.7519723. [DOI] [PubMed] [Google Scholar]
  • 3.Zhong Z, Wen Z, Darnell JE., Jr Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. Science. 1994;264:95–98. doi: 10.1126/science.8140422. [DOI] [PubMed] [Google Scholar]
  • 4.Wegenka UM, Lutticken C, Buschmann J, Yuan J, Lottspeich F, Muller-Esterl W, et al. The interleukin-6-activated acute-phase response factor is antigenically and functionally related to members of the signal transducer and activator of transcription (STAT) family. Mol Cell Biol. 1994;14:3186–3196. doi: 10.1128/mcb.14.5.3186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Akira S, Nishio Y, Inoue M, Wang XJ, Wei S, Matsusaka T, et al. Molecular cloning of APRF, a novel IFN-stimulated gene factor 3 p91-related transcription factor involved in the gp130-mediated signaling pathway. Cell. 1994;77:63–71. doi: 10.1016/0092-8674(94)90235-6. [DOI] [PubMed] [Google Scholar]
  • 6.Raz R, Durbin JE, Levy DE. Acute phase response factor and additional members of the interferon-stimulated gene factor 3 family integrate diverse signals from cytokines, interferons, and growth factors. J Biol Chem. 1994;269:24391–24395. [PubMed] [Google Scholar]
  • 7.Ruff-Jamison S, Zhong Z, Wen Z, Chen K, Darnell JE, Jr, Cohen S. Epidermal growth factor and lipopolysaccharide activate Stat3 transcription factor in mouse liver. J Biol Chem. 1994;269:21933–21935. [PubMed] [Google Scholar]
  • 8.Tian SS, Lamb P, Seidel HM, Stein RB, Rosen J. Rapid activation of the STAT3 transcription factor by granulocyte colony-stimulating factor. Blood. 1994;84:1760–1764. [PubMed] [Google Scholar]
  • 9.Yu CL, Meyer DJ, Campbell GS, Larner AC, Carter-Su C, Schwartz J, et al. Enhanced DNA-binding activity of a Stat3-related protein in cells transformed by the Src oncoprotein. Science. 1995;269:81–83. doi: 10.1126/science.7541555. [DOI] [PubMed] [Google Scholar]
  • 10.Bromberg JF, Horvath CM, Besser D, Lathem WW, Darnell JE., Jr Stat3 activation is required for cellular transformation by v-src. Mol Cell Biol. 1998;18:2553–2558. doi: 10.1128/mcb.18.5.2553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ram PT, Horvath CM, Iyengar R. Stat3-mediated transformation of NIH-3T3 cells by the constitutively active Q205L Galphao protein. Science. 2000;287:142–144. doi: 10.1126/science.287.5450.142. [DOI] [PubMed] [Google Scholar]
  • 12.Zhang Y, Turkson J, Carter-Su C, Smithgall T, Levitzki A, Kraker A, et al. Activation of Stat3 in v-Src-transformed fibroblasts requires cooperation of Jak1 kinase activity. J Biol Chem. 2000;275:24935–24944. doi: 10.1074/jbc.M002383200. [DOI] [PubMed] [Google Scholar]
  • 13.Gough DJ, Corlett A, Schlessinger K, Wegrzyn J, Larner AC, Levy DE. Mitochondrial STAT3 supports Ras-dependent oncogenic transformation. Science. 2009;324:1713–1716. doi: 10.1126/science.1171721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gough DJ, Koetz L, Levy DE. The MEK-ERK pathway is necessary for serine phosphorylation of mitochondrial STAT3 and Ras-mediated transformation. PLoS One. 2013;8:e83395. doi: 10.1371/journal.pone.0083395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Boulton TG, Zhong Z, Wen Z, Darnell JE, Jr, Stahl N, Yancopoulos GD. STAT3 activation by cytokines utilizing gp130 and related transducers involves a secondary modification requiring an H7-sensitive kinase. Proc Natl Acad Sci USA. 1995;92:6915–6919. doi: 10.1073/pnas.92.15.6915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wen Z, Zhong Z, Darnell JE., Jr Maximal activation of transcription by Stat1 and Stat3 requires both tyrosine and serine phosphorylation. Cell. 1995;82:241–250. doi: 10.1016/0092-8674(95)90311-9. [DOI] [PubMed] [Google Scholar]
  • 17.Shen Y, Schlessinger K, Zhu X, Meffre E, Quimby F, Levy DE, et al. Essential role of STAT3 in postnatal survival and growth revealed by mice lacking STAT3 serine 727 phosphorylation. Mol Cell Biol. 2004;24:407–419. doi: 10.1128/MCB.24.1.407-419.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wen Z, Darnell JE., Jr Mapping of Stat3 serine phosphorylation to a single residue (727) and evidence that serine phosphorylation has no influence on DNA binding of Stat1 and Stat3. Nucleic Acids Res. 1997;25:2062–2067. doi: 10.1093/nar/25.11.2062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Shuai K, Stark GR, Kerr IM, Darnell JE., Jr A single phosphotyrosine residue of Stat91 required for gene activation by interferon-gamma. Science. 1993;261:1744–1746. doi: 10.1126/science.7690989. [DOI] [PubMed] [Google Scholar]
  • 20.Shuai K, Schindler C, Prezioso VR, Darnell JE., Jr Activation of transcription by IFN-gamma: tyrosine phosphorylation of a 91-kD DNA binding protein. Science. 1992;258:1808–1812. doi: 10.1126/science.1281555. [DOI] [PubMed] [Google Scholar]
  • 21.Schindler C, Shuai K, Prezioso VR, Darnell JE., Jr Interferon-dependent tyrosine phosphorylation of a latent cytoplasmic transcription factor. Science. 1992;257:809–813. doi: 10.1126/science.1496401. [DOI] [PubMed] [Google Scholar]
  • 22.Becker S, Groner B, Muller CW. Three-dimensional structure of the Stat3beta homodimer bound to DNA. Nature. 1998;394:145–151. doi: 10.1038/28101. [DOI] [PubMed] [Google Scholar]
  • 23.Heim MH, Kerr IM, Stark GR, Darnell JE., Jr Contribution of STAT SH2 groups to specific interferon signaling by the Jak-STAT pathway. Science. 1995;267:1347–1349. doi: 10.1126/science.7871432. [DOI] [PubMed] [Google Scholar]
  • 24.Horvath CM, Wen Z, Darnell JE., Jr A STAT protein domain that determines DNA sequence recognition suggests a novel DNA-binding domain. Genes Dev. 1995;9:984–994. doi: 10.1101/gad.9.8.984. [DOI] [PubMed] [Google Scholar]
  • 25.Shuai K, Horvath CM, Huang LH, Qureshi SA, Cowburn D, Darnell JE., Jr Interferon activation of the transcription factor Stat91 involves dimerization through SH2-phosphotyrosyl peptide interactions. Cell. 1994;76:821–828. doi: 10.1016/0092-8674(94)90357-3. [DOI] [PubMed] [Google Scholar]
  • 26.Paulson M, Pisharody S, Pan L, Guadagno S, Mui AL, Levy DE. Stat protein transactivation domains recruit p300/CBP through widely divergent sequences. J Biol Chem. 1999;274:25343–25349. doi: 10.1074/jbc.274.36.25343. [DOI] [PubMed] [Google Scholar]
  • 27.Vinkemeier U, Moarefi I, Darnell JE, Jr, Kuriyan J. Structure of the amino-terminal protein interaction domain of STAT-4. Science. 1998;279:1048–1052. doi: 10.1126/science.279.5353.1048. [DOI] [PubMed] [Google Scholar]
  • 28.Chen X, Vinkemeier U, Zhao Y, Jeruzalmi D, Darnell JE, Jr, Kuriyan J. Crystal structure of a tyrosine phosphorylated STAT-1 dimer bound to DNA. Cell. 1998;93:827–839. doi: 10.1016/s0092-8674(00)81443-9. [DOI] [PubMed] [Google Scholar]
  • 29.Zhang T, Kee WH, Seow KT, Fung W, Cao X. The coiled-coil domain of Stat3 is essential for its SH2 domain-mediated receptor binding and subsequent activation induced by epidermal growth factor and interleukin-6. Mol Cell Biol. 2000;20:7132–7139. doi: 10.1128/mcb.20.19.7132-7139.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ota N, Brett TJ, Murphy TL, Fremont DH, Murphy KM. N-domain-dependent nonphosphorylated STAT4 dimers required for cytokine-driven activation. Nat Immunol. 2004;5:208–215. doi: 10.1038/ni1032. [DOI] [PubMed] [Google Scholar]
  • 31.Hou T, Ray S, Lee C, Brasier AR. The STAT3 NH2-terminal domain stabilizes enhanceosome assembly by interacting with the p300 bromodomain. J Biol Chem. 2008;283:30725–30734. doi: 10.1074/jbc.M805941200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Yuan ZL, Guan YJ, Chatterjee D, Chin YE. Stat3 dimerization regulated by reversible acetylation of a single lysine residue. Science. 2005;307:269–273. doi: 10.1126/science.1105166. [DOI] [PubMed] [Google Scholar]
  • 33.Yang J, Huang J, Dasgupta M, Sears N, Miyagi M, Wang B, et al. Reversible methylation of promoter-bound STAT3 by histone-modifying enzymes. Proc Natl Acad Sci USA. 2010;107:21499–21504. doi: 10.1073/pnas.1016147107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Dasgupta M, Unal H, Willard B, Yang J, Karnik SS, Stark GR. Critical role for lysine 685 in gene expression mediated by transcription factor unphosphorylated STAT3. J Biol Chem. 2014;289:30763–30771. doi: 10.1074/jbc.M114.603894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Dasgupta M, Dermawan JK, Willard B, Stark GR. STAT3-driven transcription depends upon the dimethylation of K49 by EZH2. Proc Natl Acad Sci USA. 2015;112:3985–3990. doi: 10.1073/pnas.1503152112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Bromberg JF, Wrzeszczynska MH, Devgan G, Zhao Y, Pestell RG, Albanese C, et al. Stat3 as an oncogene. Cell. 1999;98:295–303. doi: 10.1016/s0092-8674(00)81959-5. [DOI] [PubMed] [Google Scholar]
  • 37.Sadowski HB, Shuai K, Darnell JE, Jr, Gilman MZ. A common nuclear signal transduction pathway activated by growth factor and cytokine receptors. Science. 1993;261:1739–1744. doi: 10.1126/science.8397445. [DOI] [PubMed] [Google Scholar]
  • 38.Darnell JE, Jr, Kerr IM, Stark GR. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science. 1994;264:1415–1421. doi: 10.1126/science.8197455. [DOI] [PubMed] [Google Scholar]
  • 39.Schindler C, Darnell JE., Jr Transcriptional responses to polypeptide ligands: the JAK-STAT pathway. Annu Rev Biochem. 1995;64:621–651. doi: 10.1146/annurev.bi.64.070195.003201. [DOI] [PubMed] [Google Scholar]
  • 40.Darnell JE., Jr STATs and gene regulation. Science. 1997;277:1630–1635. doi: 10.1126/science.277.5332.1630. [DOI] [PubMed] [Google Scholar]
  • 41.Levy DE, Darnell JE., Jr Stats: transcriptional control and biological impact. Nat Rev Mol Cell Biol. 2002;3:651–662. doi: 10.1038/nrm909. [DOI] [PubMed] [Google Scholar]
  • 42.Stark GR. How cells respond to interferons revisited: from early history to current complexity. Cytokine Growth Factor Rev. 2007;18:419–423. doi: 10.1016/j.cytogfr.2007.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Yang J, Chatterjee-Kishore M, Staugaitis SM, Nguyen H, Schlessinger K, Levy DE, et al. Novel roles of unphosphorylated STAT3 in oncogenesis and transcriptional regulation. Cancer Res. 2005;65:939–947. [PubMed] [Google Scholar]
  • 44.Yang J, Liao X, Agarwal MK, Barnes L, Auron PE, Stark GR. Unphosphorylated STAT3 accumulates in response to IL-6 and activates transcription by binding to NFkappaB. Genes Dev. 2007;21:1396–1408. doi: 10.1101/gad.1553707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Yang J, Stark GR. Roles of unphosphorylated STATs in signaling. Cell Res. 2008;18:443–451. doi: 10.1038/cr.2008.41. [DOI] [PubMed] [Google Scholar]
  • 46.Timofeeva OA, Chasovskikh S, Lonskaya I, Tarasova NI, Khavrutskii L, Tarasov SG, et al. Mechanisms of unphosphorylated STAT3 transcription factor binding to DNA. J Biol Chem. 2012;287:14192–14200. doi: 10.1074/jbc.M111.323899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Narimatsu M, Maeda H, Itoh S, Atsumi T, Ohtani T, Nishida K, et al. Tissue-specific autoregulation of the stat3 gene and its role in interleukin-6-induced survival signals in T cells. Mol Cell Biol. 2001;21:6615–6625. doi: 10.1128/MCB.21.19.6615-6625.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wegrzyn J, Potla R, Chwae YJ, Sepuri NB, Zhang Q, Koeck T, et al. Function of mitochondrial Stat3 in cellular respiration. Science. 2009;323:793–797. doi: 10.1126/science.1164551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Garama DJ, Harris TJ, White CL, Rossello FJ, Abdul-Hay M, Gough DJ, et al. A Synthetic Lethal Interaction between Glutathione Synthesis and Mitochondrial Reactive Oxygen Species Provides a Tumor-Specific Vulnerability Dependent on STAT3. Mol Cell Biol. 2015;35:3646–3656. doi: 10.1128/MCB.00541-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Carbognin E, Betto RM, Soriano ME, Smith AG, Martello G. Stat3 promotes mitochondrial transcription and oxidative respiration during maintenance and induction of naive pluripotency. Embo j. 2016 doi: 10.15252/embj.201592629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lipinski MM, Hoffman G, Ng A, Zhou W, Py BF, Hsu E, et al. A genome-wide siRNA screen reveals multiple mTORC1 independent signaling pathways regulating autophagy under normal nutritional conditions. DevCell. 2010;18:1041–1052. doi: 10.1016/j.devcel.2010.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Shen S, Niso-Santano M, Adjemian S, Takehara T, Malik SA, Minoux H, et al. Cytoplasmic STAT3 represses autophagy by inhibiting PKR activity. Mol Cell. 2012;48:667–680. doi: 10.1016/j.molcel.2012.09.013. [DOI] [PubMed] [Google Scholar]
  • 53.Niso-Santano M, Shen S, Adjemian S, Malik SA, Marino G, Lachkar S, et al. Direct interaction between STAT3 and EIF2AK2 controls fatty acid-induced autophagy. Autophagy. 2013;9:415–417. doi: 10.4161/auto.22910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Der CJ, Krontiris TG, Cooper GM. Transforming genes of human bladder and lung carcinoma cell lines are homologous to the ras genes of Harvey and Kirsten sarcoma viruses. Proc Natl Acad Sci USA. 1982;79:3637–3640. doi: 10.1073/pnas.79.11.3637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Parada LF, Tabin CJ, Shih C, Weinberg RA. Human EJ bladder carcinoma oncogene is homologue of Harvey sarcoma virus ras gene. Nature. 1982;297:474–478. doi: 10.1038/297474a0. [DOI] [PubMed] [Google Scholar]
  • 56.Santos E, Tronick SR, Aaronson SA, Pulciani S, Barbacid M. T24 human bladder carcinoma oncogene is an activated form of the normal human homologue of BALB-and Harvey-MSV transforming genes. Nature. 1982;298:343–347. doi: 10.1038/298343a0. [DOI] [PubMed] [Google Scholar]
  • 57.Corcoran RB, Contino G, Deshpande V, Tzatsos A, Conrad C, Benes CH, et al. STAT3 plays a critical role in KRAS-induced pancreatic tumorigenesis. Cancer Res. 2011;71:5020–5029. doi: 10.1158/0008-5472.CAN-11-0908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Gough DJ, Marie IJ, Lobry C, Aifantis I, Levy DE. STAT3 supports experimental K-RasG12D-induced murine myeloproliferative neoplasms dependent on serine phosphorylation. Blood. 2014;124:2252–2261. doi: 10.1182/blood-2013-02-484196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Grabner B, Schramek D, Mueller KM, Moll HP, Svinka J, Hoffmann T, et al. Disruption of STAT3 signalling promotes KRAS-induced lung tumorigenesis. Nat Commun. 2015;6:6285. doi: 10.1038/ncomms7285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Minegishi Y, Saito M, Morio T, Watanabe K, Agematsu K, Tsuchiya S, et al. Human tyrosine kinase 2 deficiency reveals its requisite roles in multiple cytokine signals involved in innate and acquired immunity. Immunity. 2006;25:745–755. doi: 10.1016/j.immuni.2006.09.009. [DOI] [PubMed] [Google Scholar]
  • 61.Zhang Q, Davis JC, Lamborn IT, Freeman AF, Jing H, Favreau AJ, et al. Combined immunodeficiency associated with DOCK8 mutations. N Engl J Med. 2009;361:2046–2055. doi: 10.1056/NEJMoa0905506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Engelhardt KR, McGhee S, Winkler S, Sassi A, Woellner C, Lopez-Herrera G, et al. Large deletions and point mutations involving the dedicator of cytokinesis 8 (DOCK8) in the autosomal-recessive form of hyper-IgE syndrome. J Allergy Clin Immunol. 2009;124:1289–1302. doi: 10.1016/j.jaci.2009.10.038. e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Holland SM, DeLeo FR, Elloumi HZ, Hsu AP, Uzel G, Brodsky N, et al. STAT3 mutations in the hyper-IgE syndrome. N Engl J Med. 2007;357:1608–1619. doi: 10.1056/NEJMoa073687. [DOI] [PubMed] [Google Scholar]
  • 64.Minegishi Y, Saito M, Tsuchiya S, Tsuge I, Takada H, Hara T, et al. Dominant-negative mutations in the DNA-binding domain of STAT3 cause hyper-IgE syndrome. Nature. 2007;448:1058–1062. doi: 10.1038/nature06096. [DOI] [PubMed] [Google Scholar]
  • 65.Freeman AF, Holland SM. The hyper-IgE syndromes. Immunol Allergy Clin North Am. 2008;28:277–291. viii. doi: 10.1016/j.iac.2008.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Freeman AF, Domingo DL, Holland SM. Hyper IgE (Jobs) syndrome: a primary immune deficiency with oral manifestations. Oral Dis. 2009;15:2–7. doi: 10.1111/j.1601-0825.2008.01463.x. [DOI] [PubMed] [Google Scholar]
  • 67.Minegishi Y. Hyper-IgE syndrome. Curr Opin Immunol. 2009;21:487–492. doi: 10.1016/j.coi.2009.07.013. [DOI] [PubMed] [Google Scholar]
  • 68.Minegishi Y, Karasuyama H. Defects in Jak-STAT-mediated cytokine signals cause hyper-IgE syndrome: lessons from a primary immunodeficiency. Int Immunol. 2009;21:105–112. doi: 10.1093/intimm/dxn134. [DOI] [PubMed] [Google Scholar]
  • 69.Takeda K, Clausen BE, Kaisho T, Tsujimura T, Terada N, Forster I, et al. Enhanced Th1 activity and development of chronic enterocolitis in mice devoid of Stat3 in macrophages and neutrophils. Immunity. 1999;10:39–49. doi: 10.1016/s1074-7613(00)80005-9. [DOI] [PubMed] [Google Scholar]
  • 70.Takeda K, Kaisho T, Yoshida N, Takeda J, Kishimoto T, Akira S. Stat3 activation is responsible for IL-6-dependent T cell proliferation through preventing apoptosis: generation and characterization of T cell-specific Stat3-deficient mice. J Immunol. 1998;161:4652–4660. [PubMed] [Google Scholar]
  • 71.Minegishi Y, Saito M, Nagasawa M, Takada H, Hara T, Tsuchiya S, et al. Molecular explanation for the contradiction between systemic Th17 defect and localized bacterial infection in hyper-IgE syndrome. J Exp Med. 2009;206:1291–1301. doi: 10.1084/jem.20082767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Steward-Tharp SM, Laurence A, Kanno Y, Kotlyar A, Villarino AV, Sciume G, et al. A mouse model of HIES reveals pro- and anti-inflammatory functions of STAT3. Blood. 2014;123:2978–2987. doi: 10.1182/blood-2013-09-523167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Barrett JC, Hansoul S, Nicolae DL, Cho JH, Duerr RH, Rioux JD, et al. Genome-wide association defines more than 30 distinct susceptibility loci for Crohns disease. Nat Genet. 2008;40:955–962. doi: 10.1038/NG.175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Cho JH. The genetics and immunopathogenesis of inflammatory bowel disease. Nat Rev Immunol. 2008;8:458–466. doi: 10.1038/nri2340. [DOI] [PubMed] [Google Scholar]
  • 75.Jakkula E, Leppa V, Sulonen AM, Varilo T, Kallio S, Kemppinen A, et al. Genome-wide association study in a high-risk isolate for multiple sclerosis reveals associated variants in STAT3 gene. Am J Hum Genet. 2010;86:285–291. doi: 10.1016/j.ajhg.2010.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Tsoi LC, Spain SL, Knight J, Ellinghaus E, Stuart PE, Capon F, et al. Identification of 15 new psoriasis susceptibility loci highlights the role of innate immunity. Nat Genet. 2012;44:1341–1348. doi: 10.1038/ng.2467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Flanagan SE, Haapaniemi E, Russell MA, Caswell R, Lango Allen H, De Franco E, et al. Activating germline mutations in STAT3 cause early-onset multi-organ autoimmune disease. Nat Genet. 2014;46:812–814. doi: 10.1038/ng.3040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Haapaniemi EM, Kaustio M, Rajala HL, van Adrichem AJ, Kainulainen L, Glumoff V, et al. Autoimmunity, hypogammaglobulinemia, lymphoproliferation, and mycobacterial disease in patients with activating mutations in STAT3. Blood. 2015;125:639–648. doi: 10.1182/blood-2014-04-570101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Milner JD, Vogel TP, Forbes L, Ma CA, Stray-Pedersen A, Niemela JE, et al. Early-onset lymphoproliferation and autoimmunity caused by germline STAT3 gain-of-function mutations. Blood. 2015;125:591–599. doi: 10.1182/blood-2014-09-602763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Vogel TP, Milner JD, Cooper MA. The Ying and Yang of STAT3 in Human Disease. J Clin Immunol. 2015;35:615–623. doi: 10.1007/s10875-015-0187-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Yao Z, Kanno Y, Kerenyi M, Stephens G, Durant L, Watford WT, et al. Nonredundant roles for Stat5a/b in directly regulating Foxp3. Blood. 2007;109:4368–4375. doi: 10.1182/blood-2006-11-055756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Xu L, Kitani A, Stuelten C, McGrady G, Fuss I, Strober W. Positive and negative transcriptional regulation of the Foxp3 gene is mediated by access and binding of the Smad3 protein to enhancer I. Immunity. 2010;33:313–325. doi: 10.1016/j.immuni.2010.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Laurence A, Amarnath S, Mariotti J, Kim YC, Foley J, Eckhaus M, et al. STAT3 transcription factor promotes instability of nTreg cells and limits generation of iTreg cells during acute murine graft-versus-host disease. Immunity. 2012;37:209–222. doi: 10.1016/j.immuni.2012.05.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Brunkow ME, Jeffery EW, Hjerrild KA, Paeper B, Clark LB, Yasayko SA, et al. Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet. 2001;27:68–73. doi: 10.1038/83784. [DOI] [PubMed] [Google Scholar]
  • 85.Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+regulatory T cells. Nat Immunol. 2003;4:330–336. doi: 10.1038/ni904. [DOI] [PubMed] [Google Scholar]
  • 86.Kim JM, Rasmussen JP, Rudensky AY. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat Immunol. 2007;8:191–197. doi: 10.1038/ni1428. [DOI] [PubMed] [Google Scholar]
  • 87.Lahl K, Loddenkemper C, Drouin C, Freyer J, Arnason J, Eberl G, et al. Selective depletion of Foxp3+ regulatory T cells induces a scurfy-like disease. J Exp Med. 2007;204:57–63. doi: 10.1084/jem.20061852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kim J, Lahl K, Hori S, Loddenkemper C, Chaudhry A, deRoos P, et al. Cutting edge: depletion of Foxp3+ cells leads to induction of autoimmunity by specific ablation of regulatory T cells in genetically targeted mice. J Immunol. 2009;183:7631–7634. doi: 10.4049/jimmunol.0804308. [DOI] [PubMed] [Google Scholar]
  • 89.Takeda K, Noguchi K, Shi W, Tanaka T, Matsumoto M, Yoshida N, et al. Targeted disruption of the mouse Stat3 gene leads to early embryonic lethality. Proc Natl Acad Sci USA. 1997;94:3801–3804. doi: 10.1073/pnas.94.8.3801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Raz R, Lee CK, Cannizzaro LA, dEustachio P, Levy DE. Essential role of STAT3 for embryonic stem cell pluripotency. Proc Natl Acad Sci USA. 1999;96:2846–2851. doi: 10.1073/pnas.96.6.2846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Lee CK, Raz R, Gimeno R, Gertner R, Wistinghausen B, Takeshita K, et al. STAT3 is a negative regulator of granulopoiesis but is not required for G-CSF-dependent differentiation. Immunity. 2002;17:63–72. doi: 10.1016/s1074-7613(02)00336-9. [DOI] [PubMed] [Google Scholar]
  • 92.Welte T, Zhang SS, Wang T, Zhang Z, Hesslein DG, Yin Z, et al. STAT3 deletion during hematopoiesis causes Crohns disease-like pathogenesis and lethality: a critical role of STAT3 in innate immunity. Proc Natl Acad Sci USA. 2003;100:1879–1884. doi: 10.1073/pnas.0237137100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Alonzi T, Newton IP, Bryce PJ, Di Carlo E, Lattanzio G, Tripodi M, et al. Induced somatic inactivation of STAT3 in mice triggers the development of a fulminant form of enterocolitis. Cytokine. 2004;26:45–56. doi: 10.1016/j.cyto.2003.12.002. [DOI] [PubMed] [Google Scholar]
  • 94.Panopoulos AD, Zhang L, Snow JW, Jones DM, Smith AM, El Kasmi KC, et al. STAT3 governs distinct pathways in emergency granulopoiesis and mature neutrophils. Blood. 2006;108:3682–3690. doi: 10.1182/blood-2006-02-003012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Akira S. Roles of STAT3 defined by tissue-specific gene targeting. Oncogene. 2000;19:2607–2611. doi: 10.1038/sj.onc.1203478. [DOI] [PubMed] [Google Scholar]
  • 96.Manz MG, Boettcher S. Emergency granulopoiesis. Nat Rev Immunol. 2014;14:302–314. doi: 10.1038/nri3660. [DOI] [PubMed] [Google Scholar]
  • 97.Cheers C, Haigh AM, Kelso A, Metcalf D, Stanley ER, Young AM. Production of colony-stimulating factors (CSFs) during infection: separate determinations of macrophage-, granulocyte-, granulocyte-macrophage-, and multi-CSFs. Infect Immun. 1988;56:247–251. doi: 10.1128/iai.56.1.247-251.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Lieschke GJ, Grail D, Hodgson G, Metcalf D, Stanley E, Cheers C, et al. Mice lacking granulocyte colony-stimulating factor have chronic neutropenia, granulocyte and macrophage progenitor cell deficiency, and impaired neutrophil mobilization. Blood. 1994;84:1737–1746. [PubMed] [Google Scholar]
  • 99.Boettcher S, Gerosa RC, Radpour R, Bauer J, Ampenberger F, Heikenwalder M, et al. Endothelial cells translate pathogen signals into G-CSF-driven emergency granulopoiesis. Blood. 2014;124:1393–1403. doi: 10.1182/blood-2014-04-570762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Panopoulos AD, Watowich SS. Granulocyte colony-stimulating factor: molecular mechanisms of action during steady state and emergency hematopoiesis. Cytokine. 2008;42:277–288. doi: 10.1016/j.cyto.2008.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.McLemore ML, Grewal S, Liu F, Archambault A, Poursine-Laurent J, Haug J, et al. STAT-3 activation is required for normal G-CSF-dependent proliferation and granulocytic differentiation. Immunity. 2001;14:193–204. doi: 10.1016/s1074-7613(01)00101-7. [DOI] [PubMed] [Google Scholar]
  • 102.Laouar Y, Welte T, Fu XY, Flavell RA. STAT3 is required for Flt3L-dependent dendritic cell differentiation. Immunity. 2003;19:903–912. doi: 10.1016/s1074-7613(03)00332-7. [DOI] [PubMed] [Google Scholar]
  • 103.Zhang H, Nguyen-Jackson H, Panopoulos AD, Li HS, Murray PJ, Watowich SS. STAT3 controls myeloid progenitor growth during emergency granulopoiesis. Blood. 2010;116:2462–2471. doi: 10.1182/blood-2009-12-259630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Nguyen-Jackson H, Panopoulos AD, Zhang H, Li HS, Watowich SS. STAT3 controls the neutrophil migratory response to CXCR2 ligands by direct activation of G-CSF-induced CXCR2 expression and via modulation of CXCR2 signal transduction. Blood. 2010;115:3354–3363. doi: 10.1182/blood-2009-08-240317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Mantel C, Messina-Graham S, Moh A, Cooper S, Hangoc G, Fu XY, et al. Mouse hematopoietic cell-targeted STAT3 deletion: stem/progenitor cell defects, mitochondrial dysfunction, ROS overproduction, and a rapid aging-like phenotype. Blood. 2012;120:2589–2599. doi: 10.1182/blood-2012-01-404004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Li HS, Yang CY, Nallaparaju KC, Zhang H, Liu YJ, Goldrath AW, et al. The signal transducers STAT5 and STAT3 control expression of Id2 and E2-2 during dendritic cell development. Blood. 2012;120:4363–4373. doi: 10.1182/blood-2012-07-441311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Johansen LM, Iwama A, Lodie TA, Sasaki K, Felsher DW, Golub TR, et al. c-Myc is a critical target for c/EBPalphain granulopoiesis. Mol Cell Biol. 2001;21:3789–3806. doi: 10.1128/MCB.21.11.3789-3806.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Hirai H, Zhang P, Dayaram T, Hetherington CJ, Mizuno S, Imanishi J, et al. C/EBPbeta is required for emergency granulopoiesis. Nat Immunol. 2006;7:732–739. doi: 10.1038/ni1354. [DOI] [PubMed] [Google Scholar]
  • 109.Nguyen-Jackson HT, Li HS, Zhang H, Ohashi E, Watowich SS. G-CSF-activated STAT3 enhances production of the chemokine MIP-2 in bone marrow neutrophils. J Leukoc Biol. 2012;92:1215–1225. doi: 10.1189/jlb.0312126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Fielding CA, McLoughlin RM, McLeod L, Colmont CS, Najdovska M, Grail D, et al. IL-6 regulates neutrophil trafficking during acute inflammation via STAT3. J Immunol. 2008;181:2189–2195. doi: 10.4049/jimmunol.181.3.2189. [DOI] [PubMed] [Google Scholar]
  • 111.Croker BA, Metcalf D, Robb L, Wei W, Mifsud S, DiRago L, et al. SOCS3 is a critical physiological negative regulator of G-CSF signaling and emergency granulopoiesis. Immunity. 2004;20:153–165. doi: 10.1016/s1074-7613(04)00022-6. [DOI] [PubMed] [Google Scholar]
  • 112.Wormald S, Hilton DJ. Inhibitors of cytokine signal transduction. J Biol Chem. 2004;279:821–824. doi: 10.1074/jbc.R300030200. [DOI] [PubMed] [Google Scholar]
  • 113.Murray PJ. The JAK-STAT signaling pathway: input and output integration. J Immunol. 2007;178:2623–2629. doi: 10.4049/jimmunol.178.5.2623. [DOI] [PubMed] [Google Scholar]
  • 114.Auernhammer CJ, Bousquet C, Melmed S. Autoregulation of pituitary corticotroph SOCS-3 expression: characterization of the murine SOCS-3 promoter. Proc Natl Acad Sci USA. 1999;96:6964–6969. doi: 10.1073/pnas.96.12.6964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Babon JJ, Kershaw NJ, Murphy JM, Varghese LN, Laktyushin A, Young SN, et al. Suppression of cytokine signaling by SOCS3: characterization of the mode of inhibition and the basis of its specificity. Immunity. 2012;36:239–250. doi: 10.1016/j.immuni.2011.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Kershaw NJ, Murphy JM, Liau NP, Varghese LN, Laktyushin A, Whitlock EL, et al. SOCS3 binds specific receptor-JAK complexes to control cytokine signaling by direct kinase inhibition. Nat Struct Mol Biol. 2013;20:469–476. doi: 10.1038/nsmb.2519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Croker BA, Mielke LA, Wormald S, Metcalf D, Kiu H, Alexander WS, et al. Socs3 maintains the specificity of biological responses to cytokine signals during granulocyte and macrophage differentiation. Exp Hematol. 2008;36:786–798. doi: 10.1016/j.exphem.2008.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Matsukawa A, Takeda K, Kudo S, Maeda T, Kagayama M, Akira S. Aberrant inflammation and lethality to septic peritonitis in mice lacking STAT3 in macrophages and neutrophils. J Immunol. 2003;171:6198–6205. doi: 10.4049/jimmunol.171.11.6198. [DOI] [PubMed] [Google Scholar]
  • 119.Hill HR, Quie PG. Raised serum-IgE levels and defective neutrophil chemotaxis in three children with eczema and recurrent bacterial infections. Lancet. 1974;1:183–187. doi: 10.1016/s0140-6736(74)92493-3. [DOI] [PubMed] [Google Scholar]
  • 120.Hill HR, Ochs HD, Quie PG, Clark RA, Pabst HF, Klebanoff SJ, et al. Defect in neutrophil granulocyte chemotaxis in Jobs syndrome of recurrent “cold” staphylococcal abscesses. Lancet. 1974;2:617–619. doi: 10.1016/s0140-6736(74)91942-4. [DOI] [PubMed] [Google Scholar]
  • 121.Mintz R, Garty BZ, Meshel T, Marcus N, Katanov C, Cohen-Hillel E, et al. Reduced expression of chemoattractant receptors by polymorphonuclear leukocytes in Hyper IgE Syndrome patients. Immunol Lett. 2010;130:97–106. doi: 10.1016/j.imlet.2009.12.006. [DOI] [PubMed] [Google Scholar]
  • 122.Mogensen TH. STAT3 and the Hyper-IgE syndrome: Clinical presentation, genetic origin, pathogenesis, novel findings and remaining uncertainties. Jakstat. 2013;2:e23435. doi: 10.4161/jkst.23435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Liu F, Poursine-Laurent J, Wu HY, Link DC. Interleukin-6 and the granulocyte colony-stimulating factor receptor are major independent regulators of granulopoiesis in vivo but are not required for lineage commitment or terminal differentiation. Blood. 1997;90:2583–2590. [PubMed] [Google Scholar]
  • 124.Liu F, Wu HY, Wesselschmidt R, Kornaga T, Link DC. Impaired production and increased apoptosis of neutrophils in granulocyte colony-stimulating factor receptor-deficient mice. Immunity. 1996;5:491–501. doi: 10.1016/s1074-7613(00)80504-x. [DOI] [PubMed] [Google Scholar]
  • 125.Richards MK, Liu F, Iwasaki H, Akashi K, Link DC. Pivotal role of granulocyte colony-stimulating factor in the development of progenitors in the common myeloid pathway. Blood. 2003;102:3562–3568. doi: 10.1182/blood-2003-02-0593. [DOI] [PubMed] [Google Scholar]
  • 126.Semerad CL, Poursine-Laurent J, Liu F, Link DC. A role for G-CSF receptor signaling in the regulation of hematopoietic cell function but not lineage commitment or differentiation. Immunity. 1999;11:153–161. doi: 10.1016/s1074-7613(00)80090-4. [DOI] [PubMed] [Google Scholar]
  • 127.Panopoulos AD, Bartos D, Zhang L, Watowich SS. Control of myeloid-specific integrin alpha Mbeta 2 (CD11b/CD 18) expression by cytokines is regulated by Stat3-dependent activation ofPU.1. J Biol Chem. 2002;277:19001–19007. doi: 10.1074/jbc.M112271200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.MacNamara KC, Oduro K, Martin O, Jones DD, McLaughlin M, Choi K, et al. Infection-induced myelopoiesis during intracellular bacterial infection is critically dependent upon IFN-gamma signaling. J Immunol. 2011;186:1032–1043. doi: 10.4049/jimmunol.1001893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.de Bruin AM, Libregts SF, Valkhof M, Boon L, Touw IP, Nolte MA. IFNgamma induces monopoiesis and inhibits neutrophil development during inflammation. Blood. 2012;119:1543–1554. doi: 10.1182/blood-2011-07-367706. [DOI] [PubMed] [Google Scholar]
  • 130.Jouanguy E, Doffinger R, Dupuis S, Pallier A, Altare F, Casanova JL. IL-12 and IFN-gamma in host defense against mycobacteria and salmonella in mice and men. Curr Opin Immunol. 1999;11:346–351. doi: 10.1016/s0952-7915(99)80055-7. [DOI] [PubMed] [Google Scholar]
  • 131.Gardner JC, Noel JG, Nikolaidis NM, Karns R, Aronow BJ, Ogle CK, et al. G-CSF drives a posttraumatic immune program that protects the host from infection. J Immunol. 2014;192:2405–2417. doi: 10.4049/jimmunol.1302752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Merad M, Manz MG. Dendritic cell homeostasis. Blood. 2009;113:3418–3427. doi: 10.1182/blood-2008-12-180646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Merad M, Sathe P, Helft J, Miller J, Mortha A. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol. 2013;31:563–604. doi: 10.1146/annurev-immunol-020711-074950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.McKenna HJ, Stocking KL, Miller RE, Brasel K, De Smedt T, Maraskovsky E, et al. Mice lacking flt3 ligand have deficient hematopoiesis affecting hematopoietic progenitor cells, dendritic cells, and natural killer cells. Blood. 2000;95:3489–3497. [PubMed] [Google Scholar]
  • 135.Esashi E, Wang YH, Perng O, Qin XF, Liu YJ, Watowich SS. The signal transducer STAT5 inhibits plasmacytoid dendritic cell development by suppressing transcription factor IRF8. Immunity. 2008;28:509–520. doi: 10.1016/j.immuni.2008.02.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Singh P, Hoggatt J, Hu P, Speth JM, Fukuda S, Breyer RM, et al. Blockade of prostaglandin E2 signaling through EP1 and EP3 receptors attenuates Flt3L-dependent dendritic cell development from hematopoietic progenitor cells. Blood. 2012;119:1671–1682. doi: 10.1182/blood-2011-03-342428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.DAmico A, Wu L. The early progenitors of mouse dendritic cells and plasmacytoid predendritic cells are within the bone marrow hemopoietic precursors expressing Flt3. J Exp Med. 2003;198:293–303. doi: 10.1084/jem.20030107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Onai N, Obata-Onai A, Tussiwand R, Lanzavecchia A, Manz MG. Activation of the Flt3 signal transduction cascade rescues and enhances type I interferon-producingand dendritic cell development. J Exp Med. 2006;203:227–238. doi: 10.1084/jem.20051645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Hegde S, Ni S, He S, Yoon D, Feng GS, Watowich SS, et al. Stat3 promotes the development of erythroleukemia by inducing Pu.1 expression and inhibiting erythroid differentiation. Oncogene. 2009;28:3349–3359. doi: 10.1038/onc.2009.202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Carotta S, Dakic A, DAmico A, Pang SHM, Greig KT, Nutt SL, et al. The transcription factor PU.1 controls dendritic cell development and Flt3 cytokine receptor expression in a dose-dependent manner. Immunity. 2010;32:628–641. doi: 10.1016/j.immuni.2010.05.005. [DOI] [PubMed] [Google Scholar]
  • 141.Watowich SS, Liu YJ. Mechanisms regulating dendritic cell specification and development. Immunol Rev. 2010;238:76–92. doi: 10.1111/j.1600-065X.2010.00949.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Melillo JA, Song L, Bhagat G, Blazquez AB, Plumlee CR, Lee C, et al. Dendritic cell (DC)-specific targeting reveals Stat3 as a negative regulator of DC function. J Immunol. 2010;184:2638–2645. doi: 10.4049/jimmunol.0902960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Cisse B, Caton ML, Lehner M, Maeda T, Scheu S, Locksley R, et al. Transcription factor E2-2 is an essential and specific regulator of plasmacytoid dendritic cell development. Cell. 2008;135:37–48. doi: 10.1016/j.cell.2008.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Hacker C, Kirsch RD, Ju XS, Hieronymus T, Gust TC, Kuhl C, et al. Transcriptional profiling identifies Id2 function in dendritic cell development. Nat Immunol. 2003;4:380–386. doi: 10.1038/ni903. [DOI] [PubMed] [Google Scholar]
  • 145.Kee BL. E and ID proteins branch out. Nat Rev Immunol. 2009;9:175–184. doi: 10.1038/nri2507. [DOI] [PubMed] [Google Scholar]
  • 146.Li HS, Watowich SS. Diversification of dendritic cell subsets: Emerging roles for STAT proteins. JAKSTAT. 2013;2:e25112. doi: 10.4161/jkst.25112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Cheng F, Wang HW, Cuenca A, Huang M, Ghansah T, Brayer J, et al. A critical role for Stat3 signaling in immune tolerance. Immunity. 2003;19:425–436. doi: 10.1016/s1074-7613(03)00232-2. [DOI] [PubMed] [Google Scholar]
  • 148.Wang T, Niu G, Kortylewski M, Burdelya L, Shain K, Zhang S, et al. Regulation of the innate and adaptive immune responses by Stat-3 signaling in tumor cells. Nat Med. 2004;10:48–54. doi: 10.1038/nm976. [DOI] [PubMed] [Google Scholar]
  • 149.Nefedova Y, Huang M, Kusmartsev S, Bhattacharya R, Cheng P, Salup R, et al. Hyperactivation of STAT3 is involved in abnormal differentiation of dendritic cells in cancer. J Immunol. 2004;172:464–474. doi: 10.4049/jimmunol.172.1.464. [DOI] [PubMed] [Google Scholar]
  • 150.Lunz JG, 3rd, Specht SM, Murase N, Isse K, Demetris AJ. Gut-derived commensal bacterial products inhibit liver dendritic cell maturation by stimulating hepatic interleukin-6/signal transducer and activator of transcription 3 activity. Hepatology. 2007;46:1946–1959. doi: 10.1002/hep.21906. [DOI] [PubMed] [Google Scholar]
  • 151.Lin A, Schildknecht A, Nguyen LT, Ohashi PS. Dendritic cells integrate signals from the tumor microenvironmentto modulate immunity and tumor growth. Immunol Lett. 2010;127:77–84. doi: 10.1016/j.imlet.2009.09.003. [DOI] [PubMed] [Google Scholar]
  • 152.Park SJ, Nakagawa T, Kitamura H, Atsumi T, Kamon H, Sawa S, et al. IL-6 regulates in vivo dendritic cell differentiation through STAT3 activation. J Immunol. 2004;173:3844–3854. doi: 10.4049/jimmunol.173.6.3844. [DOI] [PubMed] [Google Scholar]
  • 153.Kitamura H, Kamon H, Sawa S, Park SJ, Katunuma N, Ishihara K, et al. IL-6-STAT3 controls intracellular MHC class II alphabeta dimer level through cathepsin S activity in dendritic cells. Immunity. 2005;23:491–502. doi: 10.1016/j.immuni.2005.09.010. [DOI] [PubMed] [Google Scholar]
  • 154.Cheng P, Corzo CA, Luetteke N, Yu B, Nagaraj S, Bui MM, et al. Inhibition of dendritic cell differentiation and accumulation of myeloid-derived suppressor cells in cancer is regulated by S100A9 protein. J Exp Med. 2008;205:2235–2249. doi: 10.1084/jem.20080132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Wolfle SJ, Strebovsky J, Bartz H, Sahr A, Arnold C, Kaiser C, et al. PD-L1 expression on tolerogenic APCs is controlled by STAT-3. Eur J Immunol. 2011;41:413–424. doi: 10.1002/eji.201040979. [DOI] [PubMed] [Google Scholar]
  • 156.Corinti S, Albanesi C, la Sala A, Pastore S, Girolomoni G. Regulatory activity of autocrine IL-10 on dendritic cell functions. J Immunol. 2001;166:4312–4318. doi: 10.4049/jimmunol.166.7.4312. [DOI] [PubMed] [Google Scholar]
  • 157.Liang S, Ristich V, Arase H, Dausset J, Carosella ED, Horuzsko A. Modulation of dendritic cell differentiation by HLA-G and ILT4 requires the IL-6--STAT3 signaling pathway. Proc Natl Acad Sci USA. 2008;105:8357–8362. doi: 10.1073/pnas.0803341105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Nefedova Y, Nagaraj S, Rosenbauer A, Muro-Cacho C, Sebti SM, Gabrilovich DI. Regulation of dendritic cell differentiation and antitumor immune response in cancer by pharmacologic-selective inhibition of the janus-activated kinase 2/signal transducers and activators of transcription 3 pathway. Cancer Res. 2005;65:9525–9535. doi: 10.1158/0008-5472.CAN-05-0529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Bharadwaj U, Li M, Zhang R, Chen C, Yao Q. Elevated interleukin-6 and G-CSF in human pancreatic cancer cell conditioned medium suppress dendritic cell differentiation and activation. Cancer Res. 2007;67:5479–5488. doi: 10.1158/0008-5472.CAN-06-3963. [DOI] [PubMed] [Google Scholar]
  • 160.Sanseverino I, Purificato C, Varano B, Conti L, Gessani S, Gauzzi MC. STAT3-silenced human dendritic cells have an enhanced ability to prime IFNgamma production by both alphabeta and gammadelta T lymphocytes. Immunobiology. 2014;219:503–511. doi: 10.1016/j.imbio.2014.02.012. [DOI] [PubMed] [Google Scholar]
  • 161.Kobayashi M, Kweon MN, Kuwata H, Schreiber RD, Kiyono H, Takeda K, et al. Toll-like receptor-dependent production of IL-12p40 causes chronic enterocolitis in myeloid cell-specific Stat3-deficient mice. J Clin Invest. 2003;111:1297–1308. doi: 10.1172/JCI17085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Reindl W, Weiss S, Lehr HA, Forster I. Essential crosstalk between myeloid and lymphoid cells for development of chronic colitis in myeloid-specific signal transducer and activator of transcription 3-deficient mice. Immunology. 2007;120:19–27. doi: 10.1111/j.1365-2567.2006.02473.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Kuhn R, Lohler J, Rennick D, Rajewsky K, Muller W. Interleukin-10-deficient mice develop chronic enterocolitis. Cell. 1993;75:263–274. doi: 10.1016/0092-8674(93)80068-p. [DOI] [PubMed] [Google Scholar]
  • 164.Hoshi N, Schenten D, Nish SA, Walther Z, Gagliani N, Flavell RA, et al. MyD88 signalling in colonic mononuclear phagocytes drives colitis in IL-10-deficientmice. NatCommun. 2012;3:1120. doi: 10.1038/ncomms2113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Lang R, Patel D, Morris JJ, Rutschman RL, Murray PJ. Shaping gene expression in activated and resting primary macrophages by IL-10. J Immunol. 2002;169:2253–2263. doi: 10.4049/jimmunol.169.5.2253. [DOI] [PubMed] [Google Scholar]
  • 166.Williams L, Bradley L, Smith A, Foxwell B. Signal transducer and activator of transcription 3 is the dominant mediator of the anti-inflammatory effects of IL-10 in human macrophages. J Immunol. 2004;172:567–576. doi: 10.4049/jimmunol.172.1.567. [DOI] [PubMed] [Google Scholar]
  • 167.El Kasmi KC, Holst J, Coffre M, Mielke L, de Pauw A, Lhocine N, et al. General nature of the STAT3-activated anti-inflammatory response. J Immunol. 2006;177:7880–7888. doi: 10.4049/jimmunol.177.11.7880. [DOI] [PubMed] [Google Scholar]
  • 168.Williams LM, Sarma U, Willets K, Smallie T, Brennan F, Foxwell BM. Expression of constitutively active STAT3 can replicate the cytokine-suppressive activity of interleukin-10 in human primary macrophages. J Biol Chem. 2007;282:6965–6975. doi: 10.1074/jbc.M609101200. [DOI] [PubMed] [Google Scholar]
  • 169.Murray PJ. The primary mechanism of the IL-10-regulated antiinflammatory response is to selectively inhibit transcription. Proc Natl Acad Sci USA. 2005;102:8686–8691. doi: 10.1073/pnas.0500419102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.El Kasmi KC, Smith AM, Williams L, Neale G, Panopoulos AD, Watowich SS, et al. Cutting edge: A transcriptional repressor and corepressor induced by the STAT3-regulated anti-inflammatory signaling pathway. J Immunol. 2007;179:7215–7219. doi: 10.4049/jimmunol.179.11.7215. [DOI] [PubMed] [Google Scholar]
  • 171.Schaljo B, Kratochvill F, Gratz N, Sadzak I, Sauer I, Hammer M, et al. Tristetraprolin is required for full anti-inflammatory response of murine macrophages to IL-10. J Immunol. 2009;183:1197–1206. doi: 10.4049/jimmunol.0803883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Chan CS, Ming-Lum A, Golds GB, Lee SJ, Anderson RJ, Mui AL. Interleukin-10 inhibits lipopolysaccharide-induced tumor necrosis factor-alpha translation through a SHIP1-dependent pathway. J Biol Chem. 2012;287:38020–38027. doi: 10.1074/jbc.M112.348599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Gaba A, Grivennikov SI, Do MV, Stumpo DJ, Blackshear PJ, Karin M. Cutting edge: IL-10-mediated tristetraprolin induction is part of a feedback loop that controls macrophage STAT3 activation and cytokine production. J Immunol. 2012;189:2089–2093. doi: 10.4049/jimmunol.1201126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Smith AM, Qualls JE, OBrien K, Balouzian L, Johnson PF, Schultz-Cherry S, et al. A distal enhancer in Il12b is the target of transcriptional repression by the STAT3 pathway and requires the basic leucine zipper (B-ZIP) protein NFIL3. J Biol Chem. 2011;286:23582–23590. doi: 10.1074/jbc.M111.249235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Hutchins AP, Poulain S, Miranda-Saavedra D. Genome-wide analysis of STAT3 binding in vivo predicts effectors of the anti-inflammatory response in macrophages. Blood. 2012;119:e110–e119. doi: 10.1182/blood-2011-09-381483. [DOI] [PubMed] [Google Scholar]
  • 176.Curtale G, Mirolo M, Renzi TA, Rossato M, Bazzoni F, Locati M. Negative regulation of Toll-like receptor 4 signaling by IL-10-dependentmicroRNA-146b. Proc Natl Acad Sci USA. 2013;110:11499–11504. doi: 10.1073/pnas.1219852110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Hutchins AP, Diez D, Miranda-Saavedra D. The IL-10/STAT3-mediated antiinflammatory response: recent developments and future challenges. Brief Funct Genomics. 2013;12:489–498. doi: 10.1093/bfgp/elt028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Zhang H, Hu H, Greeley N, Jin J, Matthews AJ, Ohashi E, et al. STAT3 restrains RANK- and TLR4-mediated signalling by suppressing expression of the E2 ubiquitin-conjugating enzyme Ubc13. NatCommun. 2014;5:5798. doi: 10.1038/ncomms6798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Yasukawa H, Ohishi M, Mori H, Murakami M, Chinen T, Aki D, et al. IL-6 induces an anti-inflammatory response in the absence of SOCS3 in macrophages. Nat Immunol. 2003;4:551–556. doi: 10.1038/ni938. [DOI] [PubMed] [Google Scholar]
  • 180.Johnston JA, OShea JJ. Matching SOCS with function. Nat Immunol. 2003;4:507–509. doi: 10.1038/ni0603-507. [DOI] [PubMed] [Google Scholar]
  • 181.Heimall J, Freeman A, Holland SM. Pathogenesis of hyper IgE syndrome. Clin Rev Allergy Immunol. 2010;38:32–38. doi: 10.1007/s12016-009-8134-1. [DOI] [PubMed] [Google Scholar]
  • 182.Saito M, Nagasawa M, Takada H, Hara T, Tsuchiya S, Agematsu K, et al. Defective IL-10 signaling in hyper-IgE syndrome results in impaired generation of tolerogenic dendritic cells and induced regulatory T cells. J Exp Med. 2011;208:235–249. doi: 10.1084/jem.20100799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Sowerwine KJ, Shaw PA, Gu W, Ling JC, Collins MT, Darnell DN, et al. Bone density and fractures in autosomal dominant hyper IgE syndrome. J Clin Immunol. 2014;34:260–264. doi: 10.1007/s10875-013-9982-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Kano A, Wolfgang MJ, Gao Q, Jacoby J, Chai GX, Hansen W, et al. Endothelial cells require STAT3 for protection against endotoxin-induced inflammation. J Exp Med. 2003;198:1517–1525. doi: 10.1084/jem.20030077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Burdelya L, Kujawski M, Niu G, Zhong B, Wang T, Zhang S, et al. Stat3 activity in melanoma cells affects migration of immune effector cells and nitric oxide-mediated antitumor effects. J Immunol. 2005;174:3925–3931. doi: 10.4049/jimmunol.174.7.3925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Sugimoto K, Ogawa A, Mizoguchi E, Shimomura Y, Andoh A, Bhan AK, et al. IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis. J Clin Invest. 2008;118:534–544. doi: 10.1172/JCI33194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Pickert G, Neufert C, Leppkes M, Zheng Y, Wittkopf N, Warntjen M, et al. STAT3 links IL-22 signaling in intestinal epithelial cells to mucosal wound healing. J Exp Med. 2009;206:1465–1472. doi: 10.1084/jem.20082683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Takatori H, Kanno Y, Watford WT, Tato CM, Weiss G, Ivanov II, et al. Lymphoid tissue inducer-like cells are an innate source of IL-17 and IL-22. J Exp Med. 2009;206:35–41. doi: 10.1084/jem.20072713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Chou WC, Levy DE, Lee CK. STAT3 positively regulates an early step in B-cell development. Blood. 2006;108:3005–3011. doi: 10.1182/blood-2006-05-024430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Fornek JL, Tygrett LT, Waldschmidt TJ, Poli V, Rickert RC, Kansas GS. Critical role for Stat3 in T-dependent terminal differentiation of IgG B cells. Blood. 2006;107:1085–1091. doi: 10.1182/blood-2005-07-2871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Leonard WJ, Zeng R, Spolski R. Interleukin 21: a cytokine/cytokine receptor system that has come of age. J Leukoc Biol. 2008;84:348–356. doi: 10.1189/jlb.0308149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Diehl SA, Schmidlin H, Nagasawa M, Blom B, Spits H. IL-6 triggers IL-21 production by human CD4+ T cells to drive STAT3-dependent plasma cell differentiation in B cells. Immunol Cell Biol. 2012;90:802–811. doi: 10.1038/icb.2012.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Ding BB, Bi E, Chen H, Yu JJ, Ye BH. IL-21 and CD40L synergistically promote plasma cell differentiation through upregulation of Blimp-1 in human B cells. J Immunol. 2013;190:1827–1836. doi: 10.4049/jimmunol.1201678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Hunter CA, Jones SA. IL-6 as a keystone cytokine in health and disease. Nat Immunol. 2015;16:448–457. doi: 10.1038/ni.3153. [DOI] [PubMed] [Google Scholar]
  • 195.Wang RX, Yu CR, Dambuza IM, Mahdi RM, Dolinska MB, Sergeev YV, et al. Interleukin-35 induces regulatory B cells that suppress autoimmune disease. Nat Med. 2014;20:633–641. doi: 10.1038/nm.3554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Speckmann C, Enders A, Woellner C, Thiel D, Rensing-Ehl A, Schlesier M, et al. Reduced memory B cells in patients with hyper IgE syndrome. Clin Immunol. 2008;129:448–454. doi: 10.1016/j.clim.2008.08.002. [DOI] [PubMed] [Google Scholar]
  • 197.Avery DT, Deenick EK, Ma CS, Suryani S, Simpson N, Chew GY, et al. B cell-intrinsic signaling through IL-21 receptor and STAT3 is required for establishing long-lived antibody responses in humans. J Exp Med. 2010;207:155–171. doi: 10.1084/jem.20091706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Ma CS, Avery DT, Chan A, Batten M, Bustamante J, Boisson-Dupuis S, et al. Functional STAT3 deficiency compromises the generation of human T follicular helper cells. Blood. 2012;119:3997–4008. doi: 10.1182/blood-2011-11-392985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Ma CS, Wong N, Rao G, Avery DT, Torpy J, Hambridge T, et al. Monogenic mutations differentially affect the quantity and quality of T follicular helper cells in patients with human primary immunodeficiencies. J Allergy Clin Immunol. 2015;136:993–1006. doi: 10.1016/j.jaci.2015.05.036. e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Kanno Y, Vahedi G, Hirahara K, Singleton K, OShea JJ. Transcriptional and epigenetic control of T helper cell specification: molecular mechanisms underlying commitment and plasticity. Annu Rev Immunol. 2012;30:707–731. doi: 10.1146/annurev-immunol-020711-075058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Liu X, Lee YS, Yu CR, Egwuagu CE. Loss of STAT3 in CD4+ T cells prevents development of experimental autoimmune diseases. J Immunol. 2008;180:6070–6076. doi: 10.4049/jimmunol.180.9.6070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Laurence A, Tato CM, Davidson TS, Kanno Y, Chen Z, Yao Z, et al. Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation. Immunity. 2007;26:371–381. doi: 10.1016/j.immuni.2007.02.009. [DOI] [PubMed] [Google Scholar]
  • 203.Yang XO, Panopoulos AD, Nurieva R, Chang SH, Wang D, Watowich SS, et al. STAT3 regulates cytokine-mediated generation of inflammatory helper T cells. J Biol Chem. 2007;282:9358–9363. doi: 10.1074/jbc.C600321200. [DOI] [PubMed] [Google Scholar]
  • 204.Zhou L, Ivanov II, Spolski R, Min R, Shenderov K, Egawa T, et al. IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat Immunol. 2007;8:967–974. doi: 10.1038/ni1488. [DOI] [PubMed] [Google Scholar]
  • 205.Nurieva R, Yang XO, Martinez G, Zhang Y, Panopoulos AD, Ma L, et al. Essential autocrine regulation by IL-21 in the generation of inflammatory T cells. Nature. 2007;448:480–483. doi: 10.1038/nature05969. [DOI] [PubMed] [Google Scholar]
  • 206.Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, et al. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 2006;126:1121–1133. doi: 10.1016/j.cell.2006.07.035. [DOI] [PubMed] [Google Scholar]
  • 207.Yang XO, Pappu BP, Nurieva R, Akimzhanov A, Kang HS, Chung Y, et al. T helper 17 lineage differentiation is programmed by orphan nuclear receptors ROR alpha and ROR gamma. Immunity. 2008;28:29–39. doi: 10.1016/j.immuni.2007.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Gaffen SL. Structure and signalling in the IL-17 receptor family. Nat Rev Immunol. 2009;9:556–567. doi: 10.1038/nri2586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Gaffen SL. Recent advances in the IL-17 cytokine family. Curr Opin Immunol. 2011;23:613–619. doi: 10.1016/j.coi.2011.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Nurieva RI, Chung Y, Hwang D, Yang XO, Kang HS, Ma L, et al. Generation of T follicular helper cells is mediated by interleukin-21 but independent of T helper 1, 2, or 17 cell lineages. Immunity. 2008;29:138–149. doi: 10.1016/j.immuni.2008.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Liu X, Yan X, Zhong B, Nurieva RI, Wang A, Wang X, et al. Bcl6 expression specifies the T follicular helper cell program in vivo. J Exp Med. 2012;209:1841–1852. s1–s24. doi: 10.1084/jem.20120219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Batten M, Ramamoorthi N, Kljavin NM, Ma CS, Cox JH, Dengler HS, et al. IL-27 supports germinal center function by enhancing IL-21 production and the function of T follicular helper cells. J Exp Med. 2010;207:2895–2906. doi: 10.1084/jem.20100064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Yang XO, Nurieva R, Martinez GJ, Kang HS, Chung Y, Pappu BP, et al. Molecular antagonism and plasticity of regulatory and inflammatory T cell programs. Immunity. 2008;29:44–56. doi: 10.1016/j.immuni.2008.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Chaudhry A, Samstein RM, Treuting P, Liang Y, Pils MC, Heinrich JM, et al. Interleukin-10 signaling in regulatory T cells is required for suppression of Th17 cell-mediated inflammation. Immunity. 2011;34:566–578. doi: 10.1016/j.immuni.2011.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Chaudhry A, Rudra D, Treuting P, Samstein RM, Liang Y, Kas A, et al. CD4+ regulatory T cells control TH17 responses in a Stat3-dependent manner. Science. 2009;326:986–991. doi: 10.1126/science.1172702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Chaudhry A, Rudensky AY. Control of inflammation by integration of environmental cues by regulatory T cells. J Clin Invest. 2013;123:939–944. doi: 10.1172/JCI57175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Yang XP, Ghoreschi K, Steward-Tharp SM, Rodriguez-Canales J, Zhu J, Grainger JR, et al. Opposing regulation of the locus encoding IL-17 through direct, reciprocal actions of STAT3 and STAT5. Nat Immunol. 2011;12:247–254. doi: 10.1038/ni.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Minegishi Y, Saito M. Molecular mechanisms of the immunological abnormalities in hyper-IgE syndrome. Ann NY Acad Sci. 2011;1246:34–40. doi: 10.1111/j.1749-6632.2011.06280.x. [DOI] [PubMed] [Google Scholar]
  • 219.Cui W, Liu Y, Weinstein JS, Craft J, Kaech SM. An interleukin-21-interleukin-10-STAT3 pathway is critical for functional maturation of memory CD8+ T cells. Immunity. 2011;35:792–805. doi: 10.1016/j.immuni.2011.09.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Kaech SM, Cui W. Transcriptional control of effector and memory CD8+ T cell differentiation. Nat Rev Immunol. 2012;12:749–761. doi: 10.1038/nri3307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Laidlaw BJ, Cui W, Amezquita RA, Gray SM, Guan T, Lu Y, et al. Production of IL-10 by CD4(+) regulatory T cells during the resolution of infection promotes the maturation of memory CD8(+) T cells. Nat Immunol. 2015;16:871–879. doi: 10.1038/ni.3224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Siegel AM, Heimall J, Freeman AF, Hsu AP, Brittain E, Brenchley JM, et al. A critical role for STAT3 transcription factor signaling in the development and maintenance of human T cell memory. Immunity. 2011;35:806–818. doi: 10.1016/j.immuni.2011.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Ives ML, Ma CS, Palendira U, Chan A, Bustamante J, Boisson-Dupuis S, et al. Signal transducer and activator of transcription 3 (STAT3) mutations underlying autosomal dominant hyper-IgE syndrome impair human CD8(+) T-cell memory formation and function. J Allergy Clin Immunol. 2013;132:400–411. doi: 10.1016/j.jaci.2013.05.029. e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Siegel AM, Stone KD, Cruse G, Lawrence MG, Olivera A, Jung MY, et al. Diminished allergic disease in patients with STAT3 mutations reveals a role for STAT3 signaling in mast cell degranulation. J Allergy Clin Immunol. 2013;132:1388–1396. doi: 10.1016/j.jaci.2013.08.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Yan B, Wei JJ, Yuan Y, Sun R, Li D, Luo J, et al. IL-6 cooperates with G-CSF to induce protumor function of neutrophils in bone marrow by enhancing STAT3 activation. J Immunol. 2013;190:5882–5893. doi: 10.4049/jimmunol.1201881. [DOI] [PubMed] [Google Scholar]

RESOURCES