Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.219
Filtrar
1.
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
2.
Mol Med Rep ; 30(3)2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38963029

RESUMO

Viral infections in the respiratory tract are common, and, in recent years, severe acute respiratory syndrome coronavirus 2 outbreaks have highlighted the effect of viral infections on antiviral innate immune and inflammatory reactions. Specific treatments for numerous viral respiratory infections have not yet been established and they are mainly treated symptomatically. Therefore, understanding the details of the innate immune system underlying the airway epithelium is crucial for the development of new therapies. The present study aimed to investigate the function and expression of interferon (IFN)­stimulated gene (ISG)60 in non­cancerous bronchial epithelial BEAS­2B cells exposed to a Toll­like receptor 3 agonist. BEAS­2B cells were treated with a synthetic TLR3 ligand, polyinosinic­polycytidylic acid (poly IC). The mRNA and protein expression levels of ISG60 were analyzed using reverse transcription­quantitative PCR and western blotting, respectively. The levels of C­X­C motif chemokine ligand 10 (CXCL10) were examined using an enzyme­linked immunosorbent assay, and the effects of knockdown of IFN­ß, ISG60 and ISG56 were examined using specific small interfering RNAs. Notably, ISG60 expression was increased in proportion to poly IC concentration, and recombinant human IFN­ß also induced ISG60 expression. By contrast, knockdown of IFN­ß and ISG56 decreased ISG60 expression, and ISG60 knockdown reduced CXCL10 and ISG56 expression. These findings suggested that ISG60 is partly implicated in CXCL10 expression and that ISG60 may serve a role in the innate immune response of bronchial epithelial cells. The present study highlights ISG60 as a potential target for new therapeutic strategies against viral infections in the airway.


Assuntos
Brônquios , Quimiocina CXCL10 , Células Epiteliais , Poli I-C , Transdução de Sinais , Receptor 3 Toll-Like , Humanos , Receptor 3 Toll-Like/metabolismo , Receptor 3 Toll-Like/genética , Quimiocina CXCL10/metabolismo , Quimiocina CXCL10/genética , Células Epiteliais/metabolismo , Células Epiteliais/efeitos dos fármacos , Brônquios/citologia , Brônquios/metabolismo , Poli I-C/farmacologia , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular , Imunidade Inata , Interferon beta/metabolismo , Interferon beta/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Ligação a RNA , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Reguladoras de Apoptose
3.
Nat Commun ; 15(1): 5961, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39013878

RESUMO

Autoreactive CD4+ T helper cells are critical players that orchestrate the immune response both in multiple sclerosis (MS) and in other neuroinflammatory autoimmune diseases. Ubiquitination is a posttranslational protein modification involved in regulating a variety of cellular processes, including CD4+ T cell differentiation and function. However, only a limited number of E3 ubiquitin ligases have been characterized in terms of their biological functions, particularly in CD4+ T cell differentiation and function. In this study, we found that the RING finger protein 213 (RNF213) specifically promoted regulatory T (Treg) cell differentiation in CD4+ T cells and attenuated autoimmune disease development in an FOXO1-dependent manner. Mechanistically, RNF213 interacts with Forkhead Box Protein O1 (FOXO1) and promotes nuclear translocation of FOXO1 by K63-linked ubiquitination. Notably, RNF213 expression in CD4+ T cells was induced by IFN-ß and exerts a crucial role in the therapeutic efficacy of IFN-ß for MS. Together, our study findings collectively emphasize the pivotal role of RNF213 in modulating adaptive immune responses. RNF213 holds potential as a promising therapeutic target for addressing disorders associated with Treg cells.


Assuntos
Diferenciação Celular , Proteína Forkhead Box O1 , Interferon beta , Linfócitos T Reguladores , Ubiquitina-Proteína Ligases , Ubiquitinação , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Animais , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Camundongos , Humanos , Interferon beta/metabolismo , Camundongos Endogâmicos C57BL , Núcleo Celular/metabolismo , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , Esclerose Múltipla/genética , Esclerose Múltipla/patologia , Transporte Ativo do Núcleo Celular , Feminino , Camundongos Knockout , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/genética , Células HEK293
4.
Int J Mol Sci ; 25(13)2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-39000519

RESUMO

The aim of the present study was to investigate the impact of CCR5 Δ32 and CTLA-4 polymorphisms on the response to IFN-ß treatment in our cohort of MS patients from Croatia and Slovenia. Genomic DNA was obtained from 295 MS patients (230 female; 65 male) classified as responders (n = 173) and non-responders (n = 122) based on clinical criteria for treatment efficacy. Genotyping was performed via PCR/PCR-RFLP. No significant differences in the genotype/allele frequencies of CCR5Δ32 and CTLA-4 +49 A/G were detected between male responders and non-responders. A significantly higher prevalence (p = 0.039) of the CTLA-4 +49 AA genotype was found in female responders (42.1%) compared to non-responders (28.9%). Using multiple forward regression analysis, the CTLA-4 +49 AA genotype significantly predicted a positive response to IFN-ß therapy in females (p = 0.011) and contributed to 4.5% of response variability. Furthermore, the combined presence of the CCR5Δ32 wtwt/CTLA-4 +49 AA genotype significantly predicted a positive response to treatment in females (p = 0.025). The age at disease onset, pretreatment relapse rate, and baseline EDSS score were not reliable predictors of treatment response in MS patients. Our results indicate that the presence of the CCR5Δ32 polymorphism was not associated with the response to IFN-ß treatment, whereas the CTLA-4 +49 polymorphism showed a positive correlation with an optimal response in female patients.


Assuntos
Antígeno CTLA-4 , Frequência do Gene , Interferon beta , Esclerose Múltipla , Polimorfismo de Nucleotídeo Único , Receptores CCR5 , Humanos , Feminino , Masculino , Antígeno CTLA-4/genética , Receptores CCR5/genética , Interferon beta/uso terapêutico , Eslovênia , Adulto , Croácia , Esclerose Múltipla/genética , Esclerose Múltipla/tratamento farmacológico , Pessoa de Meia-Idade , Genótipo , Resultado do Tratamento
5.
Sci Adv ; 10(29): eadl5638, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39018414

RESUMO

Viruses can selectively repress the translation of mRNAs involved in the antiviral response. RNA viruses exploit the Grb10-interacting GYF (glycine-tyrosine-phenylalanine) proteins 2 (GIGYF2) and eukaryotic translation initiation factor 4E (eIF4E) homologous protein 4EHP to selectively repress the translation of transcripts such as Ifnb1, which encodes the antiviral cytokine interferon-ß (IFN-ß). Herein, we reveal that GIGYF1, a paralog of GIGYF2, robustly represses cellular mRNA translation through a distinct 4EHP-independent mechanism. Upon recruitment to a target mRNA, GIGYF1 binds to subunits of eukaryotic translation initiation factor 3 (eIF3) at the eIF3-eIF4G1 interaction interface. This interaction disrupts the eIF3 binding to eIF4G1, resulting in transcript-specific translational repression. Depletion of GIGYF1 induces a robust immune response by derepressing IFN-ß production. Our study highlights a unique mechanism of translational regulation by GIGYF1 that involves sequestering eIF3 and abrogating its binding to eIF4G1. This mechanism has profound implications for the host response to viral infections.


Assuntos
Fator de Iniciação 3 em Eucariotos , Fator de Iniciação Eucariótico 4G , Ligação Proteica , RNA Mensageiro , Fator de Iniciação Eucariótico 4G/metabolismo , Fator de Iniciação Eucariótico 4G/genética , Fator de Iniciação 3 em Eucariotos/metabolismo , Fator de Iniciação 3 em Eucariotos/genética , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Interferon beta/metabolismo , Interferon beta/genética , Proteínas de Transporte/metabolismo , Proteínas de Transporte/genética , Iniciação Traducional da Cadeia Peptídica , Animais , Biossíntese de Proteínas , Regulação da Expressão Gênica
6.
Viruses ; 16(6)2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38932169

RESUMO

Repression of human cytomegalovirus (HCMV) immediate-early (IE) gene expression is a key regulatory step in the establishment and maintenance of latent reservoirs. Viral IE transcription and protein accumulation can be elevated during latency by treatment with histone deacetylase inhibitors such as valproic acid (VPA), rendering infected cells visible to adaptive immune responses. However, the latency-associated viral protein UL138 inhibits the ability of VPA to enhance IE gene expression during infection of incompletely differentiated myeloid cells that support latency. UL138 also limits the accumulation of IFNß transcripts by inhibiting the cGAS-STING-TBK1 DNA-sensing pathway. Here, we show that, in the absence of UL138, the cGAS-STING-TBK1 pathway promotes both IFNß accumulation and VPA-responsive IE gene expression in incompletely differentiated myeloid cells. Inactivation of this pathway by either genetic or pharmacological inhibition phenocopied UL138 expression and reduced VPA-responsive IE transcript and protein accumulation. This work reveals a link between cytoplasmic pathogen sensing and epigenetic control of viral lytic phase transcription and suggests that manipulation of pattern recognition receptor signaling pathways could aid in the refinement of MIEP regulatory strategies to target latent viral reservoirs.


Assuntos
Citomegalovirus , Proteínas de Membrana , Células Mieloides , Nucleotidiltransferases , Proteínas Serina-Treonina Quinases , Transdução de Sinais , Ácido Valproico , Humanos , Ácido Valproico/farmacologia , Células Mieloides/virologia , Células Mieloides/metabolismo , Células Mieloides/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Citomegalovirus/fisiologia , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/genética , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Infecções por Citomegalovirus/virologia , Infecções por Citomegalovirus/metabolismo , Infecções por Citomegalovirus/genética , Latência Viral/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Genes Precoces , Interferon beta/metabolismo , Interferon beta/genética
7.
Biol Pharm Bull ; 47(6): 1196-1203, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38897970

RESUMO

Microglia, resident immune cells in the central nervous system (CNS), play a critical role in maintaining CNS homeostasis. However, microglia activated in response to brain injury produce various inflammatory mediators, including nitric oxide (NO) and proinflammatory cytokines, leading to considerable neuronal damage. NO generated by inducible NO synthase (iNOS) rapidly reacts with superoxide to form a highly toxic product, peroxynitrite. Therefore, iNOS is considered to be a putative therapeutic target for cerebral ischemia. Here, we examined the effects of panobinostat (Pano), a histone deacetylase inhibitor, on lipopolysaccharide (LPS)-induced iNOS expression using rat immortalized microglia HAPI cells. Pano inhibited LPS-induced expression of iNOS mRNA and NO production in a dose-dependent manner; however, it had little effect on the LPS-induced activation of c-Jun N-terminal kinase (JNK) and p38 or nuclear translocation of nuclear factor-κB (NF-κB). The interferon-ß (IFN-ß)/signal transducer and activator of transcription (STAT) pathway is essential for LPS-induced iNOS expression in macrophages/microglia. We also examined the effects of Pano on LPS-induced IFN-ß signaling. Pano markedly inhibited LPS-induced IFN-ß expression and subsequent tyrosine phosphorylation of STAT1. However, the addition of IFN-ß restored the decreased STAT1 phosphorylation but not the decreased iNOS expression. In addition, Pano inhibited the LPS-increased expression of octamer binding protein-2 and interferon regulatory factor 9 responsible for iNOS expression, but IFN-ß addition also failed to restore the decreased expression of these factors. Thus, we conclude that the inhibitory effects of Pano are due not only to the inhibition of the IFN-ß/STAT axis but also to the downregulation of other factors not involved in this axis.


Assuntos
Inibidores de Histona Desacetilases , Lipopolissacarídeos , Microglia , Óxido Nítrico Sintase Tipo II , Óxido Nítrico , Panobinostat , Animais , Microglia/efeitos dos fármacos , Microglia/metabolismo , Lipopolissacarídeos/farmacologia , Óxido Nítrico Sintase Tipo II/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Ratos , Panobinostat/farmacologia , Óxido Nítrico/metabolismo , NF-kappa B/metabolismo , Linhagem Celular , Interferon beta/metabolismo , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos dos fármacos
8.
Nat Commun ; 15(1): 5442, 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38937436

RESUMO

Although patients benefit from immune checkpoint inhibition (ICI) therapy in a broad variety of tumors, resistance may arise from immune suppressive tumor microenvironments (TME), which is particularly true of hepatocellular carcinoma (HCC). Since oncolytic viruses (OV) can generate a highly immune-infiltrated, inflammatory TME, OVs could potentially restore ICI responsiveness via recruitment, priming, and activation of anti-tumor T cells. Here we find that on the contrary, an oncolytic vesicular stomatitis virus, expressing interferon-ß (VSV-IFNß), antagonizes the effect of anti-PD-L1 therapy in a partially anti-PD-L1-responsive model of HCC. Cytometry by Time of Flight shows that VSV-IFNß expands dominant anti-viral effector CD8 T cells with concomitant relative disappearance of anti-tumor T cell populations, which are the target of anti-PD-L1. However, by expressing a range of HCC tumor antigens within VSV, combination OV and anti-PD-L1 therapeutic benefit could be restored. Our data provide a cautionary message for the use of highly immunogenic viruses as tumor-specific immune-therapeutics by showing that dominant anti-viral T cell responses can inhibit sub-dominant anti-tumor T cell responses. However, through encoding tumor antigens within the virus, oncolytic virotherapy can generate anti-tumor T cell populations upon which immune checkpoint blockade can effectively work.


Assuntos
Antígenos de Neoplasias , Antígeno B7-H1 , Linfócitos T CD8-Positivos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Terapia Viral Oncolítica , Vírus Oncolíticos , Microambiente Tumoral , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Animais , Terapia Viral Oncolítica/métodos , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/imunologia , Microambiente Tumoral/imunologia , Camundongos , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Humanos , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/imunologia , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Interferon beta/metabolismo , Interferon beta/imunologia , Camundongos Endogâmicos C57BL , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/farmacologia , Linfócitos T/imunologia , Feminino , Vesiculovirus/imunologia , Vesiculovirus/genética
9.
Vet Microbiol ; 295: 110148, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38851152

RESUMO

Water buffalo Hunnivirus (BufHuV) belongs to the family Picornaviridae and is a newly discovered member of the Hunnivirus A genus. It causes intestinal diseases in cattle, mainly lead to subclinical infections, thereby seriously threatening the health of cattle herds. In addition, it can also bring about various clinical disease syndromes which results in severe economic losses to the cattle industry. To date, there have been no reports worldwide on the study of Hunnivirus virus infecting host cells and causing innate immune responses. In this study, we found that interferon treatment effectively blocked BufHuV replication and infection with the virus weakened the host antiviral responses. Inhibiting the transcription of IFN-ß and ISGs induced by either Sendai virus (SeV) or poly(I:C) in MDBK and HCT-8 cells, were dependent on the IRF3 or NF-κB signaling pathways, and this inhibited the activation of IFN-ß promoter by TBK1 and its upstream molecules, RIGI and MDA5. By constructing and screening five BufHuV proteins, we found that VP2, 2 C, 3 C and 3D inhibited the activation of IFN-ß promoter induced by SeV. Subsequently, we showed that VP2 inhibited the activation of IRF3 induced by SeV or poly (I:C), and it inhibited IRF3 activation by inhibiting its phosphorylation and nuclear translocation. In addition, we confirmed that VP2 inhibited the activation of IFNß induced by signaling molecules, MDA5 and TBKI. In summary, these findings provide new insights into the pathogenesis of Hunnivirus and its mechanisms involved in evading host immune responses.


Assuntos
Fator Regulador 3 de Interferon , Interferon beta , Interferon beta/genética , Interferon beta/imunologia , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética , Animais , Humanos , Linhagem Celular , Transdução de Sinais/efeitos dos fármacos , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/metabolismo , Replicação Viral/efeitos dos fármacos , Imunidade Inata , Bovinos , Búfalos/virologia , NF-kappa B/metabolismo
10.
J Immunol ; 213(3): 347-361, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38847616

RESUMO

The cyclic GMP-AMP synthase (cGAS)-stimulator of IFN genes (STING) pathway is instrumental to antitumor immunity, yet the underlying molecular and cellular mechanisms are complex and still unfolding. A new paradigm suggests that cancer cells' cGAS-synthesized cGAMP can be transferred to tumor-infiltrating immune cells, eliciting STING-dependent IFN-ß response for antitumor immunity. Nevertheless, how the tumor microenvironment may shape this process remains unclear. In this study, we found that extracellular ATP, an immune regulatory molecule widely present in the tumor microenvironment, can potentiate cGAMP transfer, thereby boosting the STING signaling and IFN-ß response in murine macrophages and fibroblasts. Notably, genetic ablation or chemical inhibition of murine volume-regulation anion channel LRRC8/volume-regulated anion channel (VRAC), a recently identified cGAMP transporter, abolished ATP-potentiated cGAMP transfer and STING-dependent IFN-ß response, revealing a crucial role of LRRC8/VRAC in the cross-talk of extracellular ATP and cGAMP. Mechanistically, ATP activation of the P2X family receptors triggered Ca2+ influx and K+ efflux, promoting reactive oxygen species production. Moreover, ATP-evoked K+ efflux alleviated the phosphorylation of VRAC's obligate subunit LRRC8A/SWELL1 on S174. Mutagenesis studies indicated that the phosphorylation of S174 on LRRC8A could act as a checkpoint for VRAC in the steady state and a rheostat of ATP responsiveness. In an MC38-transplanted tumor model, systemically blocking CD39 and ENPP1, hydroxylases of extracellular ATP and cGAMP, respectively, elevated antitumor NK, NKT, and CD8+ T cell responses and restrained tumor growth in mice. Altogether, this study establishes a crucial role of ATP in facilitating LRRC8/VRAC transport cGAMP in the tumor microenvironment and provides new insight into harnessing cGAMP transfer for antitumor immunity.


Assuntos
Trifosfato de Adenosina , Proteínas de Membrana , Nucleotídeos Cíclicos , Microambiente Tumoral , Animais , Nucleotídeos Cíclicos/metabolismo , Camundongos , Trifosfato de Adenosina/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana/imunologia , Microambiente Tumoral/imunologia , Interferon beta/metabolismo , Interferon beta/imunologia , Camundongos Endogâmicos C57BL , Humanos , Transdução de Sinais/imunologia , Camundongos Knockout , Linhagem Celular Tumoral , Cátions/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Nucleotidiltransferases/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo
11.
Nat Commun ; 15(1): 5423, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38926338

RESUMO

Oncogene-induced senescence (OIS) arrests cell proliferation in response to replication stress (RS) induced by oncogenes. OIS depends on the DNA damage response (DDR), but also on the cGAS-STING pathway, which detects cytosolic DNA and induces type I interferons (IFNs). Whether and how RS and IFN responses cooperate to promote OIS remains unknown. Here, we show that the induction of OIS by the H-RASV12 oncogene in immortalized human fibroblasts depends on the MRE11 nuclease. Indeed, treatment with the MRE11 inhibitor Mirin prevented RS, micronuclei formation and IFN response induced by RASV12. Overexpression of the cytosolic nuclease TREX1 also prevented OIS. Conversely, overexpression of a dominant negative mutant of TREX1 or treatment with IFN-ß was sufficient to induce RS and DNA damage, independent of RASV12 induction. These data suggest that the IFN response acts as a positive feedback loop to amplify DDR in OIS through a process regulated by MRE11 and TREX1.


Assuntos
Senescência Celular , Dano ao DNA , Replicação do DNA , Exodesoxirribonucleases , Proteína Homóloga a MRE11 , Fosfoproteínas , Transdução de Sinais , Humanos , Exodesoxirribonucleases/metabolismo , Exodesoxirribonucleases/genética , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Proteína Homóloga a MRE11/metabolismo , Proteína Homóloga a MRE11/genética , Senescência Celular/genética , Fibroblastos/metabolismo , Interferon beta/metabolismo , Interferon beta/genética
12.
Exp Mol Pathol ; 137: 104897, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38691979

RESUMO

BACKGROUND: Signaling by toll-like receptors (TLRs) initiates important immune responses against viral infection. The role of TLRs in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is not well elucidated. Thus, we investigated the interaction of TLRs agonists and SARS-COV-2 antigens with immune cells in vitro. MATERIAL & METHODS: 30 coronavirus disease 2019 (COVID-19) patients (15 severe and 15 moderate) and 10 age and sex-matched healthy control (HC) were enrolled. Peripheral blood mononuclear cells (PBMCs) were isolated and activated with TLR3, 7, 8, and 9 agonists, the spike protein (SP) of SARS-CoV-2, and the receptor binding domain (RBD) of SP. Frequencies of CD3+IFN-ß+ T cells, and CD3+IFN-γ+ T cells were evaluated by flow cytometry. Interferon (IFN)-ß gene expression was assessed by qRT-PCR. RESULTS: The frequency of CD3+IFN-ß+ T cells was higher in PBMCs from moderate (p < 0.0001) and severe (p = 0.009) patients at baseline in comparison with HCs. The highest increase in the frequency of CD3+IFN-ß+ T cells in cell from moderate patients was induced by TLR8 agonist and SP (p < 0.0001 for both) when compared to HC, while, the highest increase of the frequency of CD3+IFN-ß+ T cells in sample of severe patients was seen with TLR8 and TLR7 agonists (both p = 0.002). The frequency of CD3+IFN-γ+ T cells was significantly increased upon stimulation with TLR agonists in cell from patients with moderate and severe COVID-19, compared with HC (all p < 0.01), except with TLR7 and TLR8 agonists. The TLR8 agonist did not significantly increase the frequency of CD3+IFN-γ+ T cells in PBMCs of severe patients, but did so in cells from patients with moderate disease (p = 0.01). Moreover, IFN-ß gene expression was significantly upregulated in CD3+T cells from moderate (p < 0.0001) and severe (p = 0.002) COVID-19 patients, compared to HC after stimulation with the TLR8 agonist, while, stimulation of T cells with SP, significantly up-regulated IFN-ß mRNA expression in cells from patients with moderate (p = 0.0003), but not severe disease. CONCLUSION: Stimulation of PBMCs from COVID-19 patients, especially patients with moderate disease, with TLR8 agonist and SP increased the frequency of IFN-ß-producing T cells and IFN-ß gene expression.


Assuntos
Complexo CD3 , COVID-19 , SARS-CoV-2 , Linfócitos T , Receptores Toll-Like , Humanos , COVID-19/imunologia , COVID-19/virologia , SARS-CoV-2/imunologia , Masculino , Feminino , Pessoa de Meia-Idade , Receptores Toll-Like/agonistas , Receptores Toll-Like/genética , Complexo CD3/imunologia , Complexo CD3/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/efeitos dos fármacos , Adulto , Interferon gama/metabolismo , Interferon gama/genética , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/genética , Interferon beta/genética , Interferon beta/imunologia , Idoso , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Agonistas do Receptor Semelhante a Toll
13.
J Immunother Cancer ; 12(5)2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38749537

RESUMO

BACKGROUND: Cancer-intrinsic type I interferon (IFN-I) production triggered by radiotherapy (RT) is mainly dependent on cytosolic double-stranded DNA (dsDNA)-mediated cGAS/STING signaling and increases cancer immunogenicity and enhances the antitumor immune response to increase therapeutic efficacy. However, cGAS/STING deficiency in colorectal cancer (CRC) may suppress the RT-induced antitumor immunity. Therefore, we aimed to evaluate the importance of the dsRNA-mediated antitumor immune response induced by RT in patients with CRC. METHODS: Cytosolic dsRNA level and its sensors were evaluated via cell-based assays (co-culture assay, confocal microscopy, pharmacological inhibition and immunofluorescent staining) and in vivo experiments. Biopsies and surgical tissues from patients with CRC who received preoperative chemoradiotherapy (neoCRT) were collected for multiplex cytokine assays, immunohistochemical analysis and SNP genotyping. We also generated a cancer-specific adenovirus-associated virus (AAV)-IFNß1 construct to evaluate its therapeutic efficacy in combination with RT, and the immune profiles were analyzed by flow cytometry and RNA-seq. RESULTS: Our studies revealed that RT stimulates the autonomous release of dsRNA from cancer cells to activate TLR3-mediated IFN-I signatures to facilitate antitumor immune responses. Patients harboring a dysfunctional TLR3 variant had reduced serum levels of IFN-I-related cytokines and intratumoral CD8+ immune cells and shorter disease-free survival following neoCRT treatment. The engineered cancer-targeted construct AAV-IFNß1 significantly improved the response to RT, leading to systematic eradication of distant tumors and prolonged survival in defective TLR3 preclinical models. CONCLUSION: Our results support that increasing cancer-intrinsic IFNß1 expression is an immunotherapeutic strategy that enhances the RT-induced antitumor immune response in locally patients with advanced CRC with dysfunctional TLR3.


Assuntos
Neoplasias Colorretais , Interferon Tipo I , Interferon beta , RNA de Cadeia Dupla , Humanos , Neoplasias Colorretais/radioterapia , Neoplasias Colorretais/imunologia , Interferon beta/metabolismo , Camundongos , Animais , Interferon Tipo I/metabolismo , Transdução de Sinais , Feminino , Masculino
14.
Artigo em Inglês | MEDLINE | ID: mdl-38821668

RESUMO

In this paper, we studied the potential genotoxic effects of human plasma from healthy volunteers, as well as patients with gastro-oesophageal reflux disease, Barrett's oesophagus (BO) and oesophageal adenocarcinoma (OAC) using the oesophageal adenocarcinoma cell line (OE33) and the lymphoblastoid cell line (TK6). Both TK6 and OE33 cells were treated with plasma (10 % volume, replacing foetal bovine serum (FBS) or horse serum (HS)) at different time points of 4 h (for the micronucleus (Mn) assay and the invasion assay) and 24 h (for the cell cycle studies). Plasma-induced effects on DNA damage levels, cell viability and the cell cycle were studied by the micronucleus assay, cytokinesis block proliferation index (CBPI) and flow cytometry respectively. The expression of IL-8 in supernatants of TK6 cells and IFN-ß in OE33 cells was also analysed by enzyme-linked immunosorbent assay (ELISA). Finally, we carried out an assessment of cellular invasion of OE33 cells following plasma treatment. The results of the micronucleus assay confirmed the genotoxicity of direct plasma treatment from some participants through the increase in DNA damage in TK6 cells. Conversely, some individual patient plasma samples reduced background levels of TK6 cell Mn frequency, in an anti-genotoxic fashion. In TK6 cells, (on average) plasma samples from patients with Barrett's oesophagus induced higher micronucleus levels than healthy volunteers (p= 0.0019). There was little difference in Mn induction when using plasma versus serum to treat the cells in vitro. Cell cycle results showed that direct plasma treatment had a marked impact on OE33 cells at 24 h (p=0.0182 for BO and p=0.0320 for OAC) by decreasing the proportion of cells in the S phase, while plasma exposure was less impactful on the cell cycle of TK6 cells. Invasion of OE33 cells was also seen to be non-significantly affected by plasma treatment of OE33 cells. The addition of N-acetyl cysteine NAC in a dose-dependent matter did not alter the formation of Mn in TK6 cells, suggesting that reactive oxygen species (ROS) are not the root cause of plasma's genotoxicity. The concentration of IL-8 in TK6 cells and IFN-ß in OE33 cells was significantly higher in cells treated with OAC-derived plasma than in the untreated negative control. Collectively, our results demonstrate that plasma-specific effects are detectable which helps us better understand some important aspects of the biology of blood-based biomarkers under development.


Assuntos
Adenocarcinoma , Esôfago de Barrett , Dano ao DNA , Neoplasias Esofágicas , Testes para Micronúcleos , Humanos , Esôfago de Barrett/patologia , Esôfago de Barrett/genética , Adenocarcinoma/patologia , Adenocarcinoma/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Plasma/metabolismo , Interleucina-8/metabolismo , Interleucina-8/genética , Linhagem Celular Tumoral , Ciclo Celular/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Adulto , Sobrevivência Celular/efeitos dos fármacos , Feminino , Micronúcleos com Defeito Cromossômico , Interferon beta , Idoso
15.
Iran J Immunol ; 21(2): 158-165, 2024 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-38761094

RESUMO

Background: The mechanisms of the function of interferon beta (IFN-ß) and natalizumab (NTZ) in multiple sclerosis (MS) patients have not yet been fully understood. Over the past decades, many studies have been conducted to evaluate gene expression changes especially regulatory non-coding RNAs such as microRNAs (miRNAs) following therapy in MS patients. Objective: To assess the changes in the expression of miR-20b in MS patients treated with IFN-ß or NTZ. Methods: Sixty patients with relapsing-remitting MS (RRMS) and 30 healthy controls (HCs) were enrolled. The patients were categorized as untreated (N=20), IFN-ß-treated (N=20), and NTZtreated (N=20). For the expression analysis, real-time PCR was performed on the whole blood. The bioinformatic tools were applied for signaling pathways enrichment analysis of miR-20b targetome. Results: The relative expression of miR-20b was significantly downregulated in the untreated patients compared with the HCs (-1.726-fold, p<0.001), while IFN-ß-treated and NTZ-treated patients showed no statistical difference compared with the HCs (0.733-fold, p=0.99 for IFN-ß and 1.025-fold, p=0.18 for NTZ). This indicates the restoration of miR-20b expression to normal level in the treated patients. Additionally, in silico analysis demonstrated that the Jak-STAT signaling pathway is enriched with miR-20b targets (p<0.0001). Conclusion: Our findings suggest that the positive effects of IFN-ß and NTZ in the RRMS patients could be potentially mediated by returning miR-20b expression to baseline.


Assuntos
Interferon beta , Janus Quinases , MicroRNAs , Esclerose Múltipla Recidivante-Remitente , Natalizumab , Fatores de Transcrição STAT , Transdução de Sinais , Humanos , MicroRNAs/genética , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Esclerose Múltipla Recidivante-Remitente/genética , Interferon beta/uso terapêutico , Natalizumab/uso terapêutico , Feminino , Estudos de Casos e Controles , Masculino , Adulto , Janus Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Adulto Jovem , Pessoa de Meia-Idade , Biologia Computacional/métodos
16.
Clin Exp Med ; 24(1): 102, 2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38758367

RESUMO

Immunotherapy is widely used in cancer treatment; however, only a subset of patients responds well to it. Significant efforts have been made to identify patients who will benefit from immunotherapy. Successful anti-tumor immunity depends on an intact cancer-immunity cycle, especially long-lasting CD8+ T-cell responses. Interferon (IFN)-α/ß/IFN-γ/interleukin (IL)-15 pathways have been reported to be involved in the development of CD8+ T cells. And these pathways may predict responses to immunotherapy. Herein, we aimed to analyze multiple public databases to investigate whether IFN-α/ß/IFN-γ/IL-15 pathways could be used to predict the response to immunotherapy. Results showed that IFN-α/ß/IFN-γ/IL-15 pathways could efficiently predict immunotherapy response, and guanylate-binding protein 1 (GBP1) could represent the IFN-α/ß/IFN-γ/IL-15 pathways. In public and private cohorts, we further demonstrated that GBP1 could efficiently predict the response to immunotherapy. Functionally, GBP1 was mainly expressed in macrophages and strongly correlated with chemokines involved in T-cell migration. Therefore, our study comprehensively investigated the potential role of GBP1 in immunotherapy, which could serve as a novel biomarker for immunotherapy and a target for drug development.


Assuntos
Proteínas de Ligação ao GTP , Imunoterapia , Neoplasias , Humanos , Linfócitos T CD8-Positivos/imunologia , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/imunologia , Imunoterapia/métodos , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Interferon gama/metabolismo , Interleucina-15/genética , Neoplasias/imunologia , Neoplasias/terapia , Transdução de Sinais
17.
Cancer Lett ; 596: 216988, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38797234

RESUMO

Type I interferons exhibit anti-proliferative and anti-cancer activities, but their detailed regulatory mechanisms in cancer have not been fully elucidated yet. RNA binding proteins are master orchestrators of gene regulation, which are closely related to tumor progression. Here we show that the upregulated RNA binding protein RBM45 correlates with poor prognosis in breast cancer. Depletion of RBM45 suppresses breast cancer progression both in cultured cells and xenograft mouse models. Mechanistically, RBM45 ablation inhibits breast cancer progression through regulating type I interferon signaling, particularly by elevating IFN-ß production. Importantly, RBM45 recruits TRIM28 to IRF7 and stimulates its SUMOylation, thereby repressing IFNB1 transcription. Loss of RBM45 reduced the SUMOylation of IRF7 by reducing the interaction between TRIM28 and IRF7 to promote IFNB1 transcription, leading to the inhibition of breast cancer progression. Taken together, our finding uncovers a vital role of RBM45 in modulating type I interferon signaling and cancer aggressive progression, implicating RBM45 as a potential therapeutic target in breast cancer.


Assuntos
Neoplasias da Mama , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Fator Regulador 7 de Interferon , Proteínas de Ligação a RNA , Sumoilação , Humanos , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Feminino , Animais , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/metabolismo , Camundongos , Transcrição Gênica , Linhagem Celular Tumoral , Interferon beta/metabolismo , Interferon beta/genética , Transdução de Sinais , Camundongos Nus , Proliferação de Células , Proteína 28 com Motivo Tripartido/metabolismo , Proteína 28 com Motivo Tripartido/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Camundongos Endogâmicos BALB C
18.
FASEB J ; 38(10): e23651, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38752537

RESUMO

Singleton-Merten syndrome (SMS) is a rare immunogenetic disorder affecting multiple systems, characterized by dental dysplasia, aortic calcification, glaucoma, skeletal abnormalities, and psoriasis. Glaucoma, a key feature of both classical and atypical SMS, remains poorly understood in terms of its molecular mechanism caused by DDX58 mutation. This study presented a novel DDX58 variant (c.1649A>C [p.Asp550Ala]) in a family with childhood glaucoma. Functional analysis showed that DDX58 variant caused an increase in IFN-stimulated gene expression and high IFN-ß-based type-I IFN. As the trabecular meshwork (TM) is responsible for controlling intraocular pressure (IOP), we examine the effect of IFN-ß on TM cells. Our study is the first to demonstrate that IFN-ß significantly reduced TM cell viability and function by activating autophagy. In addition, anterior chamber injection of IFN-ß remarkably increased IOP level in mice, which can be attenuated by treatments with autophagy inhibitor chloroquine. To uncover the specific mechanism underlying IFN-ß-induced autophagy in TM cells, we performed microarray analysis in IFN-ß-treated and DDX58 p.Asp550Ala TM cells. It showed that RSAD2 is necessary for IFN-ß-induced autophagy. Knockdown of RSAD2 by siRNA significantly decreased autophagy flux induced by IFN-ß. Our findings suggest that DDX58 mutation leads to the overproduction of IFN-ß, which elevates IOP by modulating autophagy through RSAD2 in TM cells.


Assuntos
Autofagia , Proteína DEAD-box 58 , Glaucoma , Pressão Intraocular , Malha Trabecular , Animais , Feminino , Humanos , Masculino , Camundongos , Doenças da Aorta , Autofagia/efeitos dos fármacos , Proteína DEAD-box 58/metabolismo , Proteína DEAD-box 58/genética , Hipoplasia do Esmalte Dentário , Glaucoma/patologia , Glaucoma/metabolismo , Glaucoma/genética , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/patologia , Perda Auditiva Neurossensorial/metabolismo , Interferon beta/metabolismo , Pressão Intraocular/genética , Metacarpo/anormalidades , Camundongos Endogâmicos C57BL , Doenças Musculares , Mutação , Odontodisplasia , Atrofia Óptica/genética , Atrofia Óptica/metabolismo , Atrofia Óptica/patologia , Osteoporose , Linhagem , Receptores Imunológicos , Malha Trabecular/metabolismo , Malha Trabecular/efeitos dos fármacos , Calcificação Vascular
19.
Virol Sin ; 39(3): 501-512, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38789039

RESUMO

The infection caused by porcine epidemic diarrhea virus (PEDV) is associated with high mortality in piglets worldwide. Host factors involved in the efficient replication of PEDV, however, remain largely unknown. Our recent proteomic study in the virus-host interaction network revealed a significant increase in the accumulation of CALML5 (EF-hand protein calmodulin-like 5) following PEDV infection. A further study unveiled a biphasic increase of CALML5 in 2 and 12 â€‹h after viral infection. Similar trends were observed in the intestines of piglets in the early and late stages of the PEDV challenge. Moreover, CALML5 depletion reduced PEDV mRNA and protein levels, leading to a one-order-of-magnitude decrease in virus titer. At the early stage of PEDV infection, CALML5 affected the endosomal trafficking pathway by regulating the expression of endosomal sorting complex related cellular proteins. CALML5 depletion also suppressed IFN-ß and IL-6 production in the PEDV-infected cells, thereby indicating its involvement in negatively regulating the innate immune response. Our study reveals the biological function of CALML5 in the virology field and offers new insights into the PEDV-host cell interaction.


Assuntos
Calmodulina , Endossomos , Imunidade Inata , Vírus da Diarreia Epidêmica Suína , Replicação Viral , Animais , Vírus da Diarreia Epidêmica Suína/imunologia , Vírus da Diarreia Epidêmica Suína/fisiologia , Suínos , Calmodulina/metabolismo , Calmodulina/genética , Endossomos/metabolismo , Endossomos/virologia , Interações Hospedeiro-Patógeno/imunologia , Doenças dos Suínos/virologia , Doenças dos Suínos/imunologia , Células Vero , Chlorocebus aethiops , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Infecções por Coronavirus/veterinária , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-6/imunologia , Interferon beta/genética , Interferon beta/imunologia , Interferon beta/metabolismo
20.
Front Immunol ; 15: 1380220, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38799458

RESUMO

African swine fever (ASF) is an acute hemorrhagic and devastating infectious disease affecting domestic pigs and wild boars. It is caused by the African swine fever virus (ASFV), which is characterized by genetic diversity and sophisticated immune evasion strategies. To facilitate infection, ASFV encodes multiple proteins to antagonize host innate immune responses, thereby contributing to viral virulence and pathogenicity. The molecular mechanisms employed by ASFV-encoded proteins to modulate host antiviral responses have not been comprehensively elucidated. In this study, it was observed that the ASFV MGF505-6R protein, a member of the multigene family 505 (MGF505), effectively suppressed the activation of the interferon-beta (IFN-ß) promoter, leading to reduced mRNA levels of antiviral genes. Additional evidence has revealed that MGF505-6R antagonizes the cGAS-STING signaling pathway by interacting with the stimulator of interferon genes (STING) for degradation in the autophagy-lysosomal pathway. The domain mapping revealed that the N-terminal region (1-260aa) of MGF505-6R is the primary domain responsible for interacting with STING, while the CTT domain of STING is crucial for its interaction with MGF505-6R. Furthermore, MGF505-6R also inhibits the activation of STING by reducing the K63-linked polyubiquitination of STING, leading to the disruption of STING oligomerization and TANK binding kinase 1 (TBK1) recruitment, thereby impairing the phosphorylation and nuclear translocation of interferon regulatory factor 3 (IRF3). Collectively, our study elucidates a novel strategy developed by ASFV MGF505-6R to counteract host innate immune responses. This discovery may offer valuable insights for further exploration of ASFV immune evasion mechanisms and antiviral strategies.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Proteínas de Membrana , Proteínas Virais , Animais , Vírus da Febre Suína Africana/imunologia , Vírus da Febre Suína Africana/genética , Suínos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Febre Suína Africana/imunologia , Febre Suína Africana/virologia , Febre Suína Africana/metabolismo , Proteínas Virais/imunologia , Proteínas Virais/metabolismo , Proteínas Virais/genética , Humanos , Imunidade Inata , Interferon Tipo I/metabolismo , Interferon Tipo I/imunologia , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/imunologia , Transdução de Sinais , Proteólise , Células HEK293 , Interações Hospedeiro-Patógeno/imunologia , Evasão da Resposta Imune , Interferon beta/metabolismo , Interferon beta/imunologia , Interferon beta/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...