Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 269
Filtrar
1.
Oxid Med Cell Longev ; 2022: 5397733, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35047106

RESUMO

The infection of coronavirus disease (COVID-19) seriously threatens human life. It is urgent to generate effective and safe specific antibodies (Abs) against the pathogenic elements of COVID-19. Mice were immunized with SARS-CoV-2 spike protein antigens: S ectodomain-1 (CoV, in short) mixed in Alum adjuvant for 2 times and boosted with CoV weekly for 6 times. A portion of mice were treated with Maotai liquor (MTL, in short) or/and heat stress (HS) together with CoV boosting. We observed that the anti-CoV Ab was successfully induced in mice that received the CoV/Alum immunization for 2 times. However, upon boosting with CoV, the CoV Ab production diminished progressively; spleen CoV Ab-producing plasma cell counts reduced, in which substantial CoV-specific Ab-producing plasma cells (sPC) were apoptotic. Apparent oxidative stress signs were observed in sPCs; the results were reproduced by exposing sPCs to CoV in the culture. The presence of MTL or/and HS prevented the CoV-induced oxidative stress in sPCs and promoted and stabilized the CoV Ab production in mice in re-exposure to CoV. In summary, CoV/Alum immunization can successfully induce CoV Ab production in mice that declines upon reexposure to CoV. Concurrent administration of MTL/HS stabilizes and promotes the CoV Ab production in mice.


Assuntos
Anticorpos Neutralizantes/biossíntese , Anticorpos Antivirais/biossíntese , Apoptose , COVID-19/imunologia , Plasmócitos/imunologia , SARS-CoV-2/fisiologia , Superóxido Dismutase-1/fisiologia , Adjuvantes Imunológicos , Bebidas Alcoólicas , Compostos de Alúmen , Enzima de Conversão de Angiotensina 2/fisiologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , COVID-19/enzimologia , Vacinas contra COVID-19/imunologia , Resposta ao Choque Térmico , Imunização Secundária , Imunogenicidade da Vacina , Janus Quinase 2/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Plasmócitos/efeitos dos fármacos , Plasmócitos/patologia , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT1/fisiologia , Transdução de Sinais , Organismos Livres de Patógenos Específicos , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinação
2.
J Immunother Cancer ; 10(1)2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35091454

RESUMO

BACKGROUND: Circulating monocytes are functionally heterogeneous and can be divided into classical (CMo), intermediate (IMo), and non-CMo/patrolling monocyte (PMo) subsets. CMo can differentiate into PMo through IMo. PMos have been shown to inhibit cancer metastasis but the role of IMo is unclear. To date, no strategy has been developed to inhibit cancer metastasis through enhancing PMo/IMo differentiation. METHODS: We screened multiple inflammatory cytokines/chemokines activity of modulating PMo/IMo associated cell markers expression using human monocyte in vitro culture system. We tested our candidate cytokine activity in vivo using multiple mice models. We identified critical key factors and cytokines for our candidate cytokine activity by using gene-knockout mice and neutralization antibodies. RESULTS: We identified IFN-γ as a candidate inflammatory cytokine in the regulation of human IMo/PMo marker expression. Our in vivo data demonstrated that IMo expansion was induced by short-term (3 days) IFN-γ treatment through increasing CMo-IMo differentiation and blocking IMo-PMo differentiation. The IMo induced by IFN-γ (IFN-IMo), but not IFN-γ activated CMo (IFN-CMo), inhibited cancer metastasis by 90%. Surprizing, the effect of IFN-γ is greater in PMo deficiency mice, indicating the effect of IFN-IMo is not mediated through further differentiation into PMo. We also found that IFN-IMos induced by short-term IFN-γ treatment robustly boosted NK cell expansion for threefold and promoted NK differentiation and function through IL-27 and CXCL9. Furthermore, we identified that FOXO1, a key molecule controlling cellular energy metabolism, mediated the effect of IFN-γ induced IL-27 expression, and that NR4A1, a key molecule controlling PMo differentiation and inhibiting cancer metastasis, inhibited the pro-NK cell and anti-metastasis activity of IFN-IMo by suppressing CXCL9 expression. CONCLUSIONS: We have discovered the antimetastasis and pro-NK cell activity of IFN-IMo, identified FOXO1 as a key molecule for IFN-γ driven monocyte differentiation and function, and found NR4A1 as an inhibitory molecule for IFN-IMo activity. Our study has not only shown novel mechanisms for a classical antitumor cytokine but also provided potential target for developing superior monocytic cell therapy against cancer metastasis.


Assuntos
Proteína Forkhead Box O1/fisiologia , Interferon gama/farmacologia , Interleucina-27/fisiologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Monócitos/efeitos dos fármacos , Metástase Neoplásica/prevenção & controle , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/fisiologia , Fator de Transcrição STAT1/fisiologia
3.
Biochem J ; 479(3): 385-399, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35084016

RESUMO

The apelin receptor (APLNR) regulates many biological processes including metabolism, angiogenesis, circulating blood volume and cardiovascular function. Additionally, APLNR is overexpressed in various types of cancer and influences cancer progression. APLNR is reported to regulate tumor recognition during immune surveillance by modulating the IFN-γ response. However, the mechanism of APLNR cross-talk with intratumoral IFN-γ signaling remains unknown. Here, we show that activation of APLNR up-regulates IFN-γ signaling in melanoma cells through APLNR mediated ß-arrestin 1 but not ß-arrestin 2 recruitment. Our data suggests that ß-arrestin 1 directly interacts with STAT1 to inhibit STAT1 phosphorylation to attenuate IFN-γ signaling. The APLNR mutant receptor, I109A, which is deficient in ß-arrestins recruitment, is unable to enhance intratumoral IFN-γ signaling. While APLNR N112G, a constitutively active mutant receptor, increases intratumoral sensitivity to IFN-γ signaling by enhancing STAT1 phosphorylation upon IFN-γ exposure. We also demonstrate in a co-culture system that APLNR regulates tumor survival rate. Taken together, our findings reveal that APLNR modulates IFN-γ signaling in melanoma cells and suggest that APLNR may be a potential target to enhance the efficacy of immunotherapy.


Assuntos
Receptores de Apelina/fisiologia , Interferon gama/fisiologia , Janus Quinases/fisiologia , Melanoma/metabolismo , Proteínas de Neoplasias/fisiologia , Fator de Transcrição STAT1/fisiologia , Transdução de Sinais/fisiologia , beta-Arrestina 1/fisiologia , Receptores de Apelina/antagonistas & inibidores , Receptores de Apelina/química , Receptores de Apelina/genética , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Células HEK293 , Humanos , Janus Quinases/antagonistas & inibidores , Melanoma/imunologia , Modelos Moleculares , Mapeamento de Interação de Proteínas , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Linfócitos T/imunologia , beta-Arrestina 2/análise
4.
Cancer Lett ; 523: 57-71, 2021 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-34563641

RESUMO

High fluence low-level laser (HF-LLL), a mitochondria-targeted tumour phototherapy, results in oxidative damage and apoptosis of tumour cells, as well as damage to normal tissue. To circumvent this, the therapeutic effect of low fluence LLL (LFL), a non-invasive and drug-free therapeutic strategy, was identified for tumours and the underlying molecular mechanisms were investigated. We observed that LFL enhanced antigen-specific immune response of macrophages and dendritic cells by upregulating MHC class II, which was induced by mitochondrial reactive oxygen species (ROS)-activated signalling, suppressing tumour growth in both CD11c-DTR and C57BL/6 mice. Mechanistically, LFL upregulated MHC class II in an MHC class II transactivator (CIITA)-dependent manner. LFL-activated protein kinase C (PKC) promoted the nuclear translocation of CIITA, as inhibition of PKC attenuated the DNA-binding efficiency of CIITA to MHC class II promoter. CIITA mRNA and protein expression also improved after LFL treatment, characterised by direct binding of Src and STAT1, and subsequent activation of STAT1. Notably, scavenging of ROS downregulated LFL-induced Src and PKC activation and antagonised the effects of LFL treatment. Thus, LFL treatment altered the adaptive immune response via the mitochondrial ROS-activated signalling pathway to control the progress of neoplastic disease.


Assuntos
Antígenos de Histocompatibilidade Classe II/imunologia , Terapia com Luz de Baixa Intensidade/métodos , Neoplasias Experimentais/terapia , Proteína Quinase C/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Linfócitos T/imunologia , Quinases da Família src/fisiologia , Transporte Ativo do Núcleo Celular , Animais , Apresentação de Antígeno , Células Dendríticas/fisiologia , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/metabolismo , Proteínas Nucleares/fisiologia , Fator de Transcrição STAT1/fisiologia , Transativadores/fisiologia
5.
Int J Mol Sci ; 22(14)2021 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-34299314

RESUMO

Signal transducer and activator of transcription 1 (STAT1) acts as a tumor suppressor molecule in colitis-associated colorectal cancer (CAC), particularly during the very early stages, modulating immune responses and controlling mechanisms such as apoptosis and cell proliferation. Previously, using an experimental model of CAC, we reported increased intestinal cell proliferation and faster tumor development, which were consistent with more signs of disease and damage, and reduced survival in STAT1-/- mice, compared with WT counterparts. However, the mechanisms through which STAT1 might prevent colorectal cancer progression preceded by chronic inflammation are still unclear. Here, we demonstrate that increased tumorigenicity related to STAT1 deficiency could be suppressed by IL-17 neutralization. The blockade of IL-17 in STAT1-/- mice reduced the accumulation of CD11b+Ly6ClowLy6G+ cells resembling granulocytic myeloid-derived suppressor cells (MDSCs) in both spleen and circulation. Additionally, IL-17 blockade reduced the recruitment of neutrophils into intestinal tissue, the expression and production of inflammatory cytokines, and the expression of intestinal STAT3. In addition, the anti-IL-17 treatment also reduced the expression of Arginase-1 and inducible nitric oxide synthase (iNOS) in the colon, both associated with the main suppressive activity of MDSCs. Thus, a lack of STAT1 signaling induces a significant change in the colonic microenvironment that supports inflammation and tumor formation. Anti-IL-17 treatment throughout the initial stages of CAC related to STAT1 deficiency abrogates the tumor formation possibly caused by myeloid cells.


Assuntos
Neoplasias Associadas a Colite/etiologia , Granulócitos/patologia , Interleucina-17/fisiologia , Fator de Transcrição STAT1/fisiologia , Animais , Anticorpos Neutralizantes/administração & dosagem , Neoplasias Associadas a Colite/patologia , Neoplasias Associadas a Colite/fisiopatologia , Progressão da Doença , Feminino , Granulócitos/imunologia , Interleucina-17/antagonistas & inibidores , Interleucina-17/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/patologia , Neoplasias Experimentais/etiologia , Neoplasias Experimentais/patologia , Neoplasias Experimentais/fisiopatologia , Fator de Transcrição STAT1/deficiência , Fator de Transcrição STAT1/genética , Microambiente Tumoral/imunologia
6.
J Virol ; 95(17): e0085921, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34132573

RESUMO

Gammaherpesviruses establish lifelong infections and are associated with B cell lymphomas. Murine gammaherpesvirus 68 (MHV68) infects epithelial and myeloid cells during acute infection, with subsequent passage of the virus to B cells, where physiological B cell differentiation is usurped to ensure the establishment of a chronic latent reservoir. Interferons (IFNs) represent a major antiviral defense system that engages the transcriptional factor STAT1 to attenuate diverse acute and chronic viral infections, including those of gammaherpesviruses. Correspondingly, global deficiency of type I or type II IFN signaling profoundly increases the pathogenesis of acute and chronic gammaherpesvirus infection, compromises host survival, and impedes mechanistic understanding of cell type-specific role of IFN signaling. Here, we demonstrate that myeloid-specific STAT1 expression attenuates acute and persistent MHV68 replication in the lungs and suppresses viral reactivation from peritoneal cells, without any effect on the establishment of viral latent reservoir in splenic B cells. All gammaherpesviruses encode a conserved protein kinase that antagonizes type I IFN signaling in vitro. Here, we show that myeloid-specific STAT1 deficiency rescues the attenuated splenic latent reservoir of the kinase-null MHV68 mutant. However, despite having gained access to splenic B cells, the protein kinase-null MHV68 mutant fails to drive B cell differentiation. Thus, while myeloid-intrinsic STAT1 expression must be counteracted by the gammaherpesvirus protein kinase to facilitate viral passage to splenic B cells, expression of the viral protein kinase continues to be required to promote optimal B cell differentiation and viral reactivation, highlighting the multifunctional nature of this conserved viral protein during chronic infection. IMPORTANCE IFN signaling is a major antiviral system of the host that suppresses replication of diverse viruses, including acute and chronic gammaherpesvirus infection. STAT1 is a critical member and the primary antiviral effector of IFN signaling pathways. Given the significantly compromised antiviral status of global type I or type II IFN deficiency, unabated gammaherpesvirus replication and pathogenesis hinders understanding of cell type-specific antiviral effects. In this study, a mouse model of myeloid-specific STAT1 deficiency unveiled site-specific antiviral effects of STAT1 in the lungs and peritoneal cavity, but not the spleen, of chronically infected hosts. Interestingly, expression of a conserved gammaherpesvirus protein kinase was required to counteract the antiviral effects of myeloid-specific STAT1 expression to facilitate latent infection of splenic B cells, revealing a cell type-specific virus-host antagonism during the establishment of chronic gammaherpesvirus infection.


Assuntos
Antivirais/farmacologia , Linfócitos B/virologia , Gammaherpesvirinae/enzimologia , Infecções por Herpesviridae/virologia , Proteínas Quinases/metabolismo , Fator de Transcrição STAT1/fisiologia , Latência Viral , Animais , Interações Hospedeiro-Patógeno , Interferons/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/metabolismo , Proteínas Quinases/genética , Baço/virologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Ativação Viral
7.
Int Urol Nephrol ; 53(6): 1247-1254, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33942213

RESUMO

AIM: High glucose (HG) induces the production of transforming growth factor (TGF)-ß and reactive oxygen species, which further activates JAK/STAT signaling and promotes the synthesis of matrix proteins, contributes to the pathophysiological processes of diabetic nephropathy. This study aims to investigate the protection role of vitamin D (VD) in the kidney in high glucose condition. METHODS: Rat glomerular mesangial cells were cultured in high glucose medium, with or without VD or VD receptor (VDR) siRNAs treatment. The levels of TGF-ß and fibronectin were detected by qRT-PCR, immunoblotting and enzyme-linked immunosorbent assay (ELISA). The levels of phosphorylated JAK2, STAT1 and STAT3, and JAK/STAT signaling downstream genes were examined by immunoblotting and qRT-PCR. RESULTS: In rat glomerular mesangial cells, VD treatment can repress the tyrosine phosphorylation of JAK2, STAT1 and STAT3. VD inhibited TGF-ß and fibronectin expression which was rescued by vitamin d receptor (VDR) siRNA and STATs inhibitor perficitinib. The JAK/STAT signaling downstream protein coding genes including SOCS1, SOCS3 and type IV collagen were repressed by VD. Meanwhile, the expression of non-coding RNAs such as miR-181a, miR-181b, was repressed by VD, and the expression of miR-34a and Let-7b was upregulated by VD. CONCLUSION: Vitamin D (VD) treatment inhibits the function of HG on fibronectin production through regulating JAK/STAT pathway. These results provide direct evidences that VD protects glomerular mesangial cells from high glucose-induced injury through repressing JAK/STAT signaling, which has the potential for clinical DN treatment.


Assuntos
Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/fisiologia , Células Mesangiais/efeitos dos fármacos , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT1/fisiologia , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/fisiologia , Transdução de Sinais/efeitos dos fármacos , Vitamina D/farmacologia , Vitaminas/farmacologia , Animais , Células Cultivadas , Glucose/metabolismo , Masculino , Células Mesangiais/fisiologia , Ratos , Ratos Sprague-Dawley
8.
Vet Res ; 51(1): 135, 2020 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-33176874

RESUMO

Duck enteritis virus (DEV) is a member of the Alphaherpesvirinae subfamily. The characteristics of some DEV genes have been reported. However, information regarding the DEV UL47 gene is limited. In this study, we identified the DEV UL47 gene encoding a late structural protein located in the nucleus of infected cells. We further found that two domains of DEV pUL47, amino acids (aa) 40 to 50 and 768 to 777, could function as nuclear localization sequence (NLS) to guide the nuclear localization of pUL47 and nuclear translocation of heterologous proteins, including enhanced green fluorescent protein (EGFP) and beta-galactosidase (ß-Gal). Moreover, pUL47 significantly inhibited polyriboinosinic:polyribocytidylic acid [poly(I:C)]-induced interferon beta (IFN-ß) production and downregulated interferon-stimulated gene (ISG) expression, such as Mx and oligoadenylate synthetase-like (OASL), by interacting with signal transducer and activator of transcription-1 (STAT1).


Assuntos
Patos , Interferon beta/fisiologia , Mardivirus/fisiologia , Doença de Marek/virologia , Doenças das Aves Domésticas/virologia , Fator de Transcrição STAT1/fisiologia , Proteínas Estruturais Virais/genética , Animais , Núcleo Celular/virologia , Transdução de Sinais
9.
PLoS Pathog ; 16(10): e1008973, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33045014

RESUMO

The liver is a central regulator of metabolic homeostasis and serum metabolite levels. Hepatocytes are the functional units of the liver parenchyma and not only responsible for turnover of biomolecules but also act as central immune signaling platforms. Hepatotropic viruses infect liver tissue, resulting in inflammatory responses, tissue damage and hepatitis. Combining well-established in vitro and in vivo model systems with transcriptomic analyses, we show that type I interferon signaling initiates a robust antiviral immune response in hepatocytes. Strikingly, we also identify IFN-I as both, sufficient and necessary, to induce wide-spread metabolic reprogramming in hepatocytes. IFN-I specifically rewired tryptophan metabolism and induced hepatic tryptophan oxidation to kynurenine via Tdo2, correlating with altered concentrations of serum metabolites upon viral infection. Infected Tdo2-deficient animals displayed elevated serum levels of tryptophan and, unexpectedly, also vast increases in the downstream immune-suppressive metabolite kynurenine. Thus, Tdo2-deficiency did not result in altered serum homeostasis of the tryptophan to kynurenine ratio during infection, which seemed to be independent of hepatocyte-intrinsic compensation via the IDO-axis. These data highlight that inflammation-induced reprogramming of systemic tryptophan metabolism is tightly regulated in viral hepatitis.


Assuntos
Antivirais/metabolismo , Hepatite Viral Animal/imunologia , Hepatócitos/imunologia , Inflamação/imunologia , Cinurenina/metabolismo , Receptor de Interferon alfa e beta/fisiologia , Triptofano/metabolismo , Animais , Feminino , Vírus de Hepatite/isolamento & purificação , Hepatite Viral Animal/metabolismo , Hepatite Viral Animal/virologia , Hepatócitos/metabolismo , Hepatócitos/virologia , Humanos , Imunidade Inata/imunologia , Inflamação/metabolismo , Inflamação/patologia , Inflamação/virologia , Fator Regulador 7 de Interferon/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição STAT1/fisiologia , Triptofano Oxigenase/fisiologia
10.
Int J Mol Sci ; 21(19)2020 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-32987855

RESUMO

STAT1 and STAT3 are nuclear transcription factors that regulate genes involved in cell cycle, cell survival and immune response. The cross-talk between these signaling pathways determines how cells integrate the environmental signals received ultimately translating them in transcriptional regulation of specific sets of genes. Despite being activated downstream of common cytokine and growth factors, STAT1 and STAT3 play essentially antagonistic roles and the disruption of their balance directs cells from survival to apoptotic cell death or from inflammatory to anti-inflammatory responses. Different mechanisms are proposed to explain this yin-yang relationship. Considering the redox aspect of STATs proteins, this review attempts to summarize the current knowledge of redox regulation of STAT1 and STAT3 signaling focusing the attention on the post-translational modifications that affect their activity.


Assuntos
Fator de Transcrição STAT1/fisiologia , Fator de Transcrição STAT3/fisiologia , Transdução de Sinais , Regulação da Expressão Gênica , Humanos , Oxirredução
11.
Commun Biol ; 3(1): 252, 2020 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-32444775

RESUMO

Tumors have evolved mechanisms to escape anti-tumor immunosurveillance. They limit humoral and cellular immune activities in the stroma and render tumors resistant to immunotherapy. Sensitizing tumor cells to immune attack is an important strategy to revert immunosuppression. However, the underlying mechanisms of immune escape are still poorly understood. Here we discover Indoleamine-2,3-dioxygenase-1 (IDO1)+ Paneth cells in the stem cell niche of intestinal crypts and tumors, which promoted immune escape of colorectal cancer (CRC). Ido1 expression in Paneth cells was strictly Stat1 dependent. Loss of IDO1+ Paneth cells in murine intestinal adenomas with tumor cell-specific Stat1 deletion had profound effects on the intratumoral immune cell composition. Patient samples and TCGA expression data suggested corresponding cells in human colorectal tumors. Thus, our data uncovered an immune escape mechanism of CRC and identify IDO1+ Paneth cells as a target for immunotherapy.


Assuntos
Neoplasias Colorretais/patologia , Tolerância Imunológica/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Neoplasias Intestinais/patologia , Celulas de Paneth/imunologia , Fator de Transcrição STAT1/fisiologia , Animais , Neoplasias Colorretais/etiologia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Neoplasias Intestinais/imunologia , Neoplasias Intestinais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
12.
J Physiol Biochem ; 76(1): 147-157, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32037480

RESUMO

Low shear stress (LSS) occurs in areas where atherosclerosis is prevalent. Many studies have revealed that signal transducer and activator of transcription 1 (STAT1) plays a significant role in cardiovascular disease. Nonetheless, the mechanism underlying the connection between STAT1 and LSS is not fully understood. The purpose of this study was to investigate the link between LSS and STAT1 in endothelial cells (ECs). Monolayer endothelial cells were stimulated or not stimulated by LSS. Protein expression and phosphorylation levels were determined by western blotting. Immunofluorescence was used to compare the protein expression differences in bifurcated and non-bifurcated human coronary arteries. Endothelial function was assessed by using a dihydroethidium assay, real-time PCR, western blotting and nitric oxide (NO)-sensitive fluorophore. Results showed that STAT1 played a key role in LSS-induced endothelium damage. Firstly, LSS activated STAT1, as evidenced by LSS-induced STAT1 (Tyr701) phosphorylation in ECs in vitro and the increased intimal STAT1 expression at bifurcation of human coronary arteries. Secondly, LSS-induced STAT1 phosphorylation was positively regulated by inhibitor of nuclear factor kappa-B kinase ε (IKKε). Additionally, LSS-promoted inflammatory factor expression was markedly reversed by silencing STAT1 (siSTAT1). LSS also increased reactive oxygen species (ROS) level and decreased endogenous NO release: however, siSTAT1 reversed these adverse effects through upregulating the antioxidant gene heme oxygenase-1(HO-1) and downregulating endothelial nitric oxide synthase (eNOS) Thr495 phosphorylation. According to our results, LSS-mediated EC injury may be associated with the activation of STAT1. Strategies designed to reduce STAT1 expression or inhibit STAT1 activation may be effective approaches for reducing the incidence of atherosclerosis.


Assuntos
Vasos Coronários/metabolismo , Endotélio Vascular/metabolismo , Fator de Transcrição STAT1/fisiologia , Estresse Mecânico , Células Cultivadas , Doença da Artéria Coronariana , Vasos Coronários/patologia , Endotélio Vascular/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação/metabolismo , Óxido Nítrico/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo
13.
Int J Radiat Biol ; 96(5): 642-650, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31914348

RESUMO

Purpose: Pulmonary inflammation is an adverse consequence of radiation therapy in breast cancer. The aim of this study was to elucidate biological pathways leading to this pathology.Materials and methods: Lung endothelial cells were isolated 24 h after thorax-irradiation (sham or 10 Gy X-ray) from female C57Bl/6 mice and cultivated for 6 days.Results: Quantitative proteomic analysis of lung endothelial cells was done using data independent acquisition (DIA) mass spectrometry. The data were analyzed using Ingenuity Pathway Analysis and STRINGdb. In total, 4220 proteins were identified using DIA of which 60 were dysregulated in the irradiated samples (fold change ≥2.00 or ≤0.50; q-value <0.05). Several (12/40) upregulated proteins formed a cluster of inflammatory proteins with STAT1 and IRF3 as predicted upstream regulators. The several-fold increased expression of STAT1 and STAT-associated ISG15 was confirmed by immunoblotting. The expression of antioxidant proteins SOD1 and PRXD5 was downregulated suggesting radiation-induced oxidative stress. Similarly, the phosphorylated (active) forms of STING and IRF3, both members of the cGAS/STING pathway, were downregulated.Conclusions: These data suggest the involvement of JAK/STAT and cGas/STING pathways in the genesis of radiation-induced lung inflammation. These pathways may be used as novel targets for the prevention of radiation-induced lung damage.


Assuntos
Células Endoteliais/efeitos da radiação , Inflamação/etiologia , Pulmão/efeitos da radiação , Espectrometria de Massas/métodos , Fator de Transcrição STAT1/fisiologia , Animais , Feminino , Fator Regulador 3 de Interferon/fisiologia , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteômica , Transdução de Sinais
14.
PLoS Biol ; 17(11): e3000530, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31730616

RESUMO

Type I interferon (IFN-I) is a family of multifunctional cytokines that modulate the innate and adaptive immunity and are used to treat mastocytosis. Although IFN-I is known to suppress mast cell function, including histamine release, the mechanisms behind its effects on mast cells have been poorly understood. We here investigated IFN-I's action on mast cells using interferon-α/ß receptor subunit 1 (Ifnar1)-deficient mice, which lack a functional IFN-I receptor complex, and revealed that IFN-I in the steady state is critical for mast cell homeostasis, the disruption of which is centrally involved in systemic anaphylaxis. Ifnar1-deficient mice showed exacerbated systemic anaphylaxis after sensitization, which was associated with increased histamine in the circulation, even though the mast cell numbers and high affinity immunoglobulin E receptor (FcεRI) expression levels were similar between Ifnar1-deficient and wild-type (WT) mice. Ifnar1-deficient mast cells showed increased secretory granule synthesis and exocytosis, which probably involved the increased transcription of Tfeb. Signal transducer and activator of transcription 1(Stat1) and Stat2 were unexpectedly insufficient to mediate these IFN-I functions, and instead, Stat3 played a critical role in a redundant manner with Stat1. Our findings revealed a novel regulation mechanism of mast cell homeostasis, in which IFN-I controls lysosome-related organelle biogenesis.


Assuntos
Anafilaxia/imunologia , Interferon Tipo I/fisiologia , Mastócitos/imunologia , Vesículas Secretórias/metabolismo , Animais , Células Cultivadas , Histamina/sangue , Homeostase , Camundongos , Receptor de Interferon alfa e beta/genética , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/fisiologia , Fator de Transcrição STAT2/genética , Fator de Transcrição STAT2/fisiologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/fisiologia , Transdução de Sinais
15.
J Clin Invest ; 129(12): 5553-5567, 2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31710313

RESUMO

Immune checkpoint inhibitors (ICIs), although promising, have variable benefit in head and neck cancer (HNC). We noted that tumor galectin-1 (Gal1) levels were inversely correlated with treatment response and survival in patients with HNC who were treated with ICIs. Using multiple HNC mouse models, we show that tumor-secreted Gal1 mediates immune evasion by preventing T cell migration into the tumor. Mechanistically, Gal1 reprograms the tumor endothelium to upregulate cell-surface programmed death ligand 1 (PD-L1) and galectin-9. Using genetic and pharmacological approaches, we show that Gal1 blockade increases intratumoral T cell infiltration, leading to a better response to anti-PD1 therapy with or without radiotherapy. Our study reveals the function of Gal1 in transforming the tumor endothelium into an immune-suppressive barrier and that its inhibition synergizes with ICIs.


Assuntos
Antígeno B7-H1/antagonistas & inibidores , Endotélio/fisiologia , Galectina 1/fisiologia , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Linfócitos T/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antígeno B7-H1/fisiologia , Feminino , Galectina 1/antagonistas & inibidores , Galectinas/fisiologia , Neoplasias de Cabeça e Pescoço/imunologia , Humanos , Tolerância Imunológica , Imunoterapia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Fator de Transcrição STAT1/fisiologia
16.
PLoS One ; 14(9): e0222697, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31545817

RESUMO

Dendritic cells (DCs) are professional antigen presenting cells involved in the induction of T cell-mediated adaptive immunity. Plasmacytoid DCs (pDCs) originate from lymphoid precursors and produce type I interferons (IFNs) in response to pathogens. A20 is considered as a negative regulator of toll-like receptor (TLR) signaling pathways, in which Toxoplasma gondii- derived profilin (TgPRF) is a TLR11/12 ligand recognised by DCs to stimulate their maturation/activation. Little is known about contributions of A20 to changes in biological properties of pDCs. The present study, therefore, explored whether pDC functions are influenced by A20. To this end, bone marrow cells were isolated and cultured with Flt3L to attain CD8DCs, CD11bDCs and pDCs and followed by challenge with TgPRP in the presence or absence of A20 siRNA. Expression of maturation markers were analysed by flow cytometry, and secretion of inflammatory cytokines by ELISA, cell migration by a transwell migration assay and expression of signalling molecules by western blotting. As a result, treatment with A20 siRNA enhanced activations of IκB-α and STAT-1, leading to increases in expressions of maturation markers and cytokine productions as well as migration of TgPRP-treated pDCs, while mature CD11bDCs produced at higher levels of TNF-α and IL-6 only. In addition, functions of CD8DCs remained unaltered following A20 silencing. The effects of A20 on pDC maturation and activation were completely abolished by IKK inhibitor and partially blunted by fludarabine. In conclusion, the inhibitory effects of A20 on pDC functions are expected to affect the immune response in T. gondii infection.


Assuntos
Células Dendríticas/fisiologia , NF-kappa B/fisiologia , Fator de Transcrição STAT1/fisiologia , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/fisiologia , Animais , Western Blotting , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Citometria de Fluxo , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Profilinas/farmacologia , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Toxoplasma/metabolismo
17.
PLoS Pathog ; 15(4): e1007709, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-31017981

RESUMO

Norovirus infection is the leading cause of food-borne gastroenteritis worldwide, being responsible for over 200,000 deaths annually. Studies with murine norovirus (MNV) showed that protective STAT1 signaling controls viral replication and pathogenesis, but the immune mechanisms that noroviruses exploit to induce pathology are elusive. Here, we show that gastrointestinal MNV infection leads to widespread IL-1ß maturation in MNV-susceptible STAT1-deficient mice. MNV activates the canonical Nlrp3 inflammasome in macrophages, leading to maturation of IL-1ß and to Gasdermin D (GSDMD)-dependent pyroptosis. STAT1-deficient macrophages displayed increased MAVS-mediated expression of pro-IL-1ß, facilitating elevated Nlrp3-dependent release of mature IL-1ß upon MNV infection. Accordingly, MNV-infected Stat1-/- mice showed Nlrp3-dependent maturation of IL-1ß as well as Nlrp3-dependent pyroptosis as assessed by in vivo cleavage of GSDMD to its active N-terminal fragment. While MNV-induced diarrheic responses were not affected, Stat1-/- mice additionally lacking either Nlrp3 or GSDMD displayed lower levels of the fecal inflammatory marker Lipocalin-2 as well as delayed lethality after gastrointestinal MNV infection. Together, these results uncover new insights into the mechanisms of norovirus-induced inflammation and cell death, thereby revealing Nlrp3 inflammasome activation and ensuing GSDMD-driven pyroptosis as contributors to MNV-induced immunopathology in susceptible STAT1-deficient mice.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Infecções por Caliciviridae/patologia , Trato Gastrointestinal/patologia , Inflamassomos/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose/fisiologia , Fator de Transcrição STAT1/fisiologia , Animais , Proteínas Reguladoras de Apoptose/genética , Infecções por Caliciviridae/imunologia , Infecções por Caliciviridae/metabolismo , Infecções por Caliciviridae/virologia , Células Cultivadas , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/virologia , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Norovirus/imunologia , Norovirus/patogenicidade , Proteínas de Ligação a Fosfato
18.
PLoS Pathog ; 15(4): e1007745, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-31009517

RESUMO

The mechanisms by which the gut luminal environment is disturbed by the immune system to foster pathogenic bacterial growth and survival remain incompletely understood. Here, we show that STAT2 dependent type I IFN signaling contributes to the inflammatory environment by disrupting hypoxia enabling the pathogenic S. Typhimurium to outgrow the microbiota. Stat2-/- mice infected with S. Typhimurium exhibited impaired type I IFN induced transcriptional responses in cecal tissue and reduced bacterial burden in the intestinal lumen compared to infected wild-type mice. Although inflammatory pathology was similar between wild-type and Stat2-/- mice, we observed decreased hypoxia in the gut tissue of Stat2-/- mice. Neutrophil numbers were similar in wild-type and Stat2-/- mice, yet Stat2-/- mice showed reduced levels of myeloperoxidase activity. In vitro, the neutrophils from Stat2-/- mice produced lower levels of superoxide anion upon stimulation with the bacterial ligand N-formylmethionyl-leucyl-phenylalanine (fMLP) in the presence of IFNα compared to neutrophils from wild-type mice, indicating that the neutrophils were less functional in Stat2-/- mice. Cytochrome bd-II oxidase-mediated respiration enhances S. Typhimurium fitness in wild-type mice, while in Stat2-/- deficiency, this respiratory pathway did not provide a fitness advantage. Furthermore, luminal expansion of S. Typhimurium in wild-type mice was blunted in Stat2-/- mice. Compared to wild-type mice which exhibited a significant perturbation in Bacteroidetes abundance, Stat2-/- mice exhibited significantly less perturbation and higher levels of Bacteroidetes upon S. Typhimurium infection. Our results highlight STAT2 dependent type I IFN mediated inflammation in the gut as a novel mechanism promoting luminal expansion of S. Typhimurium.


Assuntos
Disbiose/imunologia , Gastroenterite/imunologia , Inflamação/imunologia , Interferon Tipo I/imunologia , Fator de Transcrição STAT2/fisiologia , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Animais , Células Cultivadas , Disbiose/metabolismo , Disbiose/patologia , Feminino , Gastroenterite/metabolismo , Gastroenterite/microbiologia , Gastroenterite/patologia , Inflamação/metabolismo , Inflamação/microbiologia , Inflamação/patologia , Interferon Tipo I/metabolismo , Intestinos/imunologia , Intestinos/microbiologia , Intestinos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Neutrófilos/patologia , Fator de Transcrição STAT1/fisiologia , Infecções por Salmonella/metabolismo , Infecções por Salmonella/microbiologia , Infecções por Salmonella/patologia
19.
Brain Behav Immun ; 79: 174-185, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30711510

RESUMO

Neuroinflammation occurs after germinal matrix hemorrhage (GMH) and induces secondary brain injury. Interferon-α (IFN-α) has been shown to exert anti-inflammatory effects in infectious diseases via activating IFNAR and its downstream signaling. We aimed to investigate the anti-inflammatory effects of Recombinant human IFN-α (rh-IFN-α) and the underlying mechanisms in a rat GMH model. Two hundred and eighteen P7 rat pups of both sexes were subjected to GMH by an intraparenchymal injection of bacterial collagenase. Rh-IFN-α was administered intraperitoneally. Small interfering RNA (siRNA) of IFNAR, and siRNA of tumor necrosis factor receptor associated factor 3 (TRAF3) were administered through intracerebroventricular (i.c.v.) injections. JAK1 inhibitor ruxolitinib was given by oral lavage. Post-GMH evaluation included neurobehavioral function, Nissl staining, Western blot analysis, and immunofluorescence. Our results showed that endogenous IFN-α and phosphorylated IFNAR levels were increased after GMH. Administration of rh-IFN-α improved neurological functions, attenuated neuroinflammation, inhibited microglial activation, and ameliorated post-hemorrhagic hydrocephalus after GMH. These observations were concomitant with IFNAR activation, increased expression of phosphorylated JAK1, phosphorylated STAT1 and TRAF3, and decreased levels of phosphorylated NF-κB, IL-6 and TNF-α. Specifically, knockdown of IFNAR, JAK1 and TRAF3 abolished the protective effects of rh-IFN-α. In conclusion, our findings demonstrated that rh-IFN-α treatment attenuated neuroinflammation, neurological deficits and hydrocephalus formation through inhibiting microglial activation after GMH, which might be mediated by IFNAR/JAK1-STAT1/TRAF3/NF-κB signaling pathway. Rh-IFN-α may be a promising therapeutic agent to attenuate brain injury via its anti-inflammatory effect.


Assuntos
Hemorragia Cerebral Intraventricular/imunologia , Interferon-alfa/metabolismo , Neuroimunomodulação/fisiologia , Animais , Animais Recém-Nascidos , Lesões Encefálicas/metabolismo , Hemorragia Cerebral Intraventricular/induzido quimicamente , Hemorragia Cerebral Intraventricular/fisiopatologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Proteínas I-kappa B/metabolismo , Inflamação/metabolismo , Interferon-alfa/farmacologia , Interferon-alfa/fisiologia , Janus Quinase 1/metabolismo , Janus Quinase 1/fisiologia , Masculino , Microglia/metabolismo , NF-kappa B/imunologia , NF-kappa B/metabolismo , Neuroimunomodulação/imunologia , Ratos , Ratos Sprague-Dawley , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT1/fisiologia , Transdução de Sinais/efeitos dos fármacos , Fator 3 Associado a Receptor de TNF/metabolismo
20.
Cell Rep ; 26(9): 2394-2406.e5, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30811989

RESUMO

Cytomegalovirus (CMV) has a high prevalence worldwide, is often fatal for immunocompromised patients, and causes bone marrow suppression. Deficiency of signal transducer and activator of transcription 1 (STAT1) results in severely impaired antiviral immunity. We have used cell-type restricted deletion of Stat1 to determine the importance of myeloid cell activity for the defense against murine CMV (MCMV). We show that myeloid STAT1 limits MCMV burden and infection-associated pathology in the spleen but does not affect ultimate clearance of infection. Unexpectedly, we found an essential role of myeloid STAT1 in the induction of extramedullary hematopoiesis (EMH). The EMH-promoting function of STAT1 was not restricted to MCMV infection but was also observed during CpG oligodeoxynucleotide-induced sterile inflammation. Collectively, we provide genetic evidence that signaling through STAT1 in myeloid cells is required to restrict MCMV at early time points post-infection and to induce compensatory hematopoiesis in the spleen.


Assuntos
Hematopoese Extramedular , Infecções por Herpesviridae/fisiopatologia , Muromegalovirus , Células Mieloides/fisiologia , Fator de Transcrição STAT1/fisiologia , Animais , Células Cultivadas , Feminino , Deleção de Genes , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/metabolismo , Células Matadoras Naturais/imunologia , Masculino , Camundongos Endogâmicos C57BL , Muromegalovirus/fisiologia , Receptor de Interferon alfa e beta/genética , Receptores de Interferon/genética , Receptores de Interleucina/genética , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Baço/patologia , Baço/virologia , Estresse Fisiológico , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA