Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 17.065
Filtrar
1.
BMC Genomics ; 25(1): 650, 2024 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-38951796

RESUMO

BACKGROUND: Viperin, also known as radical S-adenosyl-methionine domain containing protein 2 (RSAD2), is an interferon-inducible protein that is involved in the innate immune response against a wide array of viruses. In mammals, Viperin exerts its antiviral function through enzymatic conversion of cytidine triphosphate (CTP) into its antiviral analog ddhCTP as well as through interactions with host proteins involved in innate immune signaling and in metabolic pathways exploited by viruses during their life cycle. However, how Viperin modulates the antiviral response in fish remains largely unknown. RESULTS: For this purpose, we developed a fathead minnow (Pimephales promelas) clonal cell line in which the unique viperin gene has been knocked out by CRISPR/Cas9 genome-editing. In order to decipher the contribution of fish Viperin to the antiviral response and its regulatory role beyond the scope of the innate immune response, we performed a comparative RNA-seq analysis of viperin-/- and wildtype cell lines upon stimulation with recombinant fathead minnow type I interferon. CONCLUSIONS: Our results revealed that Viperin does not exert positive feedback on the canonical type I IFN but acts as a negative regulator of the inflammatory response by downregulating specific pro-inflammatory genes and upregulating repressors of the NF-κB pathway. It also appeared to play a role in regulating metabolic processes, including one carbon metabolism, bone formation, extracellular matrix organization and cell adhesion.


Assuntos
Cyprinidae , Inflamação , Animais , Cyprinidae/metabolismo , Cyprinidae/genética , Inflamação/metabolismo , Inflamação/genética , Imunidade Inata , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Linhagem Celular , Sistemas CRISPR-Cas , Interferon Tipo I/metabolismo , Edição de Genes , Regulação da Expressão Gênica
2.
Sci Immunol ; 9(97): eadm8185, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38968338

RESUMO

The past 20 years have seen the definition of human monogenic disorders and their autoimmune phenocopies underlying either defective or enhanced type I interferon (IFN) activity. These disorders delineate the impact of type I IFNs in natural conditions and demonstrate that only a narrow window of type I IFN activity is beneficial. Insufficient type I IFN predisposes humans to life-threatening viral diseases (albeit unexpectedly few) with a central role in immunity to respiratory and cerebral viral infection. Excessive type I IFN, perhaps counterintuitively, appears to underlie a greater number of autoinflammatory and/or autoimmune conditions known as type I interferonopathies, whose study has revealed multiple molecular programs involved in the induction of type I IFN signaling. These observations suggest that the manipulation of type I IFN activity to within a physiological range may be clinically relevant for the prevention and treatment of viral and inflammatory diseases.


Assuntos
Interferon Tipo I , Humanos , Interferon Tipo I/imunologia , Animais , Viroses/imunologia , Doenças Autoimunes/imunologia , Transdução de Sinais/imunologia
3.
Int J Rheum Dis ; 27(7): e15256, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38982864

RESUMO

The cyclic GMP-AMP synthase (cGAS), a prominent intracellular DNA sensor in mammalian cells, controls the innate immune response and the stimulator of interferon genes (STING)-mediated synthesis of pro-inflammatory cytokines, such as type-I interferon (IFN-I). For decades, IFN-I has been hypothesized to be essential in the development of systemic lupus erythematosus (SLE), a chronic multisystem autoimmunity characterized by immune complex (IC) deposition in small vessels. Recent findings revealed that the activation of the cGAS-STING pathway by self-DNA would propagate the autoimmune responses via upregulating IFN-I production in SLE. In this review, we aimed to provide a comprehensive outlook of the role of the cGAS-STING pathway in SLE pathobiology, as well as, a better understanding of current therapeutic opportunities targeting this axis.


Assuntos
Lúpus Eritematoso Sistêmico , Proteínas de Membrana , Nucleotidiltransferases , Transdução de Sinais , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Nucleotidiltransferases/metabolismo , Proteínas de Membrana/metabolismo , Animais , Autoimunidade , Interferon Tipo I/metabolismo , Interferon Tipo I/imunologia , Terapia de Alvo Molecular , Imunidade Inata
4.
Cells ; 13(13)2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38995014

RESUMO

PD-1 (Programmed cell death protein 1) regulates the metabolic reprogramming of myeloid-derived suppressor cells and myeloid cell differentiation, as well as the type I interferon (IFN-I) signaling pathway in myeloid cells in the tumor microenvironment. PD-1, therefore, is a key inhibitory receptor in myeloid cells. However, the regulation of PD-1 expression in myeloid cells is unknown. We report that the expression level of PDCD1, the gene that encodes the PD-1 protein, is positively correlated with the levels of IFNB1 and IFNAR1 in myeloid cells in human colorectal cancer. Treatment of mouse myeloid cell lines with recombinant IFNß protein elevated PD-1 expression in myeloid cells in vitro. Knocking out IFNAR1, the gene that encodes the IFN-I-specific receptor, diminished the inductive effect of IFNß on PD-1 expression in myeloid cells in vitro. Treatment of tumor-bearing mice with a lipid nanoparticle-encapsulated IFNß-encoding plasmid (IFNBCOL01) increased IFNß expression, resulting in elevated PD-1 expression in tumor-infiltrating myeloid cells. At the molecular level, we determined that IFNß activates STAT1 (signal transducer and activator of transcription 1) and IRFs (interferon regulatory factors) in myeloid cells. Analysis of the cd279 promoter identified IRF2-binding consensus sequence elements. ChIP (chromatin immunoprecipitation) analysis determined that the pSTAT1 directly binds to the irf2 promoter and that IRF2 directly binds to the cd279 promoter in myeloid cells in vitro and in vivo. In colon cancer patients, the expression levels of STAT1, IRF2 and PDCD1 are positively correlated in tumor-infiltrating myeloid cells. Our findings determine that IFNß activates PD-1 expression at least in part by an autocrine mechanism via the stimulation of the pSTAT1-IRF2 axis in myeloid cells.


Assuntos
Fator Regulador 2 de Interferon , Células Mieloides , Receptor de Morte Celular Programada 1 , Fator de Transcrição STAT1 , Transdução de Sinais , Células Mieloides/metabolismo , Células Mieloides/efeitos dos fármacos , Animais , Humanos , Fator de Transcrição STAT1/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Receptor de Morte Celular Programada 1/genética , Camundongos , Fator Regulador 2 de Interferon/metabolismo , Fator Regulador 2 de Interferon/genética , Transdução de Sinais/efeitos dos fármacos , Interferon Tipo I/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Receptor de Interferon alfa e beta/genética , Interferon beta/metabolismo , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Camundongos Endogâmicos C57BL
5.
Int J Mol Sci ; 25(13)2024 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-39000569

RESUMO

Regulation of neuroinflammation is critical for maintaining central nervous system (CNS) homeostasis and holds therapeutic promise in autoimmune diseases such as multiple sclerosis (MS). Previous studies have highlighted the significance of selective innate signaling in triggering anti-inflammatory mechanisms, which play a protective role in an MS-like disease, experimental autoimmune encephalomyelitis (EAE). However, the individual intra-CNS administration of specific innate receptor ligands or agonists, such as for toll-like receptor 7 (TLR7) and nucleotide-binding oligomerization-domain-containing protein 2 (NOD2), failed to elicit the desired anti-inflammatory response in EAE. In this study, we investigated the potential synergistic effect of targeting both TLR7 and NOD2 simultaneously to prevent EAE progression. Our findings demonstrate that simultaneous intrathecal administration of NOD2- and TLR7-agonists led to synergistic induction of Type I IFN (IFN I) and effectively suppressed EAE in an IFN I-dependent manner. Suppression of EAE was correlated with a significant decrease in the infiltration of monocytes, granulocytes, and natural killer cells, reduced demyelination, and downregulation of IL-1ß, CCL2, and IFNγ gene expression in the spinal cord. These results underscore the therapeutic promise of concurrently targeting the TLR7 and NOD2 pathways in alleviating neuroinflammation associated with MS, paving the way for novel and more efficacious treatment strategies.


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Proteína Adaptadora de Sinalização NOD2 , Receptor 7 Toll-Like , Animais , Receptor 7 Toll-Like/metabolismo , Receptor 7 Toll-Like/agonistas , Proteína Adaptadora de Sinalização NOD2/metabolismo , Proteína Adaptadora de Sinalização NOD2/genética , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Imunidade Inata/efeitos dos fármacos , Feminino , Medula Espinal/metabolismo , Medula Espinal/patologia , Medula Espinal/efeitos dos fármacos , Glicoproteínas de Membrana/metabolismo , Interferon Tipo I/metabolismo , Transdução de Sinais/efeitos dos fármacos
6.
Front Immunol ; 15: 1428232, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39040112

RESUMO

In the decades since the discovery, Type I interferon (IFN-I) has been intensively studied for their antiviral activity. However, increasing evidences suggest that it may also play an important role in the infection of Toxoplasma gondii, a model organism for intracellular parasites. Recent studies demonstrated that the induction of IFN-I by the parasite depends on cell type, strain genotype, and mouse strain. IFN-I can inhibit the proliferation of T. gondii, but few studies showed that it is beneficial to the growth of the parasite. Meanwhile, T. gondii also can secrete proteins that impact the pathway of IFN-I production and downstream induced interferon-stimulated genes (ISGs) regulation, thereby escaping immune destruction by the host. This article reviews the major findings and progress in the production, function, and regulation of IFN-I during T. gondii infection, to thoroughly understand the innate immune mechanism of T. gondii infection, which provides a new target for subsequent intervention and treatment.


Assuntos
Interferon Tipo I , Toxoplasma , Toxoplasmose , Toxoplasma/imunologia , Animais , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Humanos , Toxoplasmose/imunologia , Toxoplasmose/parasitologia , Interações Hospedeiro-Parasita/imunologia , Imunidade Inata , Transdução de Sinais , Regulação da Expressão Gênica , Camundongos
7.
J Exp Med ; 221(9)2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39042188

RESUMO

The contribution of placental immune responses to congenital Zika virus (ZIKV) syndrome remains poorly understood. Here, we leveraged a mouse model of ZIKV infection to identify mechanisms of innate immune restriction exclusively in the fetal compartment of the placenta. ZIKV principally infected mononuclear trophoblasts in the junctional zone, which was limited by mitochondrial antiviral-signaling protein (MAVS) and type I interferon (IFN) signaling mechanisms. Single nuclear RNA sequencing revealed MAVS-dependent expression of IFN-stimulated genes (ISGs) in spongiotrophoblasts but not in other placental cells that use alternate pathways to induce ISGs. ZIKV infection of Ifnar1-/- or Mavs-/- placentas was associated with greater infection of the adjacent immunocompetent decidua, and heterozygous Mavs+/- or Ifnar1+/- dams carrying immunodeficient fetuses sustained greater maternal viremia and tissue infection than dams carrying wild-type fetuses. Thus, MAVS-IFN signaling in the fetus restricts ZIKV infection in junctional zone trophoblasts, which modulates dissemination and outcome for both the fetus and the pregnant mother.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Decídua , Feto , Interferon Tipo I , Placenta , Receptor de Interferon alfa e beta , Transdução de Sinais , Trofoblastos , Infecção por Zika virus , Zika virus , Feminino , Animais , Gravidez , Interferon Tipo I/metabolismo , Interferon Tipo I/imunologia , Transdução de Sinais/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Placenta/imunologia , Placenta/virologia , Placenta/metabolismo , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia , Zika virus/imunologia , Zika virus/fisiologia , Camundongos , Decídua/imunologia , Decídua/virologia , Decídua/metabolismo , Feto/imunologia , Feto/virologia , Trofoblastos/imunologia , Trofoblastos/virologia , Trofoblastos/metabolismo , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Imunidade Inata , Complicações Infecciosas na Gravidez/imunologia , Complicações Infecciosas na Gravidez/virologia , Modelos Animais de Doenças
8.
JCI Insight ; 9(13)2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38973611

RESUMO

Optimization of protective immune responses against SARS-CoV-2 remains an urgent worldwide priority. In this regard, type III IFN (IFN-λ) restricts SARS-CoV-2 infection in vitro, and treatment with IFN-λ limits infection, inflammation, and pathogenesis in murine models. Furthermore, IFN-λ has been developed for clinical use to limit COVID-19 severity. However, whether endogenous IFN-λ signaling has an effect on SARS-CoV-2 antiviral immunity and long-term immune protection in vivo is unknown. In this study, we identified a requirement for IFN-λ signaling in promoting viral clearance and protective immune programming in SARS-CoV-2 infection of mice. Expression of both IFN and IFN-stimulated gene (ISG) in the lungs were minimally affected by the absence of IFN-λ signaling and correlated with transient increases in viral titers. We found that IFN-λ supported the generation of protective CD8 T cell responses against SARS-CoV-2 by facilitating accumulation of CD103+ DC in lung draining lymph nodes (dLN). IFN-λ signaling specifically in DCs promoted the upregulation of costimulatory molecules and the proliferation of CD8 T cells. Intriguingly, antigen-specific CD8 T cell immunity to SARS-CoV-2 was independent of type I IFN signaling, revealing a nonredundant function of IFN-λ. Overall, these studies demonstrate a critical role for IFN-λ in protective innate and adaptive immunity upon infection with SARS-CoV-2 and suggest that IFN-λ serves as an immune adjuvant to support CD8 T cell immunity.


Assuntos
Linfócitos T CD8-Positivos , COVID-19 , Interferon Tipo I , SARS-CoV-2 , Animais , Linfócitos T CD8-Positivos/imunologia , SARS-CoV-2/imunologia , Camundongos , COVID-19/imunologia , COVID-19/virologia , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Pulmão/imunologia , Pulmão/virologia , Transdução de Sinais/imunologia , Modelos Animais de Doenças , Interferon lambda , Interferons/imunologia , Interferons/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Dendríticas/imunologia , Humanos
9.
Life Sci Alliance ; 7(9)2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38977311

RESUMO

The ubiquitin-like modifier FAT10 is upregulated under pro-inflammatory conditions, targets its substrates for proteasomal degradation and functions as a negative regulator of the type-I IFN response. Influenza A virus infection upregulates the production of type-I IFN and the expression of the E3 ligase TRIM21, which regulates type-I IFN production in a positive feedback manner. In this study, we show that FAT10 becomes covalently conjugated to TRIM21 and that this targets TRIM21 for proteasomal degradation. We further show that the coiled-coil and PRYSPRY domains of TRIM21 and the C-terminal diglycine motif of FAT10 are important for the TRIM21-FAT10 interaction. Moreover, upon influenza A virus infection and in the presence of FAT10 the total ubiquitination of TRIM21 is reduced and our data reveal that the FAT10-mediated degradation of TRIM21 diminishes IFNß production. Overall, this study provides strong evidence that FAT10 down-regulates the antiviral type-I IFN production by modulating additional molecules of the RIG-I signaling pathway besides the already published OTUB1. In addition, we elucidate a novel mechanism of FAT10-mediated proteasomal degradation of TRIM21 that regulates its stability.


Assuntos
Interferon Tipo I , Complexo de Endopeptidases do Proteassoma , Ribonucleoproteínas , Ubiquitinação , Ubiquitinas , Humanos , Ribonucleoproteínas/metabolismo , Ribonucleoproteínas/genética , Interferon Tipo I/metabolismo , Ubiquitinas/metabolismo , Ubiquitinas/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Regulação para Baixo , Células HEK293 , Transdução de Sinais , Vírus da Influenza A/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Proteólise , Animais
10.
Nat Commun ; 15(1): 5759, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38982116

RESUMO

Type I interferons have been well recognized for their roles in various types of immune cells during tumor immunotherapy. However, their direct effects on tumor cells are less understood. Oxidative phosphorylation is typically latent in tumor cells. Whether oxidative phosphorylation can be targeted for immunotherapy remains unclear. Here, we find that tumor cell responsiveness to type I, but not type II interferons, is essential for CD47-SIRPα blockade immunotherapy in female mice. Mechanistically, type I interferons directly reprogram tumor cell metabolism by activating oxidative phosphorylation for ATP production in an ISG15-dependent manner. ATP extracellular release is also promoted by type I interferons due to enhanced secretory autophagy. Functionally, tumor cells with genetic deficiency in oxidative phosphorylation or autophagy are resistant to CD47-SIRPα blockade. ATP released upon CD47-SIRPα blockade is required for antitumor T cell response induction via P2X7 receptor-mediated dendritic cell activation. Based on this mechanism, combinations with inhibitors of ATP-degrading ectoenzymes, CD39 and CD73, are designed and show synergistic antitumor effects with CD47-SIRPα blockade. Together, these data reveal an important role of type I interferons on tumor cell metabolic reprograming for tumor immunotherapy and provide rational strategies harnessing this mechanism for enhanced efficacy of CD47-SIRPα blockade.


Assuntos
Trifosfato de Adenosina , Antígeno CD47 , Interferon Tipo I , Fosforilação Oxidativa , Receptores Imunológicos , Transdução de Sinais , Animais , Antígeno CD47/metabolismo , Antígeno CD47/genética , Interferon Tipo I/metabolismo , Receptores Imunológicos/metabolismo , Receptores Imunológicos/genética , Feminino , Camundongos , Trifosfato de Adenosina/metabolismo , Fosforilação Oxidativa/efeitos dos fármacos , Linhagem Celular Tumoral , Camundongos Endogâmicos C57BL , Imunoterapia/métodos , Humanos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Receptores Purinérgicos P2X7/genética , Autofagia/efeitos dos fármacos , Apirase/metabolismo , Camundongos Knockout , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Citocinas/metabolismo
11.
J Immunol ; 213(2): 109-114, 2024 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-38950331

RESUMO

ATPase cation transporting 13A2 (ATP13A2) is an endolysosomal P-type ATPase known to be a polyamine transporter, explored mostly in neurons. As endolysosomal functions are also crucial in innate immune cells, we aimed to explore the potential role of ATP13A2 in the human immunocellular compartment. We found that human plasmacytoid dendritic cells (pDCs), the professional type I IFN-producing immune cells, especially have a prominent enrichment of ATP13A2 expression in endolysosomal compartments. ATP13A2 knockdown in human pDCs interferes with cytokine induction in response to TLR9/7 activation in response to bona fide ligands. ATP13A2 plays this crucial role in TLR9/7 activation in human pDCs by regulating endolysosomal pH and mitochondrial reactive oxygen generation. This (to our knowledge) hitherto unknown regulatory mechanism in pDCs involving ATP13A2 opens up a new avenue of research, given the crucial role of pDC-derived type I IFNs in protective immunity against infections as well as in the immunopathogenesis of myriad contexts of autoreactive inflammation.


Assuntos
Células Dendríticas , Endossomos , Lisossomos , Receptor Toll-Like 9 , Humanos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Lisossomos/metabolismo , Lisossomos/imunologia , Receptor Toll-Like 9/metabolismo , Receptor Toll-Like 9/imunologia , Endossomos/metabolismo , Endossomos/imunologia , ATPases Translocadoras de Prótons/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/imunologia , Células Cultivadas , Interferon Tipo I/metabolismo , Interferon Tipo I/imunologia , Receptor 7 Toll-Like
12.
Oncoimmunology ; 13(1): 2377830, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39005546

RESUMO

Attenuated measles virus (MV) exerts its oncolytic activity in malignant pleural mesothelioma (MPM) cells that lack type-I interferon (IFN-I) production or responsiveness. However, other cells in the tumor microenvironment (TME), such as myeloid cells, possess functional antiviral pathways. In this study, we aimed to characterize the interplay between MV and the myeloid cells in human MPM. We cocultured MPM cell lines with monocytes or macrophages and infected them with MV. We analyzed the transcriptome of each cell type and studied their secretion and phenotypes by high-dimensional flow cytometry. We also measured transgene expression using an MV encoding GFP (MV-GFP). We show that MPM cells drive the differentiation of monocytes into M2-like macrophages. These macrophages inhibit GFP expression in tumor cells harboring a defect in IFN-I production and a functional signaling downstream of the IFN-I receptor, while having minimal effects on GFP expression in tumor cells with defect of responsiveness to IFN-I. Interestingly, inhibition of the IFN-I signaling by ruxolitinib restores GFP expression in tumor cells. Upon MV infection, cocultured macrophages express antiviral pro-inflammatory genes and induce the expression of IFN-stimulated genes in tumor cells. MV also increases the expression of HLA and costimulatory molecules on macrophages and their phagocytic activity. Finally, MV induces the secretion of inflammatory cytokines, especially IFN-I, and PD-L1 expression in tumor cells and macrophages. These results show that macrophages reduce viral proteins expression in some MPM cell lines through their IFN-I production and generate a pro-inflammatory interplay that may stimulate the patient's anti-tumor immune response.


Assuntos
Técnicas de Cocultura , Macrófagos , Vírus do Sarampo , Terapia Viral Oncolítica , Vírus Oncolíticos , Microambiente Tumoral , Humanos , Vírus do Sarampo/genética , Vírus do Sarampo/fisiologia , Microambiente Tumoral/imunologia , Macrófagos/metabolismo , Macrófagos/imunologia , Macrófagos/virologia , Vírus Oncolíticos/genética , Terapia Viral Oncolítica/métodos , Linhagem Celular Tumoral , Mesotelioma Maligno/patologia , Mesotelioma Maligno/terapia , Interferon Tipo I/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/virologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/terapia , Neoplasias Pulmonares/virologia , Diferenciação Celular
13.
Proc Natl Acad Sci U S A ; 121(29): e2403917121, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-38980903

RESUMO

Pancreatic ductal adenocarcinoma (PDA) is a potentially lethal disease lacking effective treatments. Its immunosuppressive tumor microenvironment (TME) allows it to evade host immunosurveillance and limits response to immunotherapy. Here, using the mouse KRT19-deficient (sgKRT19-edited) PDA model, we find that intratumoral accumulation of natural killer T (NKT) cells is required to establish an immunologically active TME. Mechanistically, intratumoral NKT cells facilitate type I interferon (IFN) production to initiate an antitumor adaptive immune response, and orchestrate the intratumoral infiltration of T cells, dendritic cells, natural killer cells, and myeloid-derived suppressor cells. At the molecular level, NKT cells promote the production of type I IFN through the interaction of their CD40L with CD40 on myeloid cells. To evaluate the therapeutic potential of these observations, we find that administration of folinic acid to mice bearing PDA increases NKT cells in the TME and improves their response to anti-PD-1 antibody treatment. In conclusion, NKT cells have an essential role in the immune response to mouse PDA and are potential targets for immunotherapy.


Assuntos
Carcinoma Ductal Pancreático , Células T Matadoras Naturais , Neoplasias Pancreáticas , Microambiente Tumoral , Animais , Camundongos , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Células T Matadoras Naturais/imunologia , Microambiente Tumoral/imunologia , Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/terapia , Carcinoma Ductal Pancreático/patologia , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Imunoterapia/métodos , Camundongos Endogâmicos C57BL , Leucovorina/administração & dosagem , Leucovorina/uso terapêutico , Humanos , Células Supressoras Mieloides/imunologia
14.
J Exp Med ; 221(9)2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39017930

RESUMO

Autoantibodies neutralizing type I interferons (IFN-Is) can underlie infection severity. Here, we trace the development of these autoantibodies at high-resolution using longitudinal samples from 1,876 well-treated individuals living with HIV over a 35-year period. Similar to general populations, ∼1.9% of individuals acquired anti-IFN-I autoantibodies as they aged (median onset ∼63 years). Once detected, anti-IFN-I autoantibodies persisted lifelong, and titers increased over decades. Individuals developed distinct neutralizing and non-neutralizing autoantibody repertoires at discrete times that selectively targeted combinations of IFNα, IFNß, and IFNω. Emergence of neutralizing anti-IFNα autoantibodies correlated with reduced baseline IFN-stimulated gene levels and was associated with subsequent susceptibility to severe COVID-19 several years later. Retrospective measurements revealed enrichment of pre-existing autoreactivity against other autoantigens in individuals who later developed anti-IFN-I autoantibodies, and there was evidence for prior viral infections or increased IFN at the time of anti-IFN-I autoantibody triggering. These analyses suggest that age-related loss of self-tolerance prior to IFN-I immune-triggering poses a risk of developing lifelong functional IFN-I deficiency.


Assuntos
Anticorpos Neutralizantes , Autoanticorpos , COVID-19 , Interferon Tipo I , Humanos , Autoanticorpos/imunologia , Interferon Tipo I/imunologia , Pessoa de Meia-Idade , Masculino , Feminino , COVID-19/imunologia , Anticorpos Neutralizantes/imunologia , Adulto , Idoso , SARS-CoV-2/imunologia , Infecções por HIV/imunologia , Interferon-alfa/imunologia , Estudos Retrospectivos
15.
Sci Adv ; 10(29): eadm8660, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39028818

RESUMO

Despite the success of immunotherapy, overcoming immunoresistance in cancer remains challenging. We identified a unique niche of tumor-associated macrophages (TAMs), coexpressing T cell immunoglobulin and mucin domain-containing 3 (TIM3) and V-domain immunoglobulin suppressor of T cell activation (VISTA), that dominated human and mouse tumors resistant to most of the currently used immunotherapies. TIM3+VISTA+ TAMs were sustained by IL-4-enriching tumors with low (neo)antigenic and T cell-depleted features. TIM3+VISTA+ TAMs showed an anti-inflammatory and protumorigenic phenotype coupled with inability to sense type I interferon (IFN). This was established with cancer cells succumbing to immunogenic cell death (ICD). Dying cancer cells not only triggered autocrine type I IFNs but also exposed HMGB1/VISTA that engaged TIM3/VISTA on TAMs to suppress paracrine IFN-responses. Accordingly, TIM3/VISTA blockade synergized with paclitaxel, an ICD-inducing chemotherapy, to repolarize TIM3+VISTA+ TAMs to proinflammatory TAMs that killed cancer cells via tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) signaling. We propose targeting TIM3+VISTA+ TAMs to overcome immunoresistant tumors.


Assuntos
Receptor Celular 2 do Vírus da Hepatite A , Imunoterapia , Macrófagos Associados a Tumor , Macrófagos Associados a Tumor/metabolismo , Macrófagos Associados a Tumor/imunologia , Macrófagos Associados a Tumor/efeitos dos fármacos , Animais , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Humanos , Imunoterapia/métodos , Camundongos , Resistencia a Medicamentos Antineoplásicos , Neoplasias/terapia , Neoplasias/imunologia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Linhagem Celular Tumoral , Microambiente Tumoral/imunologia , Interferon Tipo I/metabolismo , Antígenos B7
16.
Nat Commun ; 15(1): 5842, 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38992037

RESUMO

Activating interferon responses with STING agonists (STINGa) is a current cancer immunotherapy strategy, and therapeutic modalities that enable tumor-targeted delivery via systemic administration could be beneficial. Here we demonstrate that tumor cell-directed STING agonist antibody-drug-conjugates (STINGa ADCs) activate STING in tumor cells and myeloid cells and induce anti-tumor innate immune responses in in vitro, in vivo (in female mice), and ex vivo tumor models. We show that the tumor cell-directed STINGa ADCs are internalized into myeloid cells by Fcγ-receptor-I in a tumor antigen-dependent manner. Systemic administration of STINGa ADCs in mice leads to STING activation in tumors, with increased anti-tumor activity and reduced serum cytokine elevations compared to a free STING agonist. Furthermore, STINGa ADCs induce type III interferons, which contribute to the anti-tumor activity by upregulating type I interferon and other key chemokines/cytokines. These findings reveal an important role for type III interferons in the anti-tumor activity elicited by STING agonism and provide rationale for the clinical development of tumor cell-directed STINGa ADCs.


Assuntos
Imunidade Inata , Imunoconjugados , Interferons , Proteínas de Membrana , Animais , Proteínas de Membrana/agonistas , Proteínas de Membrana/imunologia , Imunidade Inata/efeitos dos fármacos , Feminino , Humanos , Camundongos , Linhagem Celular Tumoral , Imunoconjugados/farmacologia , Imunoconjugados/administração & dosagem , Interferons/metabolismo , Interferon lambda , Neoplasias/imunologia , Neoplasias/tratamento farmacológico , Interferon Tipo I/imunologia , Citocinas/metabolismo , Células Mieloides/imunologia , Células Mieloides/efeitos dos fármacos , Imunoterapia/métodos , Camundongos Endogâmicos C57BL , Receptores de IgG/agonistas , Receptores de IgG/metabolismo , Receptores de IgG/imunologia
17.
Front Immunol ; 15: 1354055, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39007143

RESUMO

Recombinant adeno-associated virus (AAV) vectors have emerged as the preferred platform for gene therapy of rare human diseases. Despite the clinical promise, host immune responses to AAV vectors and transgene remain a major barrier to the development of successful AAV-based human gene therapies. Here, we assessed the human innate immune response to AAV9, the preferred serotype for AAV-mediated gene therapy of the CNS. We showed that AAV9 induced type I interferon (IFN) and IL-6 responses in human blood from healthy donors. This innate response was replicated with AAV6, required full viral particles, but was not observed in every donor. Depleting CpG motifs from the AAV transgene or inhibiting TLR9 signaling reduced type I IFN response to AAV9 in responding donors, highlighting the importance of TLR9-mediated DNA sensing for the innate response to AAV9. Remarkably, we further demonstrated that only seropositive donors with preexisting antibodies to AAV9 capsid mounted an innate immune response to AAV9 in human whole blood and that anti-AAV9 antibodies were necessary and sufficient to promote type I IFN release and plasmacytoid dendritic (pDC) cell activation in response to AAV9. Thus, our study reveals a previously unidentified requirement for AAV preexisting antibodies for TLR9-mediated type I IFN response to AAV9 in human blood.


Assuntos
Dependovirus , Vetores Genéticos , Imunidade Humoral , Interferon Tipo I , Receptor Toll-Like 9 , Humanos , Receptor Toll-Like 9/imunologia , Receptor Toll-Like 9/genética , Dependovirus/genética , Dependovirus/imunologia , Interferon Tipo I/imunologia , Vetores Genéticos/genética , Imunidade Inata , Células Dendríticas/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Terapia Genética , Interleucina-6/sangue , Interleucina-6/imunologia
18.
Clin Rheumatol ; 43(8): 2403-2416, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38963465

RESUMO

Systemic lupus erythematosus (SLE) is a common autoimmune disease with a polymorphic clinical presentation involving multisystem damages with significant differences in prevalence and disease severity among different ethnic groups. Although genetic, hormonal, and environmental factors have been demonstrated to contribute a lot to SLE, the pathogenesis of SLE is still unknown. Numerous evidence revealed that gene variants within the type I interferons (IFN) signaling pathway performed the great genetic associations with autoimmune diseases including SLE. To date, through genome-wide association studies (GWAS), genetic association studies showed that more than 100 susceptibility genes have been linked to the pathogenesis of SLE, among which TYK2, STAT1, STAT4, and IRF5 are important molecules directly connected to the type I interferon signaling system. The review summarized the genetic associations and the detailed risk loci of STAT4 and IRF5 with Asian SLE patients, explored the genotype distributions associated with the main clinical manifestations of SLE, and sorted out the potential reasons for the differences in susceptibility in Asia and Europe. Moreover, the therapies targeting STAT4 and IRF5 were also evaluated in order to propose more personalized and targeted treatment plans in SLE.


Assuntos
Povo Asiático , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Fatores Reguladores de Interferon , Interferon Tipo I , Lúpus Eritematoso Sistêmico , Fator de Transcrição STAT4 , Humanos , Fator de Transcrição STAT4/genética , Lúpus Eritematoso Sistêmico/genética , Fatores Reguladores de Interferon/genética , Povo Asiático/genética , Interferon Tipo I/genética , Polimorfismo de Nucleotídeo Único , Transdução de Sinais/genética
19.
PLoS Pathog ; 20(6): e1012287, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38843304

RESUMO

The kinetics of type I interferon (IFN) induction versus the virus replication compete, and the result of the competition determines the outcome of the infection. Chaperone proteins that involved in promoting the activation kinetics of PRRs rapidly trigger antiviral innate immunity. We have previously shown that prior to the interaction with MAVS to induce type I IFN, 14-3-3η facilitates the oligomerization and intracellular redistribution of activated MDA5. Here we report that the cleavage of 14-3-3η upon MDA5 activation, and we identified Caspase-3 activated by MDA5-dependent signaling was essential to produce sub-14-3-3η lacking the C-terminal helix (αI) and tail. The cleaved form of 14-3-3η (sub-14-3-3η) could strongly interact with MDA5 but could not support MDA5-dependent type I IFN induction, indicating the opposite functions between the full-length 14-3-3η and sub-14-3-3η. During human coronavirus or enterovirus infections, the accumulation of sub-14-3-3η was observed along with the activation of Caspase-3, suggesting that RNA viruses may antagonize 14-3-3η by promoting the formation of sub-14-3-3η to impair antiviral innate immunity. In conclusion, sub-14-3-3η, which could not promote MDA5 activation, may serve as a negative feedback to return to homeostasis to prevent excessive type I IFN production and unnecessary inflammation.


Assuntos
Proteínas 14-3-3 , Caspase 3 , Helicase IFIH1 Induzida por Interferon , Proteínas 14-3-3/metabolismo , Humanos , Helicase IFIH1 Induzida por Interferon/metabolismo , Helicase IFIH1 Induzida por Interferon/genética , Caspase 3/metabolismo , Imunidade Inata , Células HEK293 , Animais , Transdução de Sinais , Interferon Tipo I/metabolismo
20.
Nat Commun ; 15(1): 4772, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38858384

RESUMO

The underlying mechanisms of atherosclerosis, the second leading cause of death among Werner syndrome (WS) patients, are not fully understood. Here, we establish an in vitro co-culture system using macrophages (iMφs), vascular endothelial cells (iVECs), and vascular smooth muscle cells (iVSMCs) derived from induced pluripotent stem cells. In co-culture, WS-iMφs induces endothelial dysfunction in WS-iVECs and characteristics of the synthetic phenotype in WS-iVSMCs. Transcriptomics and open chromatin analysis reveal accelerated activation of type I interferon signaling and reduced chromatin accessibility of several transcriptional binding sites required for cellular homeostasis in WS-iMφs. Furthermore, the H3K9me3 levels show an inverse correlation with retrotransposable elements, and retrotransposable element-derived double-stranded RNA activates the DExH-box helicase 58 (DHX58)-dependent cytoplasmic RNA sensing pathway in WS-iMφs. Conversely, silencing type I interferon signaling in WS-iMφs rescues cell proliferation and suppresses cellular senescence and inflammation. These findings suggest that Mφ-specific inhibition of type I interferon signaling could be targeted to treat atherosclerosis in WS patients.


Assuntos
Aterosclerose , Inflamação , Interferon Tipo I , Macrófagos , Retroelementos , Síndrome de Werner , Interferon Tipo I/metabolismo , Síndrome de Werner/genética , Síndrome de Werner/metabolismo , Humanos , Aterosclerose/metabolismo , Aterosclerose/imunologia , Aterosclerose/genética , Aterosclerose/patologia , Macrófagos/metabolismo , Macrófagos/imunologia , Retroelementos/genética , Inflamação/metabolismo , Inflamação/patologia , Inflamação/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Transdução de Sinais , Técnicas de Cocultura , Miócitos de Músculo Liso/metabolismo , Células Endoteliais/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , RNA Helicases DEAD-box/metabolismo , RNA Helicases DEAD-box/genética , Senescência Celular , Proliferação de Células
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA