T helper 1 immunity requires complement-driven NLRP3 inflammasome activity in CD4+ T cells

Innate immune crosstalk in T cells The classical view of immune activation is that innate immune cells, such as macrophages and dendritic cells, recognize invading microbes and then alert adaptive immune cells, such as T cells, to respond. Arbore et al. now show that innate and adaptive immunity converge in human and mouse T cells. Activated T cells express components of the complement cascade, which in turn leads to the assembly of NLRP3 inflammasomes—both critical components of innate immunity that help hosts detect and eliminate microbes. In T cells, complement and inflammasomes work together to push T cells to differentiate into a specialized subset of T cells important for eliminating intracellular bacteria. Science, this issue p. 10.1126/science.aad1210 Complement and NLRP3 inflammasomes work together to promote T helper 1 cell differentiation. INTRODUCTION The inflammasomes and the complement system are traditionally viewed as quintessential components of innate immunity required for the detection and elimination of pathogens. Assembly of the NLRP3 inflammasome in innate immune cells controls the maturation of interleukin (IL)–1β, a proinflammatory cytokine critical to host defense, whereas activation of the liver-derived complement key components C3 and C5 in serum leads to opsonization and removal of microbes and induction of the inflammatory reaction. Recent studies, however, have highlighted an unanticipated direct role for complement C3 also in human T cell immunity: The anaphylatoxin C3a receptor (C3aR) and the complement regulator CD46 (which binds C3b) are critical checkpoints in human T cell lineage commitment, and they control initiation and resolution of T helper 1 (TH1) responses in an autocrine fashion via T cell–derived and intracellularly activated C3. We explored a novel functional cross-talk of complement with the NLRP3 inflammasome within CD4+ T cells and determined how the cooperation between these two “classically” innate systems directly affects interferon-γ (IFN-γ) production by adaptive immune cells. RATIONALE Given the critical role of intracellular C3 activation in human TH1 responses and the importance of C5 activation products in inflammation, we investigated whether human CD4+ T cells also harbor an “intracellular C5 activation system” and by what means this system may contribute to effector responses by using C5aR1 and C5aR2 agonists and antagonists, T cells from patients with cryopyrin-associated periodic syndromes (CAPS), and mouse models of infection and autoimmunity. RESULTS Human CD4+ T cells expressed C5 and generated increased intracellular C5a upon T cell receptor activation and CD46 autocrine costimulation. Subsequent engagement of the intracellular C5aR1 by C5a induced the generation of reactive oxygen species (ROS) and the unexpected assembly of a functional NLRP3 inflammasome in CD4+ T cells, whereas the surface-expressed C5aR2 negatively controlled this process. NLRP3 inflammasome–dependent autocrine IL-1β secretion and activity were required for optimal IFN-γ production by T cells; consequently, dysregulation of NLRP3 function in these cells affected their normal effector responses. For example, mutated, constitutively active NLRP3 in T cells from patients with CAPS induced hyperactive TH1 responses that could be normalized with a NLRP3 inhibitor. The in vivo importance of a T cell–intrinsic NLRP3 inflammasome was further supported by the finding that IFN-γ production by Nlrp3–/– CD4+ T cells was significantly reduced during viral infections in mice and that diminished TH1 induction due to lack of NLRP3 function in a CD4+ T cell transfer model of colitis led to uncontrolled TH17 infiltration and/or expansion in the intestine and aggravated disease. CONCLUSION Our results demonstrate that the regulated cross-talk between intracellularly activated complement components (the “complosome”) and the NLRP3 inflammasome is fundamental to human TH1 induction and regulation. The finding that established innate immune pathways are also operative in adaptive immune cells and orchestrate immunological responses contributes to our understanding of immunobiology and immune system evolution. In addition, the results suggest that the complement-NLRP3 axis in T cells represents a novel therapeutic target for the modulation of TH1 activity in autoimmunity and infection. An intrinsic complement-NLRP3 axis regulates human TH1 responses. T cell receptor activation and CD46 costimulation trigger NLRP3 expression and intracellular C5a generation. Subsequent intracellular C5aR1 engagement induces ROS production (and possibly IL1B gene transcription) and NLRP3 assembly, which in turn mediates IL-1β maturation. Autocrine IL-1β promotes TH1 induction (IFN-γ production) but restricts TH1 contraction (IL-10 coexpression). C5aR2 cell surface activation by secreted C5a negatively controls these events via undefined mechanisms. Dysfunction of this system contributes to impaired TH1 responses in infection or increased TH17 responses during intestinal inflammation. The NLRP3 inflammasome controls interleukin-1β maturation in antigen-presenting cells, but a direct role for NLRP3 in human adaptive immune cells has not been described. We found that the NLRP3 inflammasome assembles in human CD4+ T cells and initiates caspase-1–dependent interleukin-1β secretion, thereby promoting interferon-γ production and T helper 1 (TH1) differentiation in an autocrine fashion. NLRP3 assembly requires intracellular C5 activation and stimulation of C5a receptor 1 (C5aR1), which is negatively regulated by surface-expressed C5aR2. Aberrant NLRP3 activity in T cells affects inflammatory responses in human autoinflammatory disease and in mouse models of inflammation and infection. Our results demonstrate that NLRP3 inflammasome activity is not confined to “innate immune cells” but is an integral component of normal adaptive TH1 responses.

[1]  B. Ryffel,et al.  The receptor NLRP3 is a transcriptional regulator of TH2 differentiation , 2015, Nature Immunology.

[2]  M. Kolev,et al.  Complement Regulates Nutrient Influx and Metabolic Reprogramming during Th1 Cell Responses , 2015, Immunity.

[3]  C. Dinarello,et al.  Cell stress increases ATP release in NLRP3 inflammasome-mediated autoinflammatory diseases, resulting in cytokine imbalance , 2015, Proceedings of the National Academy of Sciences.

[4]  K. Schroder,et al.  A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases , 2015, Nature Medicine.

[5]  J. Köhl,et al.  Monitoring and Cell-Specific Deletion of C5aR1 Using a Novel Floxed GFP-C5aR1 Reporter Knock-in Mouse , 2015, The Journal of Immunology.

[6]  M. Kolev,et al.  Complement — tapping into new sites and effector systems , 2014, Nature Reviews Immunology.

[7]  K. Reymann,et al.  Nlrp3-inflammasome activation in non-myeloid-derived cells aggravates diabetic nephropathy , 2014, Kidney international.

[8]  A. Sher,et al.  Host-directed therapy of tuberculosis based on interleukin-1 and type I interferon crosstalk , 2014, Nature.

[9]  Markus F. Neurath,et al.  Cytokines in inflammatory bowel disease , 2014, Nature Reviews Immunology.

[10]  John D Lambris,et al.  Cholesterol Crystals Induce Complement-Dependent Inflammasome Activation and Cytokine Release , 2014, The Journal of Immunology.

[11]  Hongyu Zhao,et al.  Signaling through the adaptor molecule MyD88 in CD4+ T cells is required to overcome suppression by regulatory T cells. , 2014, Immunity.

[12]  Xin Geng,et al.  Cell death by pyroptosis drives CD4 T-cell depletion in HIV-1 infection , 2013, Nature.

[13]  T. Reinheckel,et al.  Intracellular Complement Activation Sustains T Cell Homeostasis and Mediates Effector Differentiation , 2013, Immunity.

[14]  P. Dasgupta,et al.  CD46: the 'multitasker' of complement proteins. , 2013, The international journal of biochemistry & cell biology.

[15]  D. Fairlie,et al.  C5a, but not C5a‐des Arg, induces upregulation of heteromer formation between complement C5a receptors C5aR and C5L2 , 2013, Immunology and cell biology.

[16]  W. Leonard,et al.  Interleukin-21 Is a Critical Cytokine for the Generation of Virus-Specific Long-Lived Plasma Cells , 2013, Journal of Virology.

[17]  X. Chen,et al.  IL-1 enhances expansion, effector function, tissue localization, and memory response of antigen-specific CD8 T cells , 2013, The Journal of experimental medicine.

[18]  T. Woodruff,et al.  C5L2: a controversial receptor of complement anaphylatoxin, C5a , 2013, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[19]  J. Licht,et al.  Mitochondria are required for antigen-specific T cell activation through reactive oxygen species signaling. , 2013, Immunity.

[20]  B. Ksander,et al.  NLRP3 inflammasome activation in retinal pigment epithelial cells by lysosomal destabilization: implications for age-related macular degeneration. , 2013, Investigative ophthalmology & visual science.

[21]  M. Heneka,et al.  NLRP3 is activated in Alzheimer´s disease and contributes to pathology in APP/PS1 mice , 2012, Nature.

[22]  G. Kolios,et al.  Host-dependent control of early regulatory and effector T-cell differentiation underlies the genetic susceptibility of RAG2-deficient mouse strains to transfer colitis , 2012, Mucosal Immunology.

[23]  C. Floudas,et al.  De novo protein design of agonists and antagonists of C5a receptors , 2012 .

[24]  Graham M Lord,et al.  The Transcription Factor T-bet Regulates Intestinal Inflammation Mediated by Interleukin-7 Receptor+ Innate Lymphoid Cells , 2012, Immunity.

[25]  H. Lehr,et al.  Protective and Aggravating Effects of Nlrp3 Inflammasome Activation in IBD Models: Influence of Genetic and Environmental Factors , 2012, Digestive Diseases.

[26]  L. O’Neill,et al.  Biochemical regulation of the inflammasome , 2012, Critical reviews in biochemistry and molecular biology.

[27]  Burkhard Becher,et al.  IL-1β mediates chronic intestinal inflammation by promoting the accumulation of IL-17A secreting innate lymphoid cells and CD4+ Th17 cells , 2012, The Journal of experimental medicine.

[28]  J. V. Sarma,et al.  New developments in C5a receptor signaling. , 2012, Cell health and cytoskeleton.

[29]  S. Bonardelli,et al.  Influence of genetic and environmental factors in peripheral arterial disease natural history: Analysis from six years follow up , 2012, International journal of applied & basic medical research.

[30]  T. Miwa,et al.  C5aR Expression in a Novel GFP Reporter Gene Knockin Mouse: Implications for the Mechanism of Action of C5aR Signaling in T Cell Immunity , 2012, The Journal of Immunology.

[31]  D. Jarrossay,et al.  Pathogen-induced human TH17 cells produce IFN-γ or IL-10 and are regulated by IL-1β , 2012, Nature.

[32]  E. Marcotte,et al.  Insights into the regulation of protein abundance from proteomic and transcriptomic analyses , 2012, Nature Reviews Genetics.

[33]  N. Scarmeas,et al.  The good, bad, and ugly? , 2012, Neurology.

[34]  P. Menu,et al.  The NLRP3 inflammasome in health and disease: the good, the bad and the ugly , 2011, Clinical and experimental immunology.

[35]  M. McDermott,et al.  Inflammasomes and autoimmunity. , 2011, Trends in molecular medicine.

[36]  R. Guimarães,et al.  Two SNPs in NLRP3 gene are involved in the predisposition to type-1 diabetes and celiac disease in a pediatric population from northeast Brazil , 2010, Autoimmunity.

[37]  Graham M Lord,et al.  Complement regulator CD46 temporally regulates cytokine production by conventional and unconventional T cells , 2010, Nature Immunology.

[38]  John D Lambris,et al.  Complement: a key system for immune surveillance and homeostasis , 2010, Nature Immunology.

[39]  Brigitta Stockinger,et al.  Effector T cell plasticity: flexibility in the face of changing circumstances , 2010, Nature Immunology.

[40]  Kate Schroder,et al.  The NLRP3 Inflammasome: A Sensor for Metabolic Danger? , 2010, Science.

[41]  C. Mackay,et al.  The C5a Receptor (C5aR) C5L2 Is a Modulator of C5aR-mediated Signal Transduction* , 2009, The Journal of Biological Chemistry.

[42]  K. Mills,et al.  Interleukin-1 and IL-23 induce innate IL-17 production from gammadelta T cells, amplifying Th17 responses and autoimmunity. , 2009, Immunity.

[43]  Helen J. Lachmann,et al.  In vivo regulation of interleukin 1β in patients with cryopyrin-associated periodic syndromes , 2009, The Journal of experimental medicine.

[44]  W. Paul,et al.  IL-1 acts directly on CD4 T cells to enhance their antigen-driven expansion and differentiation , 2009, Proceedings of the National Academy of Sciences.

[45]  P. Monk,et al.  The human complement fragment receptor, C5L2, is a recycling decoy receptor , 2009, Molecular immunology.

[46]  D. Gerlier,et al.  Human C3 Deficiency Associated with Impairments in Dendritic Cell Differentiation, Memory B Cells, and Regulatory T Cells1 , 2008, The Journal of Immunology.

[47]  C. Gabay,et al.  IL‐1, IL‐18, and IL‐33 families of cytokines , 2008, Immunological reviews.

[48]  G. Dubyak,et al.  Locally produced complement fragments C5a and C3a provide both costimulatory and survival signals to naive CD4+ T cells. , 2008, Immunity.

[49]  P. Söderkvist,et al.  Genetic variation in proteins of the cryopyrin inflammasome influences susceptibility and severity of rheumatoid arthritis (the Swedish TIRA project). , 2007, Rheumatology.

[50]  K. Tracey,et al.  IL-1α and IL-1β Are Endogenous Mediators Linking Cell Injury to the Adaptive Alloimmune Response1 , 2007, The Journal of Immunology.

[51]  G. Meiffren,et al.  Alterations in CD46-mediated Tr1 regulatory T cells in patients with multiple sclerosis. , 2006, The Journal of clinical investigation.

[52]  K. Mills,et al.  A crucial role for interleukin (IL)-1 in the induction of IL-17–producing T cells that mediate autoimmune encephalomyelitis , 2006, The Journal of experimental medicine.

[53]  Pixu Liu,et al.  An Anti-inflammatory Function for the Complement Anaphylatoxin C5a-binding Protein, C5L2* , 2005, Journal of Biological Chemistry.

[54]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[55]  F. Martinon,et al.  NLRs join TLRs as innate sensors of pathogens. , 2005, Trends in immunology.

[56]  P. Monk,et al.  C5a Mutants Are Potent Antagonists of the C5a Receptor (CD88) and of C5L2 , 2004, Journal of Biological Chemistry.

[57]  P. Monk,et al.  The C terminus of the human C5a receptor (CD88) is required for normal ligand‐dependent receptor internalization , 1997, European journal of immunology.

[58]  C. Dinarello,et al.  Biologic basis for interleukin-1 in disease. , 1996, Blood.

[59]  H. Okamura,et al.  Cloning of a new cytokine that induces IFN-γ production by T cells , 1995, Nature.

[60]  P. Handford,et al.  The CD 46-Jagged 1 interaction is critical for human TH 1 immunity , 2012 .

[61]  J. V. Sarma,et al.  New developments in C 5 a receptor signaling , 2012 .

[62]  Scott W. Cousins,et al.  Implications for Age-Related Macular Degeneration , 2010 .

[63]  K. Tracey,et al.  IL-1alpha and IL-1beta are endogenous mediators linking cell injury to the adaptive alloimmune response. , 2007, Journal of immunology.

[64]  G. Meiffren,et al.  Alterations in CD 46-mediated Tr 1 regulatory T cells in patients with multiple sclerosis , 2006 .

[65]  P. Monk,et al.  Therapeutic activity of C5a receptor antagonists in a rat model of neurodegeneration , 2006, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.