Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.424
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 178(3): 585-599.e15, 2019 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-31303383

RESUMO

New opportunities are needed to increase immune checkpoint blockade (ICB) benefit. Whereas the interferon (IFN)γ pathway harbors both ICB resistance factors and therapeutic opportunities, this has not been systematically investigated for IFNγ-independent signaling routes. A genome-wide CRISPR/Cas9 screen to sensitize IFNγ receptor-deficient tumor cells to CD8 T cell elimination uncovered several hits mapping to the tumor necrosis factor (TNF) pathway. Clinically, we show that TNF antitumor activity is only limited in tumors at baseline and in ICB non-responders, correlating with its low abundance. Taking advantage of the genetic screen, we demonstrate that ablation of the top hit, TRAF2, lowers the TNF cytotoxicity threshold in tumors by redirecting TNF signaling to favor RIPK1-dependent apoptosis. TRAF2 loss greatly enhanced the therapeutic potential of pharmacologic inhibition of its interaction partner cIAP, another screen hit, thereby cooperating with ICB. Our results suggest that selective reduction of the TNF cytotoxicity threshold increases the susceptibility of tumors to immunotherapy.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunoterapia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Interferon gama/metabolismo , Estimativa de Kaplan-Meier , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/mortalidade , Neoplasias/terapia , RNA Guia de Cinetoplastídeos/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Transdução de Sinais/efeitos dos fármacos , Fator 2 Associado a Receptor de TNF/deficiência , Fator 2 Associado a Receptor de TNF/genética , Fator de Necrose Tumoral alfa/farmacologia , Receptor de Interferon gama
2.
Cell ; 175(5): 1336-1351.e17, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30318148

RESUMO

As a critical step during innate response, the cytoplasmic ß subunit (IFN-γR2) of interferon-γ receptor (IFN-γR) is induced and translocates to plasma membrane to join α subunit to form functional IFN-γR to mediate IFN-γ signaling. However, the mechanism driving membrane translocation and its significance remain largely unknown. We found, unexpectedly, that mice deficient in E-selectin, an endothelial cell-specific adhesion molecule, displayed impaired innate activation of macrophages upon Listeria monocytogenes infection yet had increased circulating IFN-γ. Inflammatory macrophages from E-selectin-deficient mice had less surface IFN-γR2 and impaired IFN-γ signaling. BTK elicited by extrinsic E-selectin engagement phosphorylates cytoplasmic IFN-γR2, facilitating EFhd2 binding and promoting IFN-γR2 trafficking from Golgi to cell membrane. Our findings demonstrate that membrane translocation of cytoplasmic IFN-γR2 is required to activate macrophage innate response against intracellular bacterial infection, identifying the assembly of functional cytokine receptors on cell membrane as an important layer in innate activation and cytokine signaling.


Assuntos
Selectina E/metabolismo , Imunidade Inata , Receptores de Interferon/metabolismo , Animais , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Membrana Celular/metabolismo , Selectina E/deficiência , Selectina E/genética , Complexo de Golgi/metabolismo , Interferon gama/sangue , Interferon gama/metabolismo , Listeria/patogenicidade , Ativação de Macrófagos , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Transporte Proteico , Células RAW 264.7 , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Transdução de Sinais , Receptor de Interferon gama
3.
Cell ; 169(6): 1130-1141.e11, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28552348

RESUMO

Regulatory T cells (Tregs) are a barrier to anti-tumor immunity. Neuropilin-1 (Nrp1) is required to maintain intratumoral Treg stability and function but is dispensable for peripheral immune tolerance. Treg-restricted Nrp1 deletion results in profound tumor resistance due to Treg functional fragility. Thus, identifying the basis for Nrp1 dependency and the key drivers of Treg fragility could help to improve immunotherapy for human cancer. We show that a high percentage of intratumoral NRP1+ Tregs correlates with poor prognosis in melanoma and head and neck squamous cell carcinoma. Using a mouse model of melanoma where Nrp1-deficient (Nrp1-/-) and wild-type (Nrp1+/+) Tregs can be assessed in a competitive environment, we find that a high proportion of intratumoral Nrp1-/- Tregs produce interferon-γ (IFNγ), which drives the fragility of surrounding wild-type Tregs, boosts anti-tumor immunity, and facilitates tumor clearance. We also show that IFNγ-induced Treg fragility is required for response to anti-PD1, suggesting that cancer therapies promoting Treg fragility may be efficacious.


Assuntos
Carcinoma de Células Escamosas/imunologia , Neoplasias de Cabeça e Pescoço/imunologia , Interferon gama/imunologia , Melanoma/imunologia , Linfócitos T Reguladores/imunologia , Animais , Feminino , Fatores de Transcrição Forkhead , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Masculino , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Neuropilina-1/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Microambiente Tumoral , Receptor de Interferon gama
4.
Nat Immunol ; 20(8): 1035-1045, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31235953

RESUMO

Type III interferon (IFN-λ) is important for innate immune protection at mucosal surfaces and has therapeutic benefit against influenza A virus (IAV) infection. However, the mechanisms by which IFN-λ programs adaptive immune protection against IAV are undefined. Here we found that IFN-λ signaling in dendritic cell (DC) populations was critical for the development of protective IAV-specific CD8+ T cell responses. Mice lacking the IFN-λ receptor (Ifnlr1-/-) had blunted CD8+ T cell responses relative to wild type and exhibited reduced survival after heterosubtypic IAV re-challenge. Analysis of DCs revealed IFN-λ signaling directed the migration and function of CD103+ DCs for development of optimal antiviral CD8+ T cell responses, and bioinformatic analyses identified IFN-λ regulation of a DC IL-10 immunoregulatory network. Thus, IFN-λ serves a critical role in bridging innate and adaptive immunity from lung mucosa to lymph nodes to program DCs to direct effective T cell immunity against IAV.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Vírus da Influenza A/imunologia , Interferon gama/imunologia , Infecções por Orthomyxoviridae/imunologia , Receptores de Interferon/imunologia , Animais , Linhagem Celular , Cães , Feminino , Imunidade Inata/imunologia , Interleucina-10/imunologia , Células Madin Darby de Rim Canino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Interferon/genética , Receptor de Interferon gama
5.
Cell ; 167(2): 397-404.e9, 2016 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-27667683

RESUMO

Antibody blockade of the inhibitory CTLA-4 pathway has led to clinical benefit in a subset of patients with metastatic melanoma. Anti-CTLA-4 enhances T cell responses, including production of IFN-γ, which is a critical cytokine for host immune responses. However, the role of IFN-γ signaling in tumor cells in the setting of anti-CTLA-4 therapy remains unknown. Here, we demonstrate that patients identified as non-responders to anti-CTLA-4 (ipilimumab) have tumors with genomic defects in IFN-γ pathway genes. Furthermore, mice bearing melanoma tumors with knockdown of IFN-γ receptor 1 (IFNGR1) have impaired tumor rejection upon anti-CTLA-4 therapy. These data highlight that loss of the IFN-γ signaling pathway is associated with primary resistance to anti-CTLA-4 therapy. Our findings demonstrate the importance of tumor genomic data, especially IFN-γ related genes, as prognostic information for patients selected to receive treatment with immune checkpoint therapy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígeno CTLA-4/antagonistas & inibidores , Resistencia a Medicamentos Antineoplásicos/genética , Interferon gama/genética , Melanoma/tratamento farmacológico , Receptores de Interferon/genética , Neoplasias Cutâneas/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Citocinas/imunologia , Técnicas de Silenciamento de Genes , Humanos , Ipilimumab , Melanoma/genética , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/genética , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Cutâneas/genética , Linfócitos T/imunologia , Receptor de Interferon gama
6.
Nature ; 633(8029): 451-458, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39112706

RESUMO

Cancer cells frequently alter their lipids to grow and adapt to their environment1-3. Despite the critical functions of lipid metabolism in membrane physiology, signalling and energy production, how specific lipids contribute to tumorigenesis remains incompletely understood. Here, using functional genomics and lipidomic approaches, we identified de novo sphingolipid synthesis as an essential pathway for cancer immune evasion. Synthesis of sphingolipids is surprisingly dispensable for cancer cell proliferation in culture or in immunodeficient mice but required for tumour growth in multiple syngeneic models. Blocking sphingolipid production in cancer cells enhances the anti-proliferative effects of natural killer and CD8+ T cells partly via interferon-γ (IFNγ) signalling. Mechanistically, depletion of glycosphingolipids increases surface levels of IFNγ receptor subunit 1 (IFNGR1), which mediates IFNγ-induced growth arrest and pro-inflammatory signalling. Finally, pharmacological inhibition of glycosphingolipid synthesis synergizes with checkpoint blockade therapy to enhance anti-tumour immune response. Altogether, our work identifies glycosphingolipids as necessary and limiting metabolites for cancer immune evasion.


Assuntos
Linfócitos T CD8-Positivos , Glicoesfingolipídeos , Receptor de Interferon gama , Interferon gama , Proteínas Proto-Oncogênicas p21(ras) , Transdução de Sinais , Evasão Tumoral , Animais , Glicoesfingolipídeos/metabolismo , Glicoesfingolipídeos/biossíntese , Camundongos , Humanos , Interferon gama/metabolismo , Interferon gama/imunologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Feminino , Linhagem Celular Tumoral , Neoplasias/imunologia , Neoplasias/genética , Neoplasias/patologia , Neoplasias/metabolismo , Proliferação de Células , Receptores de Interferon/metabolismo , Receptores de Interferon/genética , Receptores de Interferon/deficiência , Masculino , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Esfingolipídeos/metabolismo , Esfingolipídeos/biossíntese , Evasão da Resposta Imune , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Camundongos Endogâmicos C57BL
7.
Immunity ; 50(5): 1172-1187.e7, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-31076359

RESUMO

Although viral infections elicit robust interferon-γ (IFN-γ) and long-lived antibody-secreting cell (ASC) responses, the roles for IFN-γ and IFN-γ-induced transcription factors (TFs) in ASC development are unclear. We showed that B cell intrinsic expression of IFN-γR and the IFN-γ-induced TF T-bet were required for T-helper 1 cell-induced differentiation of B cells into ASCs. IFN-γR signaling induced Blimp1 expression in B cells but also initiated an inflammatory gene program that, if not restrained, prevented ASC formation. T-bet did not affect Blimp1 upregulation in IFN-γ-activated B cells but instead regulated chromatin accessibility within the Ifng and Ifngr2 loci and repressed the IFN-γ-induced inflammatory gene program. Consistent with this, B cell intrinsic T-bet was required for formation of long-lived ASCs and secondary ASCs following viral, but not nematode, infection. Therefore, T-bet facilitates differentiation of IFN-γ-activated inflammatory effector B cells into ASCs in the setting of IFN-γ-, but not IL-4-, induced inflammatory responses.


Assuntos
Linfócitos B/imunologia , Interferon gama/imunologia , Receptores de Interferon/metabolismo , Proteínas com Domínio T/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Células Produtoras de Anticorpos/imunologia , Linfócitos B/citologia , Diferenciação Celular/imunologia , Células Cultivadas , Cromatina/metabolismo , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nematospiroides dubius/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Fator 1 de Ligação ao Domínio I Regulador Positivo/biossíntese , Infecções por Strongylida/imunologia , Infecções por Strongylida/parasitologia , Proteínas com Domínio T/genética , Receptor de Interferon gama
8.
PLoS Genet ; 20(6): e1011162, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38885280

RESUMO

Very little is known about the process of meiosis in the apicomplexan parasite Cryptosporidium despite the essentiality of sex in its life cycle. Most cell lines only support asexual growth of Cryptosporidium parvum (C. parvum), but stem cell derived intestinal epithelial cells grown under air-liquid interface (ALI) conditions support the sexual cycle. To examine chromosomal dynamics during meiosis in C. parvum, we generated two transgenic lines of parasites that were fluorescently tagged with mCherry or GFP on chromosomes 1 or 5, respectively. Infection of ALI cultures or Ifngr1-/- mice with mCherry and GFP parasites resulted in cross-fertilization and the formation of "yellow" oocysts, which contain 4 haploid sporozoites that are the product of meiosis. Recombinant oocysts from the F1 generation were purified and used to infect HCT-8 cultures, and phenotypes of the progeny were observed by microscopy. All possible phenotypes predicted by independent segregation were represented equally (~25%) in the population, indicating that C. parvum chromosomes exhibit a Mendelian inheritance pattern. The most common pattern observed from the outgrowth of single oocysts included all possible parental and recombinant phenotypes derived from a single meiotic event, suggesting a high rate of crossover. To estimate the frequency of crossover, additional loci on chromosomes 1 and 5 were tagged and used to monitor intrachromosomal crosses in Ifngr1-/- mice. Both chromosomes showed a high frequency of crossover compared to other apicomplexans with map distances (i.e., 1% recombination) of 3-12 kb. Overall, a high recombination rate may explain many unique characteristics observed in Cryptosporidium spp. such as high rates of speciation, wide variation in host range, and rapid evolution of host-specific virulence factors.


Assuntos
Criptosporidiose , Cryptosporidium parvum , Meiose , Oocistos , Recombinação Genética , Animais , Cryptosporidium parvum/genética , Camundongos , Criptosporidiose/parasitologia , Criptosporidiose/genética , Meiose/genética , Humanos , Receptores de Interferon/genética , Receptor de Interferon gama , Segregação de Cromossomos/genética , Esporozoítos/genética , Camundongos Knockout , Fenótipo
9.
PLoS Pathog ; 20(5): e1012020, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38743761

RESUMO

Scrub typhus is an acute febrile disease due to Orientia tsutsugamushi (Ot) infection and can be life-threatening with organ failure, hemorrhage, and fatality. Yet, little is known as to how the host reacts to Ot bacteria at early stages of infection; no reports have addressed the functional roles of type I versus type II interferon (IFN) responses in scrub typhus. In this study, we used comprehensive intradermal (i.d.) inoculation models and two clinically predominant Ot strains (Karp and Gilliam) to uncover early immune events. Karp infection induced sequential expression of Ifnb and Ifng in inflamed skin and draining lymph nodes at days 1 and 3 post-infection. Using double Ifnar1-/-Ifngr1-/- and Stat1-/- mice, we found that deficiency in IFN/STAT1 signaling resulted in lethal infection with profound pathology and skin eschar lesions, which resembled to human scrub typhus. Further analyses demonstrated that deficiency in IFN-γ, but not IFN-I, resulted in impaired NK cell and macrophage activation and uncontrolled bacterial growth and dissemination, leading to metabolic dysregulation, excessive inflammatory cell infiltration, and exacerbated tissue damage. NK cells were found to be the major cellular source of innate IFN-γ, contributing to the initial Ot control in the draining lymph nodes. In vitro studies with dendritic cell cultures revealed a superior antibacterial effect offered by IFN-γ than IFN-ß. Comparative in vivo studies with Karp- and Gilliam-infection revealed a crucial role of IFN-γ signaling in protection against progression of eschar lesions and Ot infection lethality. Additionally, our i.d. mouse models of lethal infection with eschar lesions are promising tools for immunological study and vaccine development for scrub typhus.


Assuntos
Interferon gama , Orientia tsutsugamushi , Tifo por Ácaros , Transdução de Sinais , Animais , Camundongos , Modelos Animais de Doenças , Interferon gama/metabolismo , Interferon gama/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Orientia tsutsugamushi/imunologia , Tifo por Ácaros/imunologia , Tifo por Ácaros/microbiologia , Pele/microbiologia , Pele/patologia , Pele/imunologia , Fator de Transcrição STAT1/metabolismo , Receptor de Interferon gama/genética , Receptor de Interferon gama/metabolismo
10.
PLoS Pathog ; 20(5): e1011820, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38718306

RESUMO

The production of IFN-γ is crucial for control of multiple enteric infections, but its impact on intestinal epithelial cells (IEC) is not well understood. Cryptosporidium parasites exclusively infect epithelial cells and the ability of interferons to activate the transcription factor STAT1 in IEC is required for parasite clearance. Here, the use of single cell RNA sequencing to profile IEC during infection revealed an increased proportion of mid-villus enterocytes during infection and induction of IFN-γ-dependent gene signatures that was comparable between uninfected and infected cells. These analyses were complemented by in vivo studies, which demonstrated that IEC expression of the IFN-γ receptor was required for parasite control. Unexpectedly, treatment of Ifng-/- mice with IFN-γ showed the IEC response to this cytokine correlates with a delayed reduction in parasite burden but did not affect parasite development. These data sets provide insight into the impact of IFN-γ on IEC and suggest a model in which IFN-γ signalling to uninfected enterocytes is important for control of Cryptosporidium.


Assuntos
Criptosporidiose , Interferon gama , Mucosa Intestinal , Camundongos Knockout , Animais , Interferon gama/metabolismo , Interferon gama/imunologia , Criptosporidiose/imunologia , Criptosporidiose/parasitologia , Camundongos , Mucosa Intestinal/parasitologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/imunologia , Cryptosporidium , Células Epiteliais/parasitologia , Células Epiteliais/metabolismo , Células Epiteliais/imunologia , Enterócitos/parasitologia , Enterócitos/metabolismo , Enterócitos/imunologia , Camundongos Endogâmicos C57BL , Receptor de Interferon gama , Fator de Transcrição STAT1/metabolismo , Receptores de Interferon/metabolismo , Receptores de Interferon/genética , Transdução de Sinais
11.
Nat Immunol ; 15(4): 333-42, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24608040

RESUMO

Diverse cellular responses to external cues are controlled by a small number of signal-transduction pathways, but how the specificity of functional outcomes is achieved remains unclear. Here we describe a mechanism for signal integration based on the functional coupling of two distinct signaling pathways widely used in leukocytes: the ITAM pathway and the Jak-STAT pathway. Through the use of the receptor for interferon-γ (IFN-γR) and the ITAM adaptor Fcγ as an example, we found that IFN-γ modified responses of the phagocytic antibody receptor FcγRI (CD64) to specify cell-autonomous antimicrobial functions. Unexpectedly, we also found that in peritoneal macrophages, IFN-γR itself required tonic signaling from Fcγ through the kinase PI(3)K for the induction of a subset of IFN-γ-specific antimicrobial functions. Our findings may be generalizable to other ITAM and Jak-STAT signaling pathways and may help explain signal integration by those pathways.


Assuntos
Motivo de Ativação do Imunorreceptor Baseado em Tirosina/imunologia , Janus Quinase 2/metabolismo , Listeriose/imunologia , Macrófagos/imunologia , Receptor Cross-Talk/imunologia , Fator de Transcrição STAT1/metabolismo , Animais , Carga Bacteriana , Células Cultivadas , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Motivo de Ativação do Imunorreceptor Baseado em Tirosina/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Janus Quinase 2/genética , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Fagocitose/genética , Inibidores de Fosfoinositídeo-3 Quinase , Engenharia de Proteínas , Receptores de IgG/genética , Receptores de IgG/metabolismo , Receptores de Interferon/metabolismo , Fator de Transcrição STAT1/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Ativação Transcricional/efeitos dos fármacos , Receptor de Interferon gama
12.
J Virol ; 98(5): e0049324, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38578092

RESUMO

CD4+ T cells play a key role in γ-herpesvirus infection control. However, the mechanisms involved are unclear. Murine herpesvirus type 4 (MuHV-4) allows relevant immune pathways to be dissected experimentally in mice. In the lungs, it colonizes myeloid cells, which can express MHC class II (MHCII), and type 1 alveolar epithelial cells (AEC1), which lack it. Nevertheless, CD4+ T cells can control AEC1 infection, and this control depends on MHCII expression in myeloid cells. Interferon-gamma (IFNγ) is a major component of CD4+ T cell-dependent MuHV-4 control. Here, we show that the action of IFNγ is also indirect, as CD4+ T cell-mediated control of AEC1 infection depended on IFNγ receptor (IFNγR1) expression in CD11c+ cells. Indirect control also depended on natural killer (NK) cells. Together, the data suggest that the activation of MHCII+ CD11c+ antigen-presenting cells is key to the CD4+ T cell/NK cell protection axis. By contrast, CD8+ T cell control of AEC1 infection appeared to operate independently. IMPORTANCE: CD4+ T cells are critical for the control of gamma-herpesvirus infection; they act indirectly, by recruiting natural killer (NK) cells to attack infected target cells. Here, we report that the CD4+ T cell/NK cell axis of gamma-herpesvirus control requires interferon-γ engagement of CD11c+ dendritic cells. This mechanism of CD4+ T cell control releases the need for the direct engagement of CD4+ T cells with virus-infected cells and may be a common strategy for host control of immune-evasive pathogens.


Assuntos
Linfócitos T CD4-Positivos , Infecções por Herpesviridae , Interferon gama , Células Matadoras Naturais , Receptores de Interferon , Rhadinovirus , Animais , Linfócitos T CD4-Positivos/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Camundongos , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Células Matadoras Naturais/imunologia , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Rhadinovirus/imunologia , Camundongos Endogâmicos C57BL , Receptor de Interferon gama , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Células Epiteliais Alveolares/imunologia , Células Epiteliais Alveolares/virologia , Linfócitos T CD8-Positivos/imunologia , Antígeno CD11c/metabolismo , Antígeno CD11c/imunologia , Pulmão/imunologia , Pulmão/virologia
13.
Immunity ; 44(2): 368-79, 2016 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-26885859

RESUMO

Humans that are heterozygous for the common S180L polymorphism in the Toll-like receptor (TLR) adaptor Mal (encoded by TIRAP) are protected from a number of infectious diseases, including tuberculosis (TB), whereas those homozygous for the allele are at increased risk. The reason for this difference in susceptibility is not clear. We report that Mal has a TLR-independent role in interferon-gamma (IFN-γ) receptor signaling. Mal-dependent IFN-γ receptor (IFNGR) signaling led to mitogen-activated protein kinase (MAPK) p38 phosphorylation and autophagy. IFN-γ signaling via Mal was required for phagosome maturation and killing of intracellular Mycobacterium tuberculosis (Mtb). The S180L polymorphism, and its murine equivalent S200L, reduced the affinity of Mal for the IFNGR, thereby compromising IFNGR signaling in macrophages and impairing responses to TB. Our findings highlight a role for Mal outside the TLR system and imply that genetic variation in TIRAP may be linked to other IFN-γ-related diseases including autoimmunity and cancer.


Assuntos
Interferon gama/metabolismo , Macrófagos/fisiologia , Glicoproteínas de Membrana/metabolismo , Mycobacterium tuberculosis/imunologia , Receptores de Interleucina-1/metabolismo , Tuberculose Pulmonar/imunologia , Animais , Autofagia/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Genótipo , Células HEK293 , Humanos , Imunidade Inata/genética , Sistema de Sinalização das MAP Quinases/genética , Macrófagos/microbiologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Polimorfismo Genético , Ligação Proteica/genética , RNA Interferente Pequeno/genética , Receptores de Interferon/metabolismo , Receptores de Interleucina-1/genética , Tuberculose Pulmonar/genética , Receptor de Interferon gama
14.
Nature ; 567(7746): 56-60, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30814731

RESUMO

The cytokine interferon-γ (IFNγ) is a central coordinator of innate and adaptive immunity, but its highly pleiotropic actions have diminished its prospects for use as an immunotherapeutic agent. Here, we took a structure-based approach to decoupling IFNγ pleiotropy. We engineered an affinity-enhanced variant of the ligand-binding chain of the IFNγ receptor IFNγR1, which enabled us to determine the crystal structure of the complete hexameric (2:2:2) IFNγ-IFNγR1-IFNγR2 signalling complex at 3.25 Å resolution. The structure reveals the mechanism underlying deficits in IFNγ responsiveness in mycobacterial disease syndrome resulting from a T168N mutation in IFNγR2, which impairs assembly of the full signalling complex. The topology of the hexameric complex offers a blueprint for engineering IFNγ variants to tune IFNγ receptor signalling output. Unexpectedly, we found that several partial IFNγ agonists exhibited biased gene-expression profiles. These biased agonists retained the ability to induce upregulation of major histocompatibility complex class I antigen expression, but exhibited impaired induction of programmed death-ligand 1 expression in a wide range of human cancer cell lines, offering a route to decoupling immunostimulatory and immunosuppressive functions of IFNγ for therapeutic applications.


Assuntos
Desenho de Fármacos , Interferon gama/agonistas , Interferon gama/imunologia , Receptores de Interferon/química , Receptores de Interferon/metabolismo , Antígeno B7-H1/biossíntese , Antígeno B7-H1/genética , Linhagem Celular Tumoral , Agonismo Parcial de Drogas , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe I/biossíntese , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Interferon gama/química , Interferon gama/genética , Ligantes , Modelos Moleculares , Mutação , Infecções por Mycobacterium/genética , Infecções por Mycobacterium/imunologia , Estabilidade Proteica , Receptores de Interferon/genética , Transdução de Sinais , Relação Estrutura-Atividade , Receptor de Interferon gama
15.
Mol Cell ; 66(5): 635-647.e7, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28575659

RESUMO

Immune cells constantly survey the host for pathogens or tumors and secrete cytokines to alert surrounding cells of these threats. In vivo, activated immune cells secrete cytokines for several hours, yet an acute immune reaction occurs over days. Given these divergent timescales, we addressed how cytokine-responsive cells translate brief cytokine exposure into phenotypic changes that persist over long timescales. We studied melanoma cell responses to transient exposure to the cytokine interferon γ (IFNγ) by combining a systems-scale analysis of gene expression dynamics with computational modeling and experiments. We discovered that IFNγ is captured by phosphatidylserine (PS) on the surface of viable cells both in vitro and in vivo then slowly released to drive long-term transcription of cytokine-response genes. This mechanism introduces an additional function for PS in dynamically regulating inflammation across diverse cancer and primary cell types and has potential to usher in new immunotherapies targeting PS and inflammatory pathways.


Assuntos
Comunicação Celular , Mediadores da Inflamação/metabolismo , Inflamação/metabolismo , Interferon gama/metabolismo , Linfócitos do Interstício Tumoral/metabolismo , Melanoma Experimental/metabolismo , Fosfatidilserinas/metabolismo , Linfócitos T/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Animais , Linhagem Celular Tumoral , Técnicas de Cocultura , Biologia Computacional , Simulação por Computador , Bases de Dados Genéticas , Feminino , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Interferon gama/imunologia , Interleucina-12/imunologia , Interleucina-12/metabolismo , Interleucina-23/imunologia , Interleucina-23/metabolismo , Janus Quinases/metabolismo , Ativação Linfocitária , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Masculino , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilserinas/imunologia , Fosforilação , Células RAW 264.7 , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T/patologia , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/imunologia , Neoplasias da Glândula Tireoide/patologia , Fatores de Tempo , Transcrição Gênica , Receptor de Interferon gama
16.
Cell Mol Life Sci ; 81(1): 380, 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39222120

RESUMO

The cytokine IFNγ is a principal effector of macrophage activation and immune resistance to mycobacterial infection; however, pathogenic mycobacteria are capable of surviving in IFNγ-activated macrophages by largely unknown mechanisms. In this study, we find that pathogenic mycobacteria, including M. bovis BCG and M. tuberculosis can sense IFNγ to promote their proliferative activity and virulence phenotype. Moreover, interaction with the host intracellular environment increases the susceptibility of mycobacteria to IFNγ through upregulating expression of mmpL10, a mycobacterial IFNγ receptor, thereby facilitating IFNγ-dependent survival and growth of mycobacteria in macrophages. Transmission electron microscopy analysis reveals that IFNγ triggers the secretion of extracellular vesicles, an essential virulence strategy of intracellular mycobacteria, while proteomics identifies numerous pivotal IFNγ-induced effectors required for mycobacterial infection in macrophages. Our study suggests that sensing host IFNγ is a crucial virulence mechanism used by pathogenic mycobacteria to survive and proliferate inside macrophages.


Assuntos
Interferon gama , Macrófagos , Camundongos Endogâmicos C57BL , Mycobacterium tuberculosis , Interferon gama/metabolismo , Interferon gama/imunologia , Macrófagos/microbiologia , Macrófagos/metabolismo , Macrófagos/imunologia , Animais , Camundongos , Mycobacterium tuberculosis/patogenicidade , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/metabolismo , Mycobacterium bovis/imunologia , Mycobacterium bovis/metabolismo , Humanos , Interações Hospedeiro-Patógeno/imunologia , Virulência , Receptores de Interferon/metabolismo , Receptores de Interferon/genética , Receptor de Interferon gama , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/imunologia , Ativação de Macrófagos , Infecções por Mycobacterium/microbiologia , Infecções por Mycobacterium/imunologia , Infecções por Mycobacterium/metabolismo , Infecções por Mycobacterium/patologia
17.
J Biol Chem ; 299(8): 104990, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37392850

RESUMO

Cycloartenyl ferulate (CF) is abundant in brown rice with multiple biologic functions. It has been reported to possess antitumor activity; however, the related mechanism of action of CF has not been clarified. Herein, we unexpectedly uncover the immunological regulation effects of CF and its molecular mechanism. We discovered that CF directly enhanced the killing capacity of natural killer (NK) cells for various cancer cells in vitro. In vivo, CF also improved cancer surveillance in mouse models of lymphoma clearance and metastatic melanoma dependent on NK cells. In addition, CF promoted anticancer efficacy of the anti-PD1 antibody with improvement of tumor immune microenvironment. Mechanistically, we first unveiled that CF acted on the canonical JAK1/2-STAT1 signaling pathway to enhance the immunity of the NK cells by selectively binding to interferon γ receptor 1. Collectively, our results indicate that CF is a promising immunoregulation agent worthy of attention in clinical application in the future. Due to broad biological significance of interferon γ, our findings also provide a capability to understand the diverse functions of CF.


Assuntos
Ácidos Cumáricos , Células Matadoras Naturais , Neoplasias , Receptores de Interferon , Animais , Camundongos , Interferon gama/imunologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Neoplasias/imunologia , Microambiente Tumoral , Ácidos Cumáricos/farmacologia , Receptores de Interferon/imunologia , Receptor de Interferon gama
18.
J Am Chem Soc ; 146(26): 17691-17699, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38888290

RESUMO

Nonproteinogenic amino acids, including d-α-, ß-, and γ-amino acids, present in bioactive peptides play pivotal roles in their biochemical activities and proteolytic stabilities. d-α-Amino acids (dαAA) are widely used building blocks that can enhance the proteolytic stability. Cyclic ß2,3-amino acids (cßAA), for instance, can fold peptides into rigid secondary structures, improving the binding affinity and proteolytic stability. Cyclic γ2,4-amino acids (cγAA) are recently highlighted as rigid residues capable of preventing the proteolysis of flanking residues. Simultaneous incorporation of all dαAA, cßAA, and cγAA into a peptide is expected to yield l-α/d-α/ß/γ-hybrid peptides with improved stability and potency. Despite challenges in the ribosomal incorporation of multiple nonproteinogenic amino acids, our engineered tRNAPro1E2 successfully reaches such a difficulty. Here, we report the ribosomal synthesis of macrocyclic l-α/d-α/ß/γ-hybrid peptide libraries and their application to in vitro selection against interferon gamma receptor 1 (IFNGR1). One of the resulting l-α/d-α/ß/γ-hybrid peptides, IB1, exhibited remarkable inhibitory activity against the IFN-γ/IFNGR1 protein-protein interaction (PPI) (IC50 = 12 nM), primarily attributed to the presence of a cßAA in the sequence. Additionally, cγAAs and dαAAs in the resulting peptides contributed to their serum stability. Furthermore, our peptides effectively inhibit IFN-γ/IFNGR1 PPI at the cellular level (best IC50 = 0.75 µM). Altogether, our platform expands the chemical space available for exploring peptides with high activity and stability, thereby enhancing their potential for drug discovery.


Assuntos
Receptor de Interferon gama , Interferon gama , Receptores de Interferon , Interferon gama/metabolismo , Receptores de Interferon/metabolismo , Receptores de Interferon/química , Humanos , Ligação Proteica , Compostos Macrocíclicos/química , Compostos Macrocíclicos/farmacologia , Peptídeos/química , Peptídeos/farmacologia , Peptídeos Cíclicos/química , Peptídeos Cíclicos/farmacologia , Peptídeos Cíclicos/metabolismo
19.
J Autoimmun ; 148: 103279, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38972102

RESUMO

B cells of people with multiple sclerosis (MS) are more responsive to IFN-γ, corresponding to their brain-homing potential. We studied how a coding single nucleotide polymorphism (SNP) in IFNGR2 (rs9808753) co-operates with Epstein-Barr virus (EBV) infection as MS risk factors to affect the IFN-γ signaling pathway in human B cells. In both cell lines and primary cells, EBV infection positively associated with IFN-γ receptor expression and STAT1 phosphorylation. The IFNGR2 risk SNP selectively promoted downstream signaling via STAT1, particularly in transitional B cells. Altogether, EBV and the IFNGR2 risk SNP independently amplify IFN-γ signaling, potentially driving B cells to enter the MS brain.


Assuntos
Linfócitos B , Infecções por Vírus Epstein-Barr , Receptor de Interferon gama , Interferon gama , Esclerose Múltipla , Polimorfismo de Nucleotídeo Único , Receptores de Interferon , Transdução de Sinais , Humanos , Esclerose Múltipla/genética , Esclerose Múltipla/imunologia , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Interferon gama/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/genética , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT1/genética , Predisposição Genética para Doença , Herpesvirus Humano 4 , Feminino , Masculino , Fosforilação
20.
Immunity ; 42(1): 108-22, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25607461

RESUMO

The probabilistic expression of cytokine genes in differentiated T helper (Th) cell populations remains ill defined. By single-cell analyses and mathematical modeling, we show that one stimulation featured stable cytokine nonproducers as well as stable producers with wide cell-to-cell variability in the magnitude of expression. Focusing on interferon-γ (IFN-γ) expression by Th1 cells, mathematical modeling predicted that this behavior reflected different cell-intrinsic capacities and not mere gene-expression noise. In vivo, Th1 cells sort purified by secreted IFN-γ amounts preserved a quantitative memory for both probability and magnitude of IFN-γ re-expression for at least 1 month. Mechanistically, this memory resulted from quantitatively distinct transcription of individual alleles and was controlled by stable expression differences of the Th1 cell lineage-specifying transcription factor T-bet. Functionally, Th1 cells with graded IFN-γ production competence differentially activated Salmonella-infected macrophages for bacterial killing. Thus, individual Th cells commit to produce distinct amounts of a given cytokine, thereby generating functional intrapopulation heterogeneity.


Assuntos
Interferon gama/metabolismo , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Macrófagos/imunologia , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Células Th1/imunologia , Animais , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Contagem de Colônia Microbiana , Regulação da Expressão Gênica , Memória Imunológica , Interferon gama/genética , Interferon gama/imunologia , Ativação Linfocitária , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Teóricos , Receptores de Interferon/genética , Análise de Célula Única , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Células Th1/virologia , Carga Viral , Receptor de Interferon gama
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA