Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Science ; 384(6702): eade8520, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38900864

ABSTRACT

Unleashing antitumor T cell activity by checkpoint inhibitor immunotherapy is effective in cancer patients, but clinical responses are limited. Cytokine signaling through the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway correlates with checkpoint immunotherapy resistance. We report a phase I clinical trial of the JAK inhibitor ruxolitinib with anti-PD-1 antibody nivolumab in Hodgkin lymphoma patients relapsed or refractory following checkpoint inhibitor immunotherapy. The combination yielded a best overall response rate of 53% (10/19). Ruxolitinib significantly reduced neutrophil-to-lymphocyte ratios and percentages of myeloid suppressor cells but increased numbers of cytokine-producing T cells. Ruxolitinib rescued the function of exhausted T cells and enhanced the efficacy of immune checkpoint blockade in preclinical solid tumor and lymphoma models. This synergy was characterized by a switch from suppressive to immunostimulatory myeloid cells, which enhanced T cell division.


Subject(s)
Hodgkin Disease , Immune Checkpoint Inhibitors , Janus Kinase Inhibitors , Nitriles , Nivolumab , Pyrazoles , Pyrimidines , T-Lymphocytes , Adult , Aged , Animals , Female , Humans , Male , Mice , Middle Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Synergism , Hodgkin Disease/drug therapy , Hodgkin Disease/immunology , Hodgkin Disease/therapy , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy , Janus Kinase Inhibitors/therapeutic use , Janus Kinases/metabolism , Janus Kinases/antagonists & inhibitors , Nitriles/therapeutic use , Nivolumab/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , T-Lymphocytes/immunology , Mice, Inbred C57BL , Mice, Inbred BALB C
2.
Cell Rep ; 29(10): 3293-3302.e3, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31801090

ABSTRACT

Inhibitory receptors (IRs) function as critical regulators of immune responses by tempering T cell activity. In humans, several persisting viruses as well as cancers exploit IR signaling by upregulating IR ligands, resulting in suppression of T cell function (i.e., exhaustion). This allows escape from immune surveillance and continuation of disease. Here, we report the design, implementation, and results of a phenotypic high-throughput screen for molecules that modulate CD8+ T cell activity. We identify 19 compounds from the ReFRAME drug-repurposing collection that restore cytokine production and enhance the proliferation of exhausted T cells. Analysis of our top hit, ingenol mebutate, a protein kinase C (PKC) inducing diterpene ester, reveals a role for this molecule in overriding the suppressive signaling cascade mediated by IR signaling on T cells. Collectively, these results demonstrate a disease-relevant methodology for identifying modulators of T cell function and reveal new targets for immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Small Molecule Libraries/pharmacology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Chlorocebus aethiops , Costimulatory and Inhibitory T-Cell Receptors/metabolism , Cytokines/metabolism , Immunotherapy/methods , Male , Mice , Mice, Inbred C57BL , Protein Kinase C/metabolism , Signal Transduction/drug effects , Vero Cells
3.
Proc Natl Acad Sci U S A ; 116(36): 18001-18008, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31427525

ABSTRACT

Lymphocytic choriomeningitis virus (LCMV) WE variant 2.2 (v2.2) generated a high level of the major mouse urinary protein: MUP. Mice infected with LCMV WE v54, which differed from v2.2 by a single amino acid in the viral glycoprotein, failed to generate MUP above baseline levels found in uninfected controls. Variant 54 bound at 2.5 logs higher affinity to the LCMV receptor α-dystroglycan (α-DG) than v2.2 and entered α-DG-expressing but not α-DG-null cells. Variant 2.2 infected both α-DG-null or -expressing cells. Variant 54 infected more dendritic cells, generated a negligible CD8 T cell response, and caused a persistent infection, while v2.2 generated cytotoxic T lymphocytes (CTLs) and cleared virus within 10 days. By 20 days postinfection and through the 80-day observation period, significantly higher amounts of MUP were found in v2.2-infected mice. Production of MUP was dependent on virus-specific CTL as deletion of such cells aborted MUP production. Furthermore, MUP production was not elevated in v2.2 persistently infected mice unless virus was cleared following transfer of virus-specific CTL.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Proteins/immunology , Animals , Dystroglycans/immunology , Lymphocytic Choriomeningitis/pathology , Mice
4.
J Virol ; 93(18)2019 09 15.
Article in English | MEDLINE | ID: mdl-31243125

ABSTRACT

CXCR2 is a chemokine receptor expressed on oligodendroglia that has been implicated in the pathogenesis of neuroinflammatory demyelinating diseases as well as enhancement of the migration, proliferation, and myelin production by oligodendroglia. Using an inducible proteolipid protein (Plp) promoter-driven Cre-loxP recombination system, we were able to assess how timed ablation of Cxcr2 in oligodendroglia affected disease following intracranial infection with the neurotropic JHM strain of mouse hepatitis virus (JHMV). Generation of Plp-Cre-ER(T)::Cxcr2flox/flox transgenic mice (termed Cxcr2-CKO mice) allows for Cxcr2 to be silenced in oligodendrocytes in adult mice following treatment with tamoxifen. Ablation of oligodendroglia Cxcr2 did not influence clinical severity in response to intracranial infection with JHMV. Infiltration of activated T cells or myeloid cells into the central nervous system (CNS) was not affected, nor was the ability to control viral infection. In addition, the severity of demyelination was similar between tamoxifen-treated mice and vehicle-treated controls. Notably, deletion of Cxcr2 resulted in increased remyelination, as assessed by g-ratio (the ratio of the inner axonal diameter to the total outer fiber diameter) calculation, compared to that in vehicle-treated control mice. Collectively, our findings argue that CXCR2 signaling in oligodendroglia is dispensable with regard to contributing to neuroinflammation, but its deletion enhances remyelination in a preclinical model of the human demyelinating disease multiple sclerosis (MS).IMPORTANCE Signaling through the chemokine receptor CXCR2 in oligodendroglia is important for developmental myelination in rodents, while chemical inhibition or nonspecific genetic deletion of CXCR2 appears to augment myelin repair in animal models of the human demyelinating disease multiple sclerosis (MS). To better understand the biology of CXCR2 signaling on oligodendroglia, we generated transgenic mice in which Cxcr2 is selectively ablated in oligodendroglia upon treatment with tamoxifen. Using a viral model of neuroinflammation and demyelination, we demonstrate that genetic silencing of CXCR2 on oligodendroglia did not affect clinical disease, neuroinflammation, or demyelination, yet there was increased remyelination. These findings support and extend previous findings suggesting that targeting CXCR2 may offer a therapeutic avenue for enhancing remyelination in patients with demyelinating diseases.


Subject(s)
Multiple Sclerosis/metabolism , Myelin Sheath/metabolism , Receptors, Interleukin-8B/metabolism , Remyelination/physiology , Animals , Axons/metabolism , Axons/pathology , Cell Differentiation/physiology , Cell Line , Central Nervous System/pathology , Demyelinating Diseases/virology , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Multiple Sclerosis/pathology , Murine hepatitis virus/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Oligodendroglia/metabolism , Receptors, Interleukin-8B/genetics , Signal Transduction , Stem Cells , Tamoxifen/pharmacology
5.
JCI Insight ; 4(2)2019 Jan 24.
Article in English | MEDLINE | ID: mdl-30674713

ABSTRACT

Here, we report a pathogenic role for type I IFN (IFN-I) signaling in macrophages, and not ß cells in the islets, for the development of type 1 diabetes (T1D). Following lymphocytic choriomeningitis (LCMV) infection in the Rip-LCMV-GP T1D model, macrophages accumulated near islets and in close contact to islet-infiltrating GP-specific (autoimmune) CD8+ T cells. Depletion of macrophages with clodronate liposomes or genetic ablation of Ifnar in macrophages aborted T1D, despite proliferation of GP-specific (autoimmune) CD8+ T cells. Histopathologically, disrupted IFNα/ß receptor (IFNAR) signaling in macrophages resulted in restriction of CD8+ T cells entering into the islets with significant lymphoid accumulation around the islet. Collectively, these results provide evidence that macrophages via IFN-I signaling, while not entering the islets, are directly involved in interacting, directing, or restricting trafficking of autoreactive-specific T cells into the islets as an important component in causing T1D.

6.
Viral Immunol ; 32(1): 25-37, 2019.
Article in English | MEDLINE | ID: mdl-30109979

ABSTRACT

Chemokines (chemotactic cytokines) are involved in a wide variety of biological processes. Following microbial infection, there is often robust chemokine signaling elicited from infected cells, which contributes to both innate and adaptive immune responses that control growth of the invading pathogen. Infection of the central nervous system (CNS) by the neuroadapted John Howard Mueller (JHM) strain of mouse hepatitis virus (JHMV) provides an excellent example of how chemokines aid in host defense as well as contribute to disease. Intracranial inoculation of the CNS of susceptible mice with JHMV results in an acute encephalomyelitis characterized by widespread dissemination of virus throughout the parenchyma. Virus-specific T cells are recruited to the CNS, and control viral replication through release of antiviral cytokines and cytolytic activity. Sterile immunity is not acquired, and virus will persist primarily in white matter tracts leading to chronic neuroinflammation and demyelination. Chemokines are expressed and contribute to defense as well as chronic disease by attracting targeted populations of leukocytes to the CNS. The T cell chemoattractant chemokine CXCL10 (interferon-inducible protein 10 kDa, IP-10) is prominently expressed in both stages of disease, and serves to attract activated T and B lymphocytes expressing CXC chemokine receptor 3 (CXCR3), the receptor for CXCL10. Functional studies that have blocked expression of either CXCL10 or CXCR3 illuminate the important role of this signaling pathway in host defense and neurodegeneration in a model of viral-induced neurologic disease.


Subject(s)
Central Nervous System Viral Diseases/immunology , Chemokine CXCL10/immunology , Coronavirus Infections/immunology , Demyelinating Diseases/immunology , Murine hepatitis virus/immunology , Animals , Central Nervous System Viral Diseases/pathology , Chemokine CXCL10/genetics , Chemotactic Factors , Coronavirus Infections/pathology , Demyelinating Diseases/virology , Humans , Mice , Mice, Inbred C57BL , Murine hepatitis virus/pathogenicity , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Signal Transduction , T-Lymphocytes/immunology
7.
Eur J Immunol ; 48(7): 1199-1210, 2018 07.
Article in English | MEDLINE | ID: mdl-29697856

ABSTRACT

Increasing evidence points to an important role for neutrophils in participating in the pathogenesis of the human demyelinating disease MS and the animal model EAE. Therefore, a better understanding of the signals controlling migration of neutrophils as well as evaluating the role of these cells in demyelination is important to define cellular components that contribute to disease in MS patients. In this study, we examined the functional role of the chemokine CXCL1 in contributing to neuroinflammation and demyelination in EAE. Using transgenic mice in which expression of CXCL1 is under the control of a tetracycline-inducible promoter active within glial fibrillary acidic protein-positive cells, we have shown that sustained CXCL1 expression within the CNS increased the severity of clinical and histologic disease that was independent of an increase in the frequency of encephalitogenic Th1 and Th17 cells. Rather, disease was associated with enhanced recruitment of CD11b+ Ly6G+ neutrophils into the spinal cord. Targeting neutrophils resulted in a reduction in demyelination arguing for a role for these cells in myelin damage. Collectively, these findings emphasize that CXCL1-mediated attraction of neutrophils into the CNS augments demyelination suggesting that this signaling pathway may offer new targets for therapeutic intervention.


Subject(s)
Central Nervous System/immunology , Chemokine CXCL1/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Neutrophils/immunology , Spinal Cord/immunology , Animals , Autoantigens/immunology , CD11b Antigen/metabolism , Cells, Cultured , Chemokine CXCL1/genetics , Disease Models, Animal , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Myelin-Oligodendrocyte Glycoprotein/immunology , Neurogenic Inflammation , Neutrophil Infiltration , Peptide Fragments/immunology , Signal Transduction , Spinal Cord/pathology
8.
Virology ; 511: 49-55, 2017 11.
Article in English | MEDLINE | ID: mdl-28822268

ABSTRACT

The present study examines the susceptibility of mouse induced pluripotent stem cell-derived neural precursor cells (iPSC-NPCs) to infection with the neurotropic JHM strain of mouse hepatitis virus (JHMV). Similar to NPCs derived from striatum of day 1 postnatal GFP-transgenic mice (GFP-NPCs), iPSC-derived NPCs (iPSC-NPCs) are able to differentiate into terminal neural cell types and express MHC class I and II in response to IFN-γ treatment. However, in contrast to postnatally-derived NPCs, iPSC-NPCs express low levels of carcinoembryonic antigen-cell adhesion molecule 1a (CEACAM1a), the surface receptor for JHMV, and are less susceptible to infection and virus-induced cytopathic effects. The relevance of this in terms of therapeutic application of NPCs resistant to viral infection is discussed.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/physiology , Murine hepatitis virus/growth & development , Murine hepatitis virus/immunology , Neural Stem Cells/immunology , Neural Stem Cells/virology , Animals , Carcinoembryonic Antigen/biosynthesis , Gene Expression , Histocompatibility Antigens Class I/biosynthesis , Histocompatibility Antigens Class II/biosynthesis , Interferon-gamma/metabolism , Mice , Mice, Transgenic
9.
Virology ; 511: 106-113, 2017 11.
Article in English | MEDLINE | ID: mdl-28843812

ABSTRACT

Plasmacytoid dendritic cells (pDCs), a main source of type I interferon in response to viral infection, are an early cell target during lymphocytic choriomeningitis virus (LCMV) infection, which has been associated with the LCMV's ability to establish chronic infections. Human blood-derived pDCs have been reported to be refractory to ex vivo LCMV infection. In the present study we show that human pDC CAL-1 cells are refractory to infection with cell-free LCMV, but highly susceptible to infection with recombinant LCMVs carrying the surface glycoprotein of VSV, indicating that LCMV infection of CAL-1 cells is restricted at the cell entry step. Co-culture of uninfected CAL-1 cells with LCMV-infected HEK293 cells enabled LCMV to infect CAL-1 cells. This cell-to-cell spread required direct cell-cell contact and did not involve exosome pathway. Our findings indicate the presence of a novel entry pathway utilized by LCMV to infect pDC.


Subject(s)
Dendritic Cells/virology , Lymphocytic choriomeningitis virus/physiology , Virus Internalization , Cells, Cultured , Coculture Techniques , Humans
10.
Proc Natl Acad Sci U S A ; 114(14): 3708-3713, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28325871

ABSTRACT

Blockade of IFN-α but not IFN-ß signaling using either an antibody or a selective S1PR1 agonist, CYM-5442, prevented type 1 diabetes (T1D) in the mouse Rip-LCMV T1D model. First, treatment with antibody or CYM-5442 limited the migration of autoimmune "antiself" T cells to the external boundaries around the islets and prevented their entry into the islets so they could not be positioned to engage, kill, and thus remove insulin-producing ß cells. Second, CYM-5442 induced an exhaustion signature in antiself T cells by up-regulating the negative immune regulator receptor genes Pdcd1, Lag3, Ctla4, Tigit, and Btla, thereby limiting their killing ability. By such means, insulin production was preserved and glucose regulation maintained, and a mechanism for S1PR1 immunomodulation described.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Indans/administration & dosage , Interferon-alpha/metabolism , Oxadiazoles/administration & dosage , Prediabetic State/drug therapy , Receptors, Lysosphingolipid/agonists , T-Lymphocytes/drug effects , Animals , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Disease Progression , Indans/pharmacology , Insulin/metabolism , Insulin-Secreting Cells/immunology , Islets of Langerhans/immunology , Mice , Oxadiazoles/pharmacology , Prediabetic State/immunology , Receptors, Immunologic/metabolism , Signal Transduction/drug effects , Sphingosine-1-Phosphate Receptors , T-Lymphocytes/immunology
11.
J Immunol ; 196(4): 1855-64, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26773148

ABSTRACT

The functional role of the ELR(+) chemokine CXCL1 in host defense and disease following infection of the CNS with the neurotropic JHM strain of mouse hepatitis virus (JHMV) was examined. Mice in which expression of CXCL1 is under the control of a tetracycline-inducible promoter active within glial fibrillary acidic protein-positive cells were generated and this allowed for selectively increasing CNS expression of CXCL1 in response to JHMV infection and evaluating the effects on neuroinflammation, control of viral replication, and demyelination. Inducible expression of CNS-derived CXCL1 resulted in increased levels of CXCL1 protein within the serum, brain, and spinal cord that correlated with increased frequency of Ly6G(+)CD11b(+) neutrophils present within the CNS. Elevated levels of CXCL1 did not influence the generation of virus-specific T cells, and there was no difference in control of JHMV replication compared with control mice, indicating that T cell infiltration into the CNS is CXCL1-independent. Sustained CXCL1 expression within the CNS resulted in increased mortality that correlated with elevated neutrophil infiltration, diminished numbers of mature oligodendrocytes, and an increase in the severity of demyelination. Neutrophil ablation in CXCL1-transgenic mice reduced the severity of demyelination in mice, arguing for a role for these cells in white matter damage. Collectively, these findings illustrate that sustained CXCL1 expression amplifies the severity of white matter damage and that neutrophils can contribute to this process in a model of viral-induced neurologic disease.


Subject(s)
Central Nervous System Diseases/immunology , Central Nervous System Diseases/pathology , Chemokine CXCL1/immunology , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Animals , Central Nervous System Diseases/virology , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Mice , Mice, Inbred C57BL , Mice, Transgenic , Murine hepatitis virus , Neutrophil Infiltration , Real-Time Polymerase Chain Reaction , White Matter/immunology , White Matter/pathology
12.
Expert Rev Neurother ; 14(10): 1169-79, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25245576

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory disease of the CNS. While a broad range of therapeutics effectively reduce the incidence of focal white matter inflammation and plaque formation for patients with relapse-remitting forms of MS, a challenge within the field is to develop therapies that allow for axonal protection and remyelination. In the last decade, growing interest has focused on utilizing neural precursor cells (NPCs) to promote remyelination. To understand how NPCs function in chronic demyelinating environments, several excellent pre-clinical mouse models have been developed. One well accepted model is infection of susceptible mice with neurotropic variants of mouse hepatitis virus (MHV) that undergo chronic demyelination exhibiting clinical and histopathologic similarities to MS patients. Combined with the possibility that an environmental agent such as a virus could trigger MS, the MHV model of demyelination presents a relevant mouse model to assess the therapeutic potential of NPCs transplanted into an environment in which inflammatory-mediated demyelination is established.


Subject(s)
Cell- and Tissue-Based Therapy , Demyelinating Diseases/therapy , Disease Models, Animal , Multiple Sclerosis/therapy , Neural Stem Cells/cytology , Animals , Cell- and Tissue-Based Therapy/methods , Demyelinating Diseases/pathology , Humans , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Regeneration/physiology
13.
Virology ; 435(1): 110-7, 2013 Jan 05.
Article in English | MEDLINE | ID: mdl-23217621

ABSTRACT

Inoculation with the neurotropic JHM strain of mouse hepatitis virus (MHV) into the central nervous system (CNS) of susceptible strains of mice results in an acute encephalomyelitis in which virus preferentially replicates within glial cells while excluding neurons. Control of viral replication during acute disease is mediated by infiltrating virus-specific T cells via cytokine secretion and cytolytic activity, however sterile immunity is not achieved and virus persists resulting in chronic neuroinflammation associated with demyelination. CXCR2 is a chemokine receptor that upon binding to specific ligands promotes host defense through recruitment of myeloid cells to the CNS as well as protecting oligodendroglia from cytokine-mediated death in response to MHV infection. These findings highlight growing evidence of the diverse and important role of CXCR2 in regulating neuroinflammatory diseases.


Subject(s)
Central Nervous System/virology , Coronavirus Infections/virology , Demyelinating Diseases/virology , Encephalomyelitis/virology , Murine hepatitis virus/physiology , Oligodendroglia/virology , Receptors, Interleukin-8B/immunology , Acute Disease , Animals , Cell Movement , Central Nervous System/immunology , Central Nervous System/pathology , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Cytokines/biosynthesis , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Encephalomyelitis/immunology , Encephalomyelitis/pathology , Mice , Murine hepatitis virus/pathogenicity , Myeloid Cells/immunology , Myeloid Cells/virology , Oligodendroglia/immunology , Oligodendroglia/pathology , Receptors, Interleukin-8B/genetics , T-Lymphocytes/immunology , T-Lymphocytes/virology , Virus Replication
14.
Future Virol ; 7(4): 349-359, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22582084

ABSTRACT

Inoculation of the neurotropic JHM strain of mouse hepatitis virus (JHMV) into the central nervous system (CNS) of susceptible strains of mice results in wide-spread replication within glial cells accompanied by infiltration of virus-specific T lymphocytes that control virus through cytokine secretion and cytolytic activity. Virus persists within white matter tracts of surviving mice resulting in demyelination that is amplified by inflammatory T cells and macrophages. In response to infection, numerous cytokines/chemokines are secreted by resident cells of the CNS and inflammatory leukocytes that participate in both host defense and disease. Among these are the ELR-positive chemokines that are able to signal through CXC chemokine receptors including CXCR2. Early following JHMV infection, ELR-positive chemokines contribute to host defense by attracting CXCR2-expressing cells including polymorphonuclear cells to the CNS that aid in host defense through increasing the permeability the blood-brain-barrier (BBB). During chronic disease, CXCR2 signaling on oligodendroglia protects these cells from apoptosis and restricts the severity of demyelination. This review covers aspects related to host defense and disease in response to JHMV infection and highlights the different roles of CXCR2 signaling in these processes.

15.
Proc Natl Acad Sci U S A ; 108(37): 15312-7, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21876153

ABSTRACT

Caspase-8 (casp8) is required for extrinsic apoptosis, and mice deficient in casp8 fail to develop and die in utero while ultimately failing to maintain the proliferation of T cells, B cells, and a host of other cell types. Paradoxically, these failures are not caused by a defect in apoptosis, but by a presumed proliferative function of this protease. Indeed, following mitogenic stimulation, T cells lacking casp8 or its adaptor protein FADD (Fas-associated death domain protein) develop a hyperautophagic morphology, and die a programmed necrosis-like death process termed necroptosis. Recent studies have demonstrated that receptor-interacting protein kinases (RIPKs) RIPK1 and RIPK3 together facilitate TNF-induced necroptosis, but the precise role of RIPKs in the demise of T cells lacking FADD or casp8 activity is unknown. Here we demonstrate that RIPK3 and FADD have opposing and complementary roles in promoting T-cell clonal expansion and homeostasis. We show that the defective proliferation of T cells bearing an interfering form of FADD (FADDdd) is rescued by crossing with RIPK3(-/-) mice, although such rescue ultimately leads to lymphadenopathy. Enhanced recovery of these double-mutant T cells following stimulation demonstrates that FADD, casp8, and RIPK3 are all essential for clonal expansion, contraction, and antiviral responses. Finally, we demonstrate that caspase-mediated cleavage of RIPK1-containing necrosis inducing complexes (necrosomes) is sufficient to prevent necroptosis in the face of death receptor signaling. These studies highlight the "two-faced" nature of casp8 activity, promoting clonal expansion in some situations and apoptotic demise in others.


Subject(s)
CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/virology , Fas-Associated Death Domain Protein/metabolism , Homeostasis/immunology , Immunity/immunology , Murine hepatitis virus/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Proliferation , Cell Survival , Crosses, Genetic , Female , Hepatitis, Viral, Animal/immunology , Hepatitis, Viral, Animal/virology , Male , Mice , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...