Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell ; 161(4): 750-61, 2015 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-25957683

RESUMO

Memory T cells are critical for long-term immunity against reinfection and require interleukin-7 (IL-7), but the mechanisms by which IL-7 controls memory T cell survival, particularly metabolic fitness, remain elusive. We discover that IL-7 induces expression of the glycerol channel aquaporin 9 (AQP9) in virus-specific memory CD8+ T cells, but not naive cells, and that AQP9 is vitally required for their long-term survival. AQP9 deficiency impairs glycerol import into memory CD8+ T cells for fatty acid esterification and triglyceride (TAG) synthesis and storage. These defects can be rescued by ectopic expression of TAG synthases, which restores lipid stores and memory T cell survival. Finally, we find that TAG synthesis is a central component of IL-7-mediated survival of human and mouse memory CD8+T cells. This study uncovers the metabolic mechanisms by which IL-7 tailors the metabolism of memory T cells to promote their longevity and fast response to rechallenge.


Assuntos
Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Sobrevivência Celular , Memória Imunológica , Triglicerídeos/metabolismo , Animais , Aquaporinas/metabolismo , Infecções por Arenaviridae/imunologia , Linfócitos T CD8-Positivos/metabolismo , Glicerol/metabolismo , Humanos , Interleucina-7/metabolismo , Vírus da Coriomeningite Linfocítica/fisiologia , Camundongos , Transdução de Sinais
2.
Cell ; 162(6): 1217-28, 2015 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-26321681

RESUMO

Activated T cells engage aerobic glycolysis and anabolic metabolism for growth, proliferation, and effector functions. We propose that a glucose-poor tumor microenvironment limits aerobic glycolysis in tumor-infiltrating T cells, which suppresses tumoricidal effector functions. We discovered a new role for the glycolytic metabolite phosphoenolpyruvate (PEP) in sustaining T cell receptor-mediated Ca(2+)-NFAT signaling and effector functions by repressing sarco/ER Ca(2+)-ATPase (SERCA) activity. Tumor-specific CD4 and CD8 T cells could be metabolically reprogrammed by increasing PEP production through overexpression of phosphoenolpyruvate carboxykinase 1 (PCK1), which bolstered effector functions. Moreover, PCK1-overexpressing T cells restricted tumor growth and prolonged the survival of melanoma-bearing mice. This study uncovers new metabolic checkpoints for T cell activity and demonstrates that metabolic reprogramming of tumor-reactive T cells can enhance anti-tumor T cell responses, illuminating new forms of immunotherapy.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos do Interstício Tumoral/imunologia , Melanoma/imunologia , Melanoma/terapia , Monitorização Imunológica , Fosfoenolpiruvato/metabolismo , Microambiente Tumoral , Animais , Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , Glicólise , Hexoquinase/metabolismo , Imunoterapia , Camundongos , Fatores de Transcrição NFATC/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/imunologia
3.
Immunity ; 43(4): 690-702, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26410627

RESUMO

The differentiation of CD4(+) helper T cell subsets with diverse effector functions is accompanied by changes in metabolism required to meet their bioenergetic demands. We find that follicular B helper T (Tfh) cells exhibited less proliferation, glycolysis, and mitochondrial respiration, accompanied by reduced mTOR kinase activity compared to T helper 1 (Th1) cells in response to acute viral infection. IL-2-mediated activation of the Akt kinase and mTORc1 signaling was both necessary and sufficient to shift differentiation away from Tfh cells, instead promoting that of Th1 cells. These findings were not the result of generalized signaling attenuation in Tfh cells, because they retained the ability to flux calcium and activate NFAT-transcription-factor-dependent cytokine production. These data identify the interleukin-2 (IL-2)-mTORc1 axis as a critical orchestrator of the reciprocal balance between Tfh and Th1 cell fates and their respective metabolic activities after acute viral infection.


Assuntos
Interleucina-2/fisiologia , Complexos Multiproteicos/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/fisiologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Auxiliares-Indutores/metabolismo , Serina-Treonina Quinases TOR/fisiologia , Animais , Apoptose , Sinalização do Cálcio , Ciclo Celular , Divisão Celular , Ativação Enzimática , Glucose/metabolismo , Glicólise , Subunidade alfa de Receptor de Interleucina-2/fisiologia , Vírus da Coriomeningite Linfocítica/imunologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFATC/fisiologia , Consumo de Oxigênio , Fator 1 de Ligação ao Domínio I Regulador Positivo , Organismos Livres de Patógenos Específicos , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/virologia , Células Th1/citologia , Células Th1/imunologia , Células Th1/metabolismo , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
4.
Int J Mol Sci ; 25(10)2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38791146

RESUMO

Crohn's disease (CD) is a subtype of inflammatory bowel disease (IBD) characterized by transmural disease. The concept of transmural healing (TH) has been proposed as an indicator of deep clinical remission of CD and as a predictor of favorable treatment endpoints. Understanding the pathophysiology involved in transmural disease is critical to achieving these endpoints. However, most studies have focused on the intestinal mucosa, overlooking the contribution of the intestinal wall in Crohn's disease. Multi-omics approaches have provided new avenues for exploring the pathogenesis of Crohn's disease and identifying potential biomarkers. We aimed to use transcriptomic and proteomic technologies to compare immune and mesenchymal cell profiles and pathways in the mucosal and submucosa/wall compartments to better understand chronic refractory disease elements to achieve transmural healing. The results revealed similarities and differences in gene and protein expression profiles, metabolic mechanisms, and immune and non-immune pathways between these two compartments. Additionally, the identification of protein isoforms highlights the complex molecular mechanisms underlying this disease, such as decreased RTN4 isoforms (RTN4B2 and RTN4C) in the submucosa/wall, which may be related to the dysregulation of enteric neural processes. These findings have the potential to inform the development of novel therapeutic strategies to achieve TH.


Assuntos
Colo , Doença de Crohn , Mucosa Intestinal , Proteômica , Humanos , Doença de Crohn/metabolismo , Doença de Crohn/patologia , Doença de Crohn/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Proteômica/métodos , Colo/metabolismo , Colo/patologia , Transcriptoma , Masculino , Feminino , Adulto , Perfilação da Expressão Gênica , Biomarcadores , Pessoa de Meia-Idade , Multiômica
5.
Immunity ; 40(3): 367-77, 2014 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-24631156

RESUMO

Follicular helper T (Tfh) cells are required for the establishment of T-dependent B cell memory and high affinity antibody-secreting cells. We have revealed herein opposing roles for signal transducer and activator of transcription 3 (STAT3) and type I interferon (IFN) signaling in the differentiation of Tfh cells following viral infection. STAT3-deficient CD4(+) T cells had a profound defect in Tfh cell differentiation, accompanied by decreased germinal center (GC) B cells and antigen-specific antibody production during acute infection with lymphocytic choriomeningitis virus. STAT3-deficient Tfh cells had strikingly increased expression of a number of IFN-inducible genes, in addition to enhanced T-bet synthesis, thus adopting a T helper 1 (Th1) cell-like effector phenotype. Conversely, IFN-αß receptor blockade restored Tfh and GC B cell phenotypes in mice containing STAT3-deficient CD4(+) T cells. These data suggest mutually repressive roles for STAT3 and type I IFN signaling pathways in the differentiation of Tfh cells following viral infection.


Assuntos
Diferenciação Celular , Interferon Tipo I/metabolismo , Fator de Transcrição STAT3/metabolismo , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/metabolismo , Animais , Anticorpos Antivirais/imunologia , Especificidade de Anticorpos/imunologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Antígenos CD4/genética , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Switching de Imunoglobulina/genética , Interferon Tipo I/genética , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/metabolismo , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Knockout , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/deficiência , Fator de Transcrição STAT3/genética , Transdução de Sinais , Linfócitos T Auxiliares-Indutores/imunologia , Transcriptoma
6.
Immunity ; 41(5): 802-14, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25464856

RESUMO

Protein kinase B (also known as AKT) and the mechanistic target of rapamycin (mTOR) are central regulators of T cell differentiation, proliferation, metabolism, and survival. Here, we show that during chronic murine lymphocytic choriomeningitis virus infection, activation of AKT and mTOR are impaired in antiviral cytotoxic T lymphocytes (CTLs), resulting in enhanced activity of the transcription factor FoxO1. Blockade of inhibitory receptor programmed cell death protein 1 (PD-1) in vivo increased mTOR activity in virus-specific CTLs, and its therapeutic effects were abrogated by the mTOR inhibitor rapamycin. FoxO1 functioned as a transcriptional activator of PD-1 that promoted the differentiation of terminally exhausted CTLs. Importantly, FoxO1-null CTLs failed to persist and control chronic viral infection. Collectively, this study shows that CTLs adapt to persistent infection through a positive feedback pathway (PD-1?FoxO1?PD-1) that functions to both desensitize virus-specific CTLs to antigen and support their survival during chronic viral infection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Fatores de Transcrição Forkhead/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Receptor de Morte Celular Programada 1/biossíntese , Linfócitos T Citotóxicos/imunologia , Animais , Anticorpos Bloqueadores/farmacologia , Anticorpos Monoclonais/farmacologia , Antígenos CD28/imunologia , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Doença Crônica , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Granzimas/biossíntese , Humanos , Interferon gama/biossíntese , Células Jurkat , Ativação Linfocitária/imunologia , Coriomeningite Linfocítica/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor de Morte Celular Programada 1/imunologia , Proteínas Proto-Oncogênicas c-akt/biossíntese , Receptores de Antígenos de Linfócitos T/imunologia , Sirolimo/farmacologia , Linfócitos T Citotóxicos/citologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/biossíntese
7.
Immunity ; 38(5): 984-97, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23623381

RESUMO

Regulation of metabolic pathways in the immune system provides a mechanism to actively control cellular function, growth, proliferation, and survival. Here, we report that miR-181 is a nonredundant determinant of cellular metabolism and is essential for supporting the biosynthetic demands of early NKT cell development. As a result, miR-181-deficient mice showed a complete absence of mature NKT cells in the thymus and periphery. Mechanistically, miR-181 modulated expression of the phosphatase PTEN to control PI3K signaling, which was a primary stimulus for anabolic metabolism in immune cells. Thus miR-181-deficient mice also showed severe defects in lymphoid development and T cell homeostasis associated with impaired PI3K signaling. These results uncover miR-181 as essential for NKT cell development and establish this family of miRNAs as central regulators of PI3K signaling and global metabolic fitness during development and homeostasis.


Assuntos
Linfopoese/genética , MicroRNAs/metabolismo , Células T Matadoras Naturais/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Animais , Diferenciação Celular , Quimiocina CXCL12/metabolismo , Regulação para Baixo , Homeostase , Linfócitos/metabolismo , Camundongos , MicroRNAs/genética , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/genética
8.
Nature ; 520(7548): 553-7, 2015 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-25642965

RESUMO

Mitochondrial DNA (mtDNA) is normally present at thousands of copies per cell and is packaged into several hundred higher-order structures termed nucleoids. The abundant mtDNA-binding protein TFAM (transcription factor A, mitochondrial) regulates nucleoid architecture, abundance and segregation. Complete mtDNA depletion profoundly impairs oxidative phosphorylation, triggering calcium-dependent stress signalling and adaptive metabolic responses. However, the cellular responses to mtDNA instability, a physiologically relevant stress observed in many human diseases and ageing, remain poorly defined. Here we show that moderate mtDNA stress elicited by TFAM deficiency engages cytosolic antiviral signalling to enhance the expression of a subset of interferon-stimulated genes. Mechanistically, we find that aberrant mtDNA packaging promotes escape of mtDNA into the cytosol, where it engages the DNA sensor cGAS (also known as MB21D1) and promotes STING (also known as TMEM173)-IRF3-dependent signalling to elevate interferon-stimulated gene expression, potentiate type I interferon responses and confer broad viral resistance. Furthermore, we demonstrate that herpesviruses induce mtDNA stress, which enhances antiviral signalling and type I interferon responses during infection. Our results further demonstrate that mitochondria are central participants in innate immunity, identify mtDNA stress as a cell-intrinsic trigger of antiviral signalling and suggest that cellular monitoring of mtDNA homeostasis cooperates with canonical virus sensing mechanisms to fully engage antiviral innate immunity.


Assuntos
DNA Mitocondrial/metabolismo , Herpesvirus Humano 1/imunologia , Imunidade Inata/imunologia , Estresse Fisiológico , Animais , Linhagem Celular , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Proteínas de Grupo de Alta Mobilidade/deficiência , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Nucleotidiltransferases/metabolismo
9.
Proc Natl Acad Sci U S A ; 112(14): 4423-8, 2015 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-25831514

RESUMO

Prolonged T-cell receptor (TCR) signaling is required for the proliferation of T lymphocytes. Ligation of the TCR activates signaling, but also causes internalization of the TCR from the cell surface. How TCR signaling is sustained for many hours despite lower surface expression is unknown. Using genetic inhibition of endocytosis, we show here that TCR internalization promotes continued TCR signaling and T-lymphocyte proliferation. T-cell-specific deletion of dynamin 2, an essential component of endocytosis, resulted in reduced TCR signaling strength, impaired homeostatic proliferation, and the inability to undergo clonal expansion in vivo. Blocking endocytosis resulted in a failure to maintain mammalian target of rapamycin (mTOR) activity and to stably induce the transcription factor myelocytomatosis oncogene (c-Myc), which led to metabolic stress and a defect in cell growth. Our results support the concept that the TCR can continue to signal after it is internalized from the cell surface, thereby enabling sustained signaling and cell proliferation.


Assuntos
Dinamina II/fisiologia , Endocitose , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Linfócitos T/metabolismo , Animais , Anticorpos Monoclonais/química , Autofagia , Membrana Celular/metabolismo , Proliferação de Células , Regulação da Expressão Gênica , Imunoterapia , Camundongos , Camundongos Knockout , Fosforilação , Proteínas Proto-Oncogênicas c-myc/metabolismo , Linfócitos T/citologia , Serina-Treonina Quinases TOR/metabolismo
10.
Immunol Rev ; 261(1): 157-68, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25123283

RESUMO

Immunity to many intracellular pathogens requires the proliferation, differentiation, and function of CD8(+) cytotoxic T lymphocytes (CTLs). While the majority of effector CTLs die upon clearance of the pathogen, a small proportion of them survive to become long-lived memory CTLs. Memory CTLs can provide protective immunity against re-exposure to the same pathogen and are the principle motivation behind T-cell- based vaccine design. While a large body of cellular immunologic research has proven invaluable to define effector and memory CTLs by their different phenotypes and functions, an emerging focus in the field has been to understand how environmental cues regulate CTL differentiation on a genomic level. Genome-wide studies to profile transcriptional and epigenetic changes during infection have revealed that dynamic changes in DNA methylation patterns and histone modifications accompany transcriptional signatures that define and regulate CTL differentiation states. In this review, we emphasize the importance of epigenetic regulation of CD8(+) T-cell differentiation and the likely role that transcription factors play in this process.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Diferenciação Celular , Metilação de DNA , Epigênese Genética , Regulação da Expressão Gênica/imunologia , Interação Gene-Ambiente , Humanos , Memória Imunológica/genética , Memória Imunológica/imunologia , Infecções/genética , Ativação Transcricional , Vacinas/imunologia
11.
Proc Natl Acad Sci U S A ; 110(17): 6877-82, 2013 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-23572575

RESUMO

Increasing evidence points to a role for the protein quality control in the endoplasmic reticulum (ER) in maintaining intestinal homeostasis. However, the specific role for general ER chaperones in this process remains unknown. Herein, we report that a major ER heat shock protein grp94 interacts with MesD, a critical chaperone for the Wnt coreceptor low-density lipoprotein receptor-related protein 6 (LRP6). Without grp94, LRP6 fails to export from the ER to the cell surface, resulting in a profound loss of canonical Wnt signaling. The significance of this finding is demonstrated in vivo in that grp94 loss causes a rapid and profound compromise in intestinal homeostasis with gut-intrinsic defect in the proliferation of intestinal crypts, compromise of nuclear ß-catenin translocation, loss of crypt-villus structure, and impaired barrier function. Taken together, our work has uncovered the role of grp94 in chaperoning LRP6-MesD in coordinating intestinal homeostasis, placing canonical Wnt-signaling pathway under the direct regulation of the general protein quality control machinery in the ER.


Assuntos
Retículo Endoplasmático/metabolismo , Trato Gastrointestinal/fisiologia , Homeostase/fisiologia , Glicoproteínas de Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Antibacterianos/farmacologia , Western Blotting , Bromodesoxiuridina , Fibroblastos , Imunofluorescência , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Células HEK293 , Proteínas de Choque Térmico HSP90/deficiência , Humanos , Imuno-Histoquímica , Imunoprecipitação , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Plasmídeos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Blood ; 117(26): 7136-44, 2011 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-21576699

RESUMO

The platelet glycoprotein Ib-IX-V complex (GPIb-IX-IV) is the receptor for VWF and is responsible for VWF-mediated platelet activation and aggregation. Loss of the GPIb-IX-V complex is pathogenic for Bernard-soulier Syndrome (BSS), which is characterized by macrothrombocytopenia and impaired platelet function. It remains unclear how the GPIb-IX-V complex is assembled and whether there is a role for a specific molecular chaperone in the process. In the present study, we report that the assembly of the GPIb-IX-V complex depends critically on a molecular chaperone in the endoplasmic reticulum (ER): gp96 (also known as grp94 and HSP90b1). gp96/grp94 deletion in the murine hematopoietic system results in thrombocytopenia, prolonged bleeding time, and giant platelets that are clinically indistinguishable from human BSS. Loss of gp96/grp94 in vivo and in vitro leads to the concomitant reduction in GPIb-IX complex expression due to ER-associated degradation. We further demonstrate that gp96/grp94 binds selectively to the GPIX subunit, but not to gpIbα or gpIbß. Therefore, we identify the platelet GPIX subunit of the GPIb-IX-V complex as an obligate and novel client of gp96/grp94.


Assuntos
Plaquetas/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Animais , Síndrome de Bernard-Soulier/metabolismo , Síndrome de Bernard-Soulier/fisiopatologia , Tempo de Sangramento , Plaquetas/ultraestrutura , Células da Medula Óssea/metabolismo , Células da Medula Óssea/ultraestrutura , Transplante de Medula Óssea/efeitos adversos , Células Cultivadas , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Inibidores Enzimáticos/toxicidade , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/genética , Hematopoese , Hemorragia/metabolismo , Hemorragia/fisiopatologia , Hemostasia , Megacariócitos/metabolismo , Megacariócitos/ultraestrutura , Camundongos , Camundongos Knockout , Agregação Plaquetária , Subunidades Proteicas/metabolismo , RNA Interferente Pequeno , Trombocitopenia/metabolismo , Trombocitopenia/fisiopatologia , Quimeras de Transplante/metabolismo
13.
Blood ; 115(12): 2380-90, 2010 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-19965672

RESUMO

Integrins contribute to lymphopoiesis, whereas Toll-like receptors (TLRs) facilitate the myeloid replenishment during inflammation. The combined role of TLRs and integrin on hematopoiesis remains unclear. gp96 (grp94, HSP90b1) is an endoplasmic reticulum master chaperone for multiple TLRs. We report herein that gp96 is also essential for expression of 14 hematopoietic system-specific integrins. Genetic deletion of gp96 thus enables us to determine the collective roles of gp96, integrins, and TLRs in hematopoiesis. We found that gp96-null hematopoietic stem cells could support long-term myelopoiesis. B- and T-cell development, however, was severely compromised with transitional block from pro-B to pre-B cells and the inability of thymocytes to develop beyond the CD4(-)CD8(-) stage. These defects were cell-intrinsic and could be recapitulated on bone marrow stromal cell culture. Furthermore, defective lymphopoiesis correlated strongly with failure of hematopoietic progenitors to form close contact with stromal cell niche and was not the result of the defect in the assembly of antigen receptor or interleukin-7 signaling. These findings define gp96 as the only known molecular chaperone to specifically regulate T- and B-cell development.


Assuntos
Linfócitos B/fisiologia , Linfopoese/fisiologia , Glicoproteínas de Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Linfócitos T/fisiologia , Animais , Linfócitos B/citologia , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Linhagem Celular , Linhagem da Célula/imunologia , Retículo Endoplasmático/metabolismo , Feminino , Integrinas/metabolismo , Interleucina-7/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Chaperonas Moleculares/genética , Mielopoese/fisiologia , Células Precursoras de Linfócitos B/citologia , Transdução de Sinais/imunologia , Células Estromais/citologia , Células Estromais/fisiologia , Linfócitos T/citologia , Timo/citologia , Timo/fisiologia , Receptores Toll-Like/metabolismo
14.
Nat Commun ; 10(1): 794, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30770827

RESUMO

Prolonged exposure of CD8+ T cells to antigenic stimulation, as in chronic viral infections, leads to a state of diminished function termed exhaustion. We now demonstrate that even during exhaustion there is a subset of functional CD8+ T cells defined by surface expression of SIRPα, a protein not previously reported on lymphocytes. On SIRPα+ CD8+ T cells, expression of co-inhibitory receptors is counterbalanced by expression of co-stimulatory receptors and it is only SIRPα+ cells that actively proliferate, transcribe IFNγ and show cytolytic activity. Furthermore, target cells that express the ligand for SIRPα, CD47, are more susceptible to CD8+ T cell-killing in vivo. SIRPα+ CD8+ T cells are evident in mice infected with Friend retrovirus, LCMV Clone 13, and in patients with chronic HCV infections. Furthermore, therapeutic blockade of PD-L1 to reinvigorate CD8+ T cells during chronic infection expands the cytotoxic subset of SIRPα+ CD8+ T cells.


Assuntos
Infecções por Arenaviridae/imunologia , Linfócitos T CD8-Positivos/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Receptores Imunológicos/imunologia , Animais , Infecções por Arenaviridae/genética , Infecções por Arenaviridae/virologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Feminino , Expressão Gênica/imunologia , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno/imunologia , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/virologia
15.
Protein Cell ; 9(1): 121-129, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28585177

RESUMO

Novel biologics that redirect cytotoxic T lymphocytes (CTLs) to kill tumor cells bearing a tumor associated antigen hold great promise in the clinic. However, the ability to safely and potently target CD3 on CTL toward tumor associated antigens (TAA) expressed on tumor cells remains a challenge of both technology and biology. Herein we describe the use of a Half DVD-Ig format that can redirect CTL to kill tumor cells. Notably, Half DVD-Ig molecules that are monovalent for each specificity demonstrated reduced non-specific CTL activation and conditional CTL activation upon binding to TAA compared to intact tetravalent DVD-Ig molecules that are bivalent for each specificity, while maintaining good drug like properties and appropriate PK properties.


Assuntos
Anticorpos Monoclonais/imunologia , Citotoxicidade Imunológica , Neoplasias/imunologia , Neoplasias/patologia , Linfócitos T Citotóxicos/imunologia , Animais , Anticorpos Biespecíficos/imunologia , Anticorpos Monoclonais/farmacocinética , Complexo CD3/metabolismo , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Feminino , Humanos , Ativação Linfocitária/imunologia , Camundongos SCID , Ratos Sprague-Dawley
16.
Nat Med ; 21(4): 327-34, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25799228

RESUMO

More than 10% of the world's population is chronically infected with HIV, hepatitis C virus (HCV) or hepatitis B virus (HBV), all of which can cause severe disease and death. These viruses persist in part because continuous antigenic stimulation causes the deterioration of virus-specific cytotoxic T lymphocyte (CTL) function and survival. Additionally, antiviral CTLs autonomously suppress their responses to limit immunopathology by upregulating inhibitory receptors such as programmed cell death 1 (PD-1). Identification and blockade of the pathways that induce CTL dysfunction may facilitate the clearance of chronic viral infections. We found that the prostaglandin E2 (PGE2) receptors EP2 and EP4 were upregulated on virus-specific CTLs during chronic lymphocytic choriomeningitis virus (LCMV) infection and suppressed CTL survival and function. We show that the combined blockade of PGE2 and PD-1 signaling was therapeutic in terms of improving viral control and augmenting the numbers of functional virus-specific CTLs. Thus, PGE2 inhibition is both an independent candidate therapeutic target and a promising adjunct therapy to PD-1 blockade for the treatment of HIV and other chronic viral infections.


Assuntos
Dinoprostona/metabolismo , Coriomeningite Linfocítica/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Linfócitos T Citotóxicos/citologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Celecoxib , Sobrevivência Celular , Feminino , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Proto-Oncogênicas/metabolismo , Pirazóis/química , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Transdução de Sinais , Sulfonamidas/química
17.
Elife ; 4: e04851, 2015 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-25569154

RESUMO

T follicular helper cells (Tfh) are crucial for the initiation and maintenance of germinal center (GC) reactions and high affinity, isotype-switched antibody responses. In this study, we demonstrate that direct TGF-ß signaling to CD4 T cells is important for the formation of influenza-specific Tfh cells, GC reactions, and development of isotype-switched, flu-specific antibody responses. Early during infection, TGF-ß signaling suppressed the expression of the high affinity IL-2 receptor α chain (CD25) on virus-specific CD4 T cells, which tempered IL-2 signaling and STAT5 and mammalian target of rapamycin (mTOR) activation in Tfh precursor CD4 T cells. Inhibition of mTOR allowed for the differentiation of Tfh cells in the absence of TGF-ßR signaling, suggesting that TGF-ß insulates Tfh progenitor cells from IL-2-delivered mTOR signals, thereby promoting Tfh differentiation during acute viral infection. These findings identify a new pathway critical for the generation of Tfh cells and humoral responses during respiratory viral infections.


Assuntos
Formação de Anticorpos/imunologia , Switching de Imunoglobulina/imunologia , Pulmão/imunologia , Mucosa/imunologia , Transdução de Sinais , Linfócitos T Auxiliares-Indutores/imunologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Linfócitos B/imunologia , Diferenciação Celular/imunologia , Perfilação da Expressão Gênica , Centro Germinativo/imunologia , Pulmão/patologia , Pulmão/virologia , Camundongos , Mucosa/patologia , Mucosa/virologia , Orthomyxoviridae/fisiologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Especificidade da Espécie , Serina-Treonina Quinases TOR/metabolismo
18.
J Clin Invest ; 124(8): 3455-68, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25003188

RESUMO

During the course of many chronic viral infections, the antiviral T cell response becomes attenuated through a process that is regulated in part by the host. While elevated expression of the immunosuppressive cytokine IL-10 is involved in the suppression of viral-specific T cell responses, the relevant cellular sources of IL-10, as well as the pathways responsible for IL-10 induction, remain unclear. In this study, we traced IL-10 production over the course of chronic lymphocytic choriomeningitis virus (LCMV) infection in an IL-10 reporter mouse line. Using this model, we demonstrated that virus-specific T cells with reduced inflammatory function, particularly Th1 cells, display elevated and sustained IL-10 expression during chronic LCMV infection. Furthermore, ablation of IL-10 from the T cell compartment partially restored T cell function and reduced viral loads in LCMV-infected animals. We found that viral persistence is needed for sustained IL-10 production by Th1 cells and that the transcription factor BLIMP-1 is required for IL-10 expression by Th1 cells. Restimulation of Th1 cells from LCMV-infected mice promoted BLIMP-1 and subsequent IL-10 expression, suggesting that constant antigen exposure likely induces the BLIMP-1/IL-10 pathway during chronic viral infection. Together, these data indicate that effector T cells self-limit their responsiveness during persistent viral infection via an IL-10-dependent negative feedback loop.


Assuntos
Interleucina-10/biossíntese , Coriomeningite Linfocítica/imunologia , Células Th1/imunologia , Fatores de Transcrição/metabolismo , Animais , Doença Crônica , Citocinas/biossíntese , Mediadores da Inflamação/metabolismo , Interleucina-10/genética , Coriomeningite Linfocítica/metabolismo , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/patogenicidade , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fator 1 de Ligação ao Domínio I Regulador Positivo , Receptores de Antígenos de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Células Th1/metabolismo
19.
PLoS One ; 7(6): e39442, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22724016

RESUMO

INTRODUCTION: Basophil has been implicated in anti-parasite defense, allergy and in polarizing T(H)2 response. Mouse model has been commonly used to study basophil function although the difference between human and mouse basophils is underappreciated. As an essential chaperone for multiple Toll-like receptors and integrins in the endoplasmic reticulum, gp96 also participates in general protein homeostasis and in the ER unfolded protein response to ensure cell survival during stress. The roles of gp96 in basophil development are unknown. METHODS: We genetically delete gp96 in mice and examined the expression of gp96 in basophils by Western blot and flow cytometry. We compared the expression pattern of gp96 between human and mouse basophils. RESULTS: We found that gp96 was dispensable for murine basophil development. Moreover, gp96 was cleaved by serine protease(s) in murine but not human basophils leading to accumulation of a nun-functional N-terminal ∼50 kDa fragment and striking induction of the unfolded protein response. The alteration of gp96 was unique to basophils and was not observed in any other cell types including mast cells. We also demonstrated that the ectopic expression of a mouse-specific tryptase mMCP11 does not lead to gp96 cleavage in human basophils. CONCLUSIONS: Our study revealed a remarkable biochemical event of gp96 silencing in murine but not human basophils, highlighting the need for caution in using mouse models to infer the function of basophils in human immune response. Our study also reveals a novel mechanism of shutting down gp96 post-translationally in regulating its function.


Assuntos
Basófilos/metabolismo , Retículo Endoplasmático/metabolismo , Glicoproteínas de Membrana/genética , Chaperonas Moleculares/genética , Processamento Alternativo , Animais , Antígenos/imunologia , Basófilos/imunologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Deleção de Genes , Regulação da Expressão Gênica , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Camundongos , Chaperonas Moleculares/metabolismo , Multimerização Proteica , Proteólise , Especificidade da Espécie , Resposta a Proteínas não Dobradas
20.
Nat Commun ; 1: 79, 2010 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-20865800

RESUMO

Cytosolic HSP90 requires multiple cochaperones in folding client proteins. However, the function of gp96 (HSP90b1, grp94), an HSP90 paralogue in the endoplasmic reticulum (ER), is believed to be independent of cochaperones. Here, we demonstrate that gp96 chaperones multiple Toll-like receptors (TLRs), but not TLR3, in a manner that is dependent on another ER luminal protein, CNPY3. gp96 directly interacts with CNPY3, and the complex dissociates in the presence of adenosine triphosphate (ATP). Genetic disruption of gp96-CNPY3 interaction completely abolishes their TLR chaperone function. Moreover, we demonstrate that TLR9 forms a multimolecular complex with gp96 and CNPY3, and the binding of TLR9 to either molecule requires the presence of the other. We suggest that CNPY3 interacts with the ATP-sensitive conformation of gp96 to promote substrate loading. Our study has thus established CNPY3 as a TLR-specific cochaperone for gp96.


Assuntos
Glicoproteínas de Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Receptores Toll-Like/metabolismo , Animais , Cromatografia em Gel , Cromatografia Líquida , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Immunoblotting , Imunoprecipitação , Glicoproteínas de Membrana/genética , Camundongos , Microscopia de Fluorescência , Chaperonas Moleculares/genética , Reação em Cadeia da Polimerase , Ligação Proteica/genética , Ligação Proteica/fisiologia , Dobramento de Proteína , Espectrometria de Massas em Tandem , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Receptores Toll-Like/química , Receptores Toll-Like/genética , Transdução Genética , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA