Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 270
Filtrar
1.
Neuromolecular Med ; 26(1): 16, 2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38668900

RESUMO

Toll-like receptor (TLR) 7 plays an important role in recognizing virus-derived nucleic acids. TLR7 signaling in astrocytes and microglia is critical for activating immune responses against neurotrophic viruses. Neurons express TLR7, similar to glial cells; however, the role of neuronal TLR7 has not yet been fully elucidated. This study sought to determine whether resiquimod, the TLR7/8 agonist, induces the expression of inflammatory chemokines in SH-SY5Y human neuroblastoma cells. Immunofluorescence microscopy revealed that TLR7 was constitutively expressed in SH-SY5Y cells. Stimulation with resiquimod induced C-C motif chemokine ligand 2 (CCL2) expression, accompanied by the activation of nuclear factor-kappa B (NF-κB) in SH-SY5Y cells. Resiquimod increased mRNA levels of C-X-C motif chemokine ligand 8 (CXCL8) and CXCL10, while the increase was slight at the protein level. Knockdown of NF-κB p65 eliminated resiquimod-induced CCL2 production. This study provides novel evidence that resiquimod has promising therapeutic potential against central nervous system viral infections through its immunostimulatory effects on neurons.


Assuntos
Quimiocina CCL2 , Quimiocina CXCL10 , Imidazóis , Interleucina-8 , Receptor 7 Toll-Like , Fator de Transcrição RelA , Humanos , Linhagem Celular Tumoral , Quimiocina CCL2/genética , Quimiocina CCL2/biossíntese , Quimiocina CXCL10/genética , Quimiocina CXCL10/biossíntese , Imidazóis/farmacologia , Interleucina-8/genética , Interleucina-8/biossíntese , Neuroblastoma , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , NF-kappa B/metabolismo , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/genética , Receptor 8 Toll-Like/agonistas , Receptor 8 Toll-Like/genética , Fator de Transcrição RelA/metabolismo , Fator de Transcrição RelA/genética
2.
Innate Immun ; 28(5): 155-163, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35548957

RESUMO

Th17 cells represent important immune cells. Ursolic acid (UA) can regulate immune cell activities. This study was aimed to explore the effects of UA on Th17 cell differentiation and Schwann cell(SCs)-mediated migration and the potential mechanism. Naïve CD4+ T cells were isolated from rat peripheral blood, induced for Th17 cell differentiation, and treated with UA. The proportion of Th17 cells was detected by flow cytometry assay. SCs were co-cultured with Th17 cells. Th17 cell migration was detected by Transwell assay. The molecule expression was determined by Western blot and qRT-PCR. UA inhibited the Th17 cell differentiation and suppressed the STAT3/RORγt pathway. STAT3 overexpression up-regulated p-STAT3 and RORγt expression and promoted Th17 cell differentiation under the UA treatment. In LPS- and IFN-γ-stimulated-SCs, UA suppressed the expression of chemokines CXCL9/10, but had no significant effect of CCL20 expression. The expression CXCL9/10 receptor CXCR3 was higher in Th17 cells than that in Treg cells, while the expression CCL20 receptor CCR6 was lower in Th17 cells than that in Treg cells. UA, anti-CXCR3, and anti-CCR6 treatment inhibited SCs-mediated Th17 cell migration, and anti-CXCR3 exerted stronger inhibitory effect than Anti-CCR6. UA inhibited Th17 cell differentiation through STAT3/RORγt pathway and suppressed Th17 cell migration through down-regulating CXCL9/10 expression in SCs.


Assuntos
Quimiocina CXCL10 , Quimiocina CXCL9 , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Fator de Transcrição STAT3 , Células de Schwann , Células Th17 , Triterpenos , Animais , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Quimiocina CXCL10/biossíntese , Quimiocina CXCL10/metabolismo , Quimiocina CXCL9/biossíntese , Quimiocina CXCL9/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Ratos , Fator de Transcrição STAT3/metabolismo , Células de Schwann/citologia , Células de Schwann/efeitos dos fármacos , Células de Schwann/metabolismo , Células Th17/citologia , Células Th17/efeitos dos fármacos , Células Th17/metabolismo , Triterpenos/farmacologia , Ácido Ursólico
3.
PLoS One ; 17(2): e0263370, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35113934

RESUMO

The cytokine Interferon-γ (IFN-γ) exerts powerful immunoregulatory effects on the adaptive immune system and also enhances functions of the neutrophil (PMN). The clinical use of IFN-γ has been driven by the finding that its administration to patients with chronic granulomatous disease (CGD) results in decreased incidence and severity of infections. However, IFN-γ has no effect on the characteristic defect of CGD, the inability to convert oxygen to microbicidal metabolites including superoxide anion (O2-) during the phagocytosis associated oxidative burst. We administered varying doses of IFN-γ to adult volunteers and studied the effects on plasma drug levels and response molecules and PMNs isolated from blood drawn at intervals over a 96- hour period. Plasma concentrations of IFN-γ, IP-10 and neopterin, and stimulated release of O2- from PMNs exhibited dose- and time-dependent increases after IFN-γ administration. Gene expression in PMNs was altered for 2775 genes; changes occurred rapidly after administration and returned to baseline in 24-36 hours. Several genes involved with neutrophil host defense were upregulated including those for components of the O2- generating NADPH oxidase; innate-immune and Fc receptors; proteins involved in MHCI and II; a regulator of circulating PMN number; guanylate binding proteins; and a key enzyme in synthesis of an essential NOS cofactor. Coordinate changes were detected in protein levels of representative products from several of these genes. Lysates from isolated neutrophils also demonstrated a spike in NO following IFN-γ administration. IFN-γ appears to increase non-oxygen dependent microbicidal functions of PMNs which could provide strategies to compensate for deficiencies, explain its clinical benefit for CGD patients and expand therapeutic applications of IFN-γ to other disorders. Trial registration: Protocol registered in ClinicalTrials.gov, NCT02609932, Effect of IFN-γ on Innate Immune Cells.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Doença Granulomatosa Crônica/tratamento farmacológico , Doença Granulomatosa Crônica/metabolismo , Interferon gama/farmacologia , Neutrófilos/efeitos dos fármacos , Adolescente , Adulto , Quimiocina CXCL10/biossíntese , Doença Granulomatosa Crônica/genética , Voluntários Saudáveis , Humanos , Interferon gama/biossíntese , Pessoa de Meia-Idade , NADPH Oxidases/metabolismo , Neopterina/biossíntese , Neutrófilos/metabolismo , Fagocitose , Fenótipo , Explosão Respiratória , Superóxidos , Adulto Jovem
4.
Biomed Res Int ; 2022: 6803154, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35036436

RESUMO

BACKGROUND: The energy delivered by a ventilator to the respiratory system in one minute is defined as mechanical power (MP). However, the effect of ventilator-induced lung injury (VILI) in patients suffering from acute respiratory distress syndrome (ARDS) is still unknown. Our previous studies revealed that CXCL10 may be a potential biomarker of lung injury in ARDS. Therefore, the aim of this study was to compare the lung injury of rats and patients under different MP conditions to explore the involvement of CXCL10 and its receptor CXCR3 in VILI. METHODS: Patients were divided into the high mechanical power group (HMPp group) and low mechanical power group (LMPp group), while rats were assigned to the high mechanical power group (HMPr group), medium mechanical power group (MMPr group), and low mechanical power group (LMPr group). CXCL10 and CXCR3 plasma content in ARDS patients and rats under ventilation at different MP was measured, as well as their protein and mRNA expression in rat lungs. RESULTS: CXCL10 and CXCR3 content in the plasma of ARDS patients in the HMPp was significantly higher than that in the LMPp. The increase of MP during mechanical ventilation in the rats gradually increased lung damage, and CXCL10 and CXCR3 levels in rat plasma gradually increased with the increase of MP. CXCL10 and CXCR3 protein and mRNA expression in the HMPr group and MMPr group was significantly higher than that in the LMPr group (P < 0.05). More mast cells were present in the trachea, bronchus, blood vessels, and lymphatic system in the rat lungs of the HMPr group, and the number of mast cells in the HMPr group (13.32 ± 3.27) was significantly higher than that in the LMPr group (3.25 ± 0.29) (P < 0.05). CONCLUSION: The higher the MP, the more severe the lung injury, and the higher the CXCL10/CXCR3 expression. Therefore, CXCL10/CXCR3 might participate in VILI by mediating mast cell chemotaxis.


Assuntos
Quimiocina CXCL10/biossíntese , Regulação da Expressão Gênica , Pulmão/metabolismo , Receptores CXCR3/biossíntese , Respiração Artificial/efeitos adversos , Síndrome do Desconforto Respiratório/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/metabolismo , Idoso , Animais , Biomarcadores/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Ratos , Ratos Sprague-Dawley , Síndrome do Desconforto Respiratório/terapia
5.
J Neuroimmunol ; 360: 577715, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34536787

RESUMO

This post-hoc analysis evaluated candidate biomarkers of long-term efficacy of subcutaneous interferon beta-1a (sc IFN ß-1a) in REFLEX/REFLEXION studies of clinically isolated syndrome. Samples from 507 REFLEX and 287 REFLEXION study participants were analyzed. All investigated biomarkers were significantly upregulated 1.5-4-fold in response to sc IFN ß-1a treatment versus baseline (p ≤ 0.008). The validity of MX1, 2'5'OAS, and IL-1RA as biomarkers of response to sc IFN ß-1a was confirmed in this large patient cohort, with biomarkers consistently upregulated in a dose-dependent manner. Neopterin, TRAIL, and IP-10 were confirmed as biomarkers associated with long-term sc IFN ß-1a treatment efficacy over 5 years.


Assuntos
Interferon beta-1a/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , 2',5'-Oligoadenilato Sintetase/biossíntese , 2',5'-Oligoadenilato Sintetase/sangue , 2',5'-Oligoadenilato Sintetase/genética , Biomarcadores , Quimiocina CXCL10/biossíntese , Quimiocina CXCL10/sangue , Quimiocina CXCL10/genética , Relação Dose-Resposta a Droga , Método Duplo-Cego , Seguimentos , Humanos , Injeções Subcutâneas , Interferon beta-1a/administração & dosagem , Interferon beta-1a/farmacocinética , Proteína Antagonista do Receptor de Interleucina 1/biossíntese , Proteína Antagonista do Receptor de Interleucina 1/sangue , Proteína Antagonista do Receptor de Interleucina 1/genética , Estudos Multicêntricos como Assunto , Esclerose Múltipla/sangue , Proteínas de Resistência a Myxovirus/biossíntese , Proteínas de Resistência a Myxovirus/sangue , Proteínas de Resistência a Myxovirus/genética , Neopterina/biossíntese , Neopterina/sangue , Neopterina/genética , Ensaios Clínicos Controlados Aleatórios como Assunto/estatística & dados numéricos , Ligante Indutor de Apoptose Relacionado a TNF/biossíntese , Ligante Indutor de Apoptose Relacionado a TNF/sangue , Ligante Indutor de Apoptose Relacionado a TNF/genética , Regulação para Cima
6.
Front Immunol ; 12: 718744, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34531865

RESUMO

COVID-19 associated multisystem inflammatory syndrome (MIS) is a rare condition mostly affecting children but also adults (MIS-A). Although severe systemic inflammation and multiorgan dysfunction are hallmarks of the syndrome, the underlying pathogenesis is unclear. We aimed to provide novel immunological and genetic descriptions of MIS-A patients. Cytokine responses (IL-6, IL-1ß, TNFα, CXCL10, type I, II and III interferons) following SARS-CoV-2 infection of peripheral blood mononuclear cells in vitro were analyzed as well as antibodies against IFNα and IFNω (by ELISA) in patients and healthy controls. We also performed whole exome sequencing (WES) of patient DNA. A total of five patients (ages 19, 23, 33, 38, 50 years) were included. The patients shared characteristic features, although organ involvement and the time course of disease varied slightly. SARS-CoV-2 in vitro infection of patient PBMCs revealed impaired type I and III interferon responses and reduced CXCL10 expression, whereas production of proinflammatory cytokines were less affected, compared to healthy controls. Presence of interferon autoantibodies was not detected. Whole exome sequencing analysis of patient DNA revealed 12 rare potentially disease-causing variants in genes related to autophagy, classical Kawasaki disease, restriction factors and immune responses. In conclusion, we observed an impaired production of type I and III interferons in response to SARS-CoV-2 infection and detected several rare potentially disease-causing gene variants potentially contributing to MIS-A.


Assuntos
COVID-19/patologia , Citocinas/sangue , Interferon-alfa/biossíntese , Interferons/biossíntese , Síndrome de Resposta Inflamatória Sistêmica/patologia , Adulto , Autoanticorpos/sangue , Quimiocina CXCL10/biossíntese , Comorbidade , Exoma/genética , Feminino , Humanos , Interferon-alfa/imunologia , Interferons/imunologia , Leucócitos Mononucleares/imunologia , Masculino , Pessoa de Meia-Idade , SARS-CoV-2/imunologia , Sequenciamento do Exoma , Adulto Jovem , Interferon lambda
7.
Eur J Immunol ; 51(9): 2317-2329, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34272885

RESUMO

Endothelial cells are key regulators of transendothelial migration and their secretion of chemokines and expression of adhesion molecules facilitates lymphocyte entry into tissues. Previously, we demonstrated that Tregs can reduce transendothelial migration of T cells into tumors by decreasing endothelial CXCL10 secretion, but the mechanism by which this occurs is still not known. In this study, we aimed to define how Tregs decrease transendothelial migration into tumors. mRNA sequencing of intestinal tumor endothelial cells from Treg depleted mice identified neutral sphingomyelinase 2 (nSMase2) as a gene downregulated in the presence of Tregs. nSMase2 is expressed in human umbilical vein endothelial cells (HUVECs) and was decreased after coculture with Tregs. Furthermore, blocking of nSMase2 activity in vitro decreased VCAM1, CX3CL1, and CXCL10 expression in HUVECs, mirroring the same decrease found in Treg cocultures. In the APCmin/+ mouse model of intestinal cancer, nSMase2 is lower in tumor endothelial cells than in unaffected small intestine and chronic treatment with a nSMase2 inhibitor suppressed the increased migration that is otherwise seen in the absence of Tregs. We conclude that nSMase2 is an important mediator in endothelial cells supporting transendothelial migration, which may be targeted by Tregs to reduce T-cell migration into tumors.


Assuntos
Quimiocina CXCL10/metabolismo , Neoplasias do Colo/patologia , Linfócitos do Interstício Tumoral/imunologia , Esfingomielina Fosfodiesterase/metabolismo , Linfócitos T Reguladores/imunologia , Migração Transendotelial e Transepitelial/fisiologia , Animais , Moléculas de Adesão Celular/biossíntese , Linhagem Celular , Quimiocina CX3CL1/biossíntese , Quimiocina CXCL10/biossíntese , Neoplasias do Colo/imunologia , Regulação para Baixo , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Subpopulações de Linfócitos T/imunologia , Versicanas/biossíntese
8.
Cancer Res ; 81(8): 2171-2183, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33558334

RESUMO

Ewing sarcoma is the second most common pediatric bone cancer, with a 5-year survival rate for metastatic disease of only 20%. Recent work indicates that survival is strongly correlated with high levels of tumor-infiltrating lymphocytes (TIL), whose abundance is associated with IFN-inducible chemokines CXCL10 and CCL5. However, the tumor-intrinsic factors that drive chemokine production and TIL recruitment have not been fully elucidated. We previously showed that ubiquitin-specific protease 6 (USP6) directly deubiquitinates and stabilizes Jak1, thereby inducing an IFN signature in Ewing sarcoma cells. Here, we show that this gene set comprises chemokines associated with immunostimulatory, antitumorigenic functions, including CXCL10 and CCL5. USP6 synergistically enhanced chemokine production in response to exogenous IFN by inducing surface upregulation of IFNAR1 and IFNGR1. USP6-expressing Ewing sarcoma cells stimulated migration of primary human monocytes and T lymphocytes and triggered activation of natural killer (NK) cells in vitro. USP6 inhibited Ewing sarcoma xenograft growth in nude but not NSG mice and was accompanied by increased intratumoral chemokine production and infiltration and activation of NK cells, dendritic cells, and macrophages, consistent with a requirement for innate immune cells in mediating the antitumorigenic effects of USP6. High USP6 expression in patients with Ewing sarcoma was associated with chemokine production, immune infiltration, and improved survival. This work reveals a previously unrecognized tumor-suppressive function for USP6, which engenders an immunostimulatory microenvironment through pleiotropic effects on multiple immune lineages. This further raises the possibility that USP6 activity may be harnessed to create a "hot" tumor microenvironment in immunotherapy. SIGNIFICANCE: This study reveals a novel tumor-suppressive function for USP6 by inducing an immunostimulatory microenvironment, suggesting that USP6 activity may be exploited to enhance immunotherapy regimens.


Assuntos
Neoplasias Ósseas/genética , Linfócitos do Interstício Tumoral , Sarcoma de Ewing/genética , Proteínas Supressoras de Tumor/fisiologia , Ubiquitina Tiolesterase/fisiologia , Animais , Neoplasias Ósseas/imunologia , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/mortalidade , Movimento Celular/efeitos dos fármacos , Quimiocina CCL5/biossíntese , Quimiocina CXCL10/biossíntese , Células Dendríticas/efeitos dos fármacos , Humanos , Imunoterapia , Interferons/farmacologia , Janus Quinase 1/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Receptor de Interferon alfa e beta/metabolismo , Receptores de Interferon/metabolismo , Sarcoma de Ewing/imunologia , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/mortalidade , Microambiente Tumoral/imunologia , Ubiquitina Tiolesterase/imunologia , Ubiquitina Tiolesterase/metabolismo , Regulação para Cima/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Receptor de Interferon gama
9.
Inflammation ; 44(1): 206-216, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32772240

RESUMO

Sjögren's syndrome (SS) is a chronic autoimmune disease targeting salivary and lacrimal glands. C-X-C motif chemokine ligand 10 (CXCL10) expression is upregulated in lip salivary glands (LSGs) of primary SS (pSS) patients, and CXCL10 involved in SS pathogenesis via immune-cell accumulation. Moreover, interferon (IFN)-γ enhances CXCL10 production via the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway. We investigated the effects of baricitinib, a selective JAK1/2 inhibitor, on both IFN-γ-induced CXCL10 production and immune-cell chemotaxis. We used immunohistochemical staining to determine the expression levels and localization of JAK1 and JAK2 in LSGs of SS patients (n = 12) and healthy controls (n = 3). We then evaluated the effect of baricitinib in an immortalized normal human salivary gland ductal (NS-SV-DC) cell line. Immunohistochemical analysis of LSGs from pSS patients revealed strong JAK1 and JAK2 expression in ductal and acinar cells, respectively. Baricitinib significantly inhibited IFN-γ-induced CXCL10 expression as well as the protein levels in an immortalized human salivary gland ductal-cell clone in a dose-dependent manner. Additionally, western blot analysis showed that baricitinib suppressed the IFN-γ-induced phosphorylation of STAT1 and STAT3, with a stronger effect observed in the case of STAT1. It also inhibited IFN-γ-mediated chemotaxis of Jurkat T cells. These results suggested that baricitinib suppressed IFN-γ-induced CXCL10 expression and attenuated immune-cell chemotaxis by inhibiting JAK/STAT signaling, suggesting its potential as a therapeutic strategy for pSS.


Assuntos
Azetidinas/farmacologia , Quimiocina CXCL10/antagonistas & inibidores , Interferon gama/farmacologia , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 2/antagonistas & inibidores , Purinas/farmacologia , Pirazóis/farmacologia , Fator de Transcrição STAT1/antagonistas & inibidores , Ductos Salivares/metabolismo , Sulfonamidas/farmacologia , Azetidinas/uso terapêutico , Linhagem Celular Transformada , Quimiocina CXCL10/biossíntese , Feminino , Humanos , Janus Quinase 1/biossíntese , Janus Quinase 2/biossíntese , Células Jurkat , Purinas/uso terapêutico , Pirazóis/uso terapêutico , Fator de Transcrição STAT1/biossíntese , Ductos Salivares/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Síndrome de Sjogren/tratamento farmacológico , Síndrome de Sjogren/metabolismo , Sulfonamidas/uso terapêutico
10.
Cell Biol Int ; 45(1): 238-244, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32926524

RESUMO

Human dental pulp cells (HDPCs) play an important role in pulpitis. Semaphorin3A (Sema3A), which is an axon guidance molecule, is a member of the secretory semaphorin family. Recently, Sema3A has been reported to be an osteoprotective factor and to be involved in the immune response. However, the role of Sema3A in dental pulp inflammation remains unknown. The aim of this study was to reveal the existence of Sema3A in human dental pulp tissue and the effect of Sema3A which is released from tumor necrosis factor (TNF)-α-stimulated HDPCs on production of proinflammatory cytokines, such as interleukin (IL)-6 and CXC chemokine ligand 10 (CXCL10), from HDPCs stimulated with TNF-α. Sema3A was detected in inflamed pulp as compared to normal pulp. HDPCs expressed Neuropilin-1(Nrp1) which is Sema3A receptor. TNF-α increased the levels of IL-6 and CXCL10 in HDPCs in time-dependent manner. Sema3A inhibited production of these two cytokines from TNF-α-stimulated HDPCs. TNF-α induced soluble Sema3A production from HDPCs. Moreover, antibody-based neutralization of Sema3A further promoted production of IL-6 and CXCL10 from TNF-α-stimulated HDPCs. Sema3A inhibited nuclear factor (NF)-κB P65 phosphorylation and inhibitor κBα degradation in TNF-α-stimulated HDPCs. These results indicated that Sema3A is induced in human dental pulp, and TNF-α acts on HDPCs to produce Sema3A, which partially inhibits the increase in IL-6 and CXCL10 production induced by TNF-α, and that the inhibition leads to suppression of NF-κB activation. Therefore, it is suggested that Sema3A may regulate inflammation in dental pulp and be novel antiinflammatory target molecule for pulpitis.


Assuntos
Quimiocina CXCL10/biossíntese , Polpa Dentária/citologia , Interleucina-6/biossíntese , NF-kappa B/metabolismo , Semaforina-3A/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Anti-Inflamatórios/metabolismo , Humanos , Inibidor de NF-kappaB alfa/metabolismo , NF-kappa B/antagonistas & inibidores , Neuropilina-1/metabolismo , Fosforilação , Proteólise
11.
Kidney Blood Press Res ; 46(1): 74-83, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33326977

RESUMO

INTRODUCTION: Various viruses including a novel coronavirus (SARS-CoV-2) can infect the kidney. When viruses invade the glomeruli from the bloodstream, glomerular endothelial cells (GECs) initiate the innate immune reactions. We investigated the expression of interferon (IFN)-induced protein with tetratricopeptide repeats (IFIT) 1/2/3, antiviral molecules, in human GECs treated with a toll-like receptor (TLR) 3 agonist. Role of IFIT1/2/3 in the expression of C-X-C motif chemokine ligand 10 (CXCL10) was also examined. METHODS: Human GECs were cultured and stimulated with polyinosinic-polycytidylic acid (poly IC), a synthetic TLR3 agonist. Real-time qPCR, Western blotting, and ELISA were used to examine the expression of IFIT1/2/3, IFN-ß, and CXCL10. RNA interference against IFN-ß or IFIT1/2/3 was also performed. RESULTS: Expression of IFIT1/2/3 and CXCL10 was induced by poly IC in GECs. The inductions were inhibited by RNA interfering of IFN-ß. Knockdown of IFIT1/2/3 decreased the CXCL10 expression. Knockdown of IFIT3 decreased the expression of IFIT1 and IFIT2 proteins. CONCLUSION: IFIT1/2/3 and CXCL10 were induced by poly IC via IFN-ß in GECs. IFIT1/2/3 may increase the expression of CXCL10 which induces lymphocyte chemotaxis and may inhibit the replication of infected viruses. These molecules may play a role in GEC innate immune reactions in response to viruses.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Reguladoras de Apoptose/biossíntese , Quimiocina CXCL10/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Glomérulos Renais/metabolismo , Proteínas de Ligação a RNA/biossíntese , Receptor 3 Toll-Like/agonistas , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose/genética , Células Cultivadas , Quimiocina CXCL10/genética , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Glomérulos Renais/citologia , Glomérulos Renais/efeitos dos fármacos , Poli I-C/farmacologia , Proteínas de Ligação a RNA/genética , Receptor 3 Toll-Like/metabolismo
12.
Immunity ; 53(1): 187-203.e8, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32640259

RESUMO

Only a small percentage of patients afflicted with gastric cancer (GC) respond to immune checkpoint blockade (ICB). To study the mechanisms underlying this resistance, we examined the immune landscape of GC. A subset of these tumors was characterized by high frequencies of regulatory T (Treg) cells and low numbers of effector T cells. Genomic analyses revealed that these tumors bore mutations in RHOA that are known to drive tumor progression. RHOA mutations in cancer cells activated the PI3K-AKT-mTOR signaling pathway, increasing production of free fatty acids that are more effectively consumed by Treg cells than effector T cells. RHOA mutant tumors were resistant to PD-1 blockade but responded to combination of PD-1 blockade with inhibitors of the PI3K pathway or therapies targeting Treg cells. We propose that the metabolic advantage conferred by RHOA mutations enables Treg cell accumulation within GC tumors, generating an immunosuppressive TME that underlies resistance to ICB.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Inibidores de Checkpoint Imunológico/farmacologia , Neoplasias Gástricas/genética , Linfócitos T Reguladores/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Animais , Contagem de Linfócito CD4 , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Quimiocina CXCL10/biossíntese , Quimiocina CXCL11/biossíntese , Ácidos Graxos não Esterificados/biossíntese , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Receptor de Morte Celular Programada 1/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/imunologia , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/patologia , Linfócitos T Reguladores/imunologia , Serina-Treonina Quinases TOR/metabolismo , Microambiente Tumoral/imunologia
13.
Inflammation ; 43(2): 752-764, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31873836

RESUMO

Cordycepin, a natural derivative of adenosine, has been shown to exert pharmacological properties including anti-oxidation, antitumor, and immune regulation. It is reported that cordycepin is involved in the regulation of macrophage function. However, the effect of cordycepin on inflammatory cell infiltration in inflammation remains ambiguous. In this study, we investigated the potential role of cordycepin playing in macrophage function in CFA-induced inflammation mice model. In this model, we found that cordycepin prevented against macrophage infiltration in paw tissue and reduced interferon-γ (IFN-γ) production in both serum and paw tissue. Using luciferase reporter assay, we found that cordycepin suppressed IFN-γ-induced activators of transcription-1 (STAT1) transcriptional activity in a dose-dependent manner. Moreover, western blotting data demonstrated that cordycepin inhibited IFN-γ-induced STAT1 activation through attenuating STAT1 phosphorylation. Further investigations revealed that cordycepin inhibited the expressions of IFN-γ-inducible protein 10 (IP-10) and monokine induced by IFN-γ (Mig), which were the effector genes in IFN-γ-induced STAT1 signaling. Meanwhile, the excessive inflammatory cell infiltration in paw tissue was reduced by cordycepin. These findings demonstrate that cordycepin alleviates excessive inflammatory cell infiltration through down-regulation of macrophage IP-10 and Mig expressions via suppressing STAT1 phosphorylation. Thus, cordycepin may be a potential therapeutic approach to prevent and treat inflammation-associated diseases.


Assuntos
Quimiocina CXCL10/antagonistas & inibidores , Quimiocina CXCL9/antagonistas & inibidores , Desoxiadenosinas/uso terapêutico , Interferon gama/toxicidade , Macrófagos/efeitos dos fármacos , Fator de Transcrição STAT1/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Quimiocina CXCL10/biossíntese , Quimiocina CXCL9/biossíntese , Desoxiadenosinas/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Adjuvante de Freund/toxicidade , Expressão Gênica , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Macrófagos/metabolismo , Camundongos , Células RAW 264.7 , Distribuição Aleatória , Fator de Transcrição STAT1/metabolismo
14.
Inflammation ; 42(6): 2148-2158, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31440939

RESUMO

Gene expression profiling of lip salivary gland (LSG) has shown that C-X-C motif chemokine 10 (CXCL10) and matrix metalloproteinase 9 (MMP9) expression is upregulated in primary Sjögren's syndrome (pSS) patients. Although CXCL10 and MMP-9 are both associated with pSS pathogenesis, the potential relationship between these two factors has not been investigated. In this study, we used LSG sections from pSS patients and human salivary gland cell lines to investigate the relationship between CXCL10 and MMP-9. Immunofluorescence analyses revealed that CXCL10 and MMP-9 were co-expressed in the LSG of pSS patients, particularly in expanded ductal cells. Furthermore, RT-qPCR analyses on human salivary gland ductal NS-SV-DC cells confirmed that CXCL10 expression was induced by interferon (IFN)-γ, whereas that of MMP9 was stimulated by IFN-α, tumor necrosis factor-α, and interleukin-1ß. Remarkably, MMP-9 inhibition in IFN-γ-stimulated NS-SV-DC cells significantly decreased CXCL10 mRNA and secreted protein levels. Further analyses established that MMP-9 inhibition in IFN-γ-stimulated NS-SV-DC cells decreased STAT1 phosphorylation and hence suppressed IFN-γ signaling. Collectively, these results suggest that in addition to its reported role in the destruction of acinar structures, MMP-9 is involved in the IFN-γ-induced production of CXCL10 in pSS lesions. We believe that our findings open the door to the development of novel treatments for pSS, based on the modulation of MMP-9 activity.


Assuntos
Quimiocina CXCL10/biossíntese , Metaloproteinase 9 da Matriz/metabolismo , Ductos Salivares/citologia , Linhagem Celular , Células Cultivadas , Humanos , Interferon gama/farmacologia , Fosforilação , Fator de Transcrição STAT1/metabolismo , Ductos Salivares/metabolismo , Glândulas Salivares/metabolismo , Síndrome de Sjogren/enzimologia , Síndrome de Sjogren/metabolismo , Síndrome de Sjogren/patologia
15.
Biol Pharm Bull ; 42(1): 57-65, 2019 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-30381616

RESUMO

CXC chemokine ligand 10 (CXCL10) is a CXC chemokine family protein that transmits signals by binding to its specific receptor, CXCR3. CXCL10 is also known as an interferon-γ-inducible chemokine involved in various biological phenomena, including chemotaxis of natural killer (NK) cells and cytotoxic T lymphocytes, that suppress tumor growth and inhibition of angiogenesis. In this study, we examined the effects of forced expression of CXCL10 in a murine colon carcinoma cell line (CT26) on growth and metastasis in syngeneic mice. We first established CT26 cells that were stably expressing murine CXCL10 (CT26/CXCL10) and compared their growth with their parental CT26 cells in vitro and in vivo. The in vitro growth of the CT26/CXCL10 and CT26 cells was comparable, whereas the in vivo growth of the CT26/CXCL10 cells in the skin was strongly suppressed. Liver metastasis of the CT26/CXCL10 cells was also significantly suppressed after intra-splenic implantation. Removal of NK cells by the administration of anti-asialo GM1 antibody canceled the suppression of subcutaneous growth and liver metastasis of CT26/CXCL10 cells. Immunofluorescence clearly showed that abundant NKp46-positive NK cells were recruited into the liver metastatic lesions of the CT26/CXCL10 cells, consistent with specific NK cell migration towards the culture supernatant from the CT26/CXCL10 cells in the chemotaxis assay using transwells. These findings indicate that CXCL10 prevents in vivo growth and metastasis of colon carcinoma cells by recruiting NK cells, suggesting that forced expression of CXCL10 in the colon tumors by gene delivery should lead to a favorable clinical outcome.


Assuntos
Quimiocina CXCL10/biossíntese , Neoplasias do Colo/metabolismo , Regulação Neoplásica da Expressão Gênica , Células Matadoras Naturais/metabolismo , Neoplasias Hepáticas/prevenção & controle , Animais , Linhagem Celular Tumoral , Quimiocina CXCL10/genética , Neoplasias do Colo/patologia , Feminino , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Carga Tumoral/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
16.
J Leukoc Biol ; 104(6): 1187-1198, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30295956

RESUMO

Reactive oxygen species (ROS) are electrophilic chemical species produced from incomplete oxidation. They have long been known as aggressive molecules that lead to direct tissue and cellular damage. Recent studies have reconsidered ROS as second messengers in the initiation and amplification of cell signaling, but how ROS regulate lung tissue and immune cell remain unknown. In this study, we used a LPS-induced acute lung injury (ALI) mouse model to observe disease, progression and determine ROS-related immune responses. We found that ROS play an essential pathogenic role in ALI, however, the major role of ROS in exacerbating ALI was increasing bronchoalveolar fluid (BALF) B cells rather than eliciting tissue damage. Moreover, these pathogenic B cells are FasL+ killer B cells, which reported to damage Fas-sensitive target cells including pulmonary epithelial cells. Furthermore, via in vitro transwell assays and in vivo treatment with neutralizing antibodies. ROS promoted pulmonary epithelial cells to produce CXCL9 and CXCL10, which recruited B cells into BALF. These results demonstrated that during lung injury, instead of causing oxidative damage, ROS mainly serve as second messengers, interacting with tissue and immune cells to enhance immune responses that lead to more severe disease.


Assuntos
Acetilcisteína/uso terapêutico , Lesão Pulmonar Aguda/imunologia , Células Epiteliais Alveolares/efeitos dos fármacos , Subpopulações de Linfócitos B/imunologia , Proteína Ligante Fas/análise , Sequestradores de Radicais Livres/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Sistemas do Segundo Mensageiro/fisiologia , Acetilcisteína/farmacologia , Lesão Pulmonar Aguda/induzido quimicamente , Células Epiteliais Alveolares/metabolismo , Animais , Apoptose , Líquido da Lavagem Broncoalveolar/citologia , Quimiocina CXCL10/biossíntese , Quimiocina CXCL9/biossíntese , Quimiotaxia de Leucócito/efeitos dos fármacos , Citotoxicidade Imunológica , Progressão da Doença , Feminino , Sequestradores de Radicais Livres/farmacologia , Lipopolissacarídeos , Pulmão/imunologia , Pulmão/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Espécies Reativas de Oxigênio/antagonistas & inibidores
17.
Adv Clin Exp Med ; 27(6): 849-856, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29893515

RESUMO

The aim of this review is to present data from the available literature concerning CXCL9, CXCL10 and CXCL11, as well as their receptor 3 (CXCR3) in selected diseases of the central nervous system (CNS), such as tickborne encephalitis (TBE), neuroborreliosis (NB), Alzheimer's disease (AD), and multiple sclerosis (MS). CXCL9, CXCL10, and CXCL11 lack glutamic acid-leucine-arginine (ELR), and are unique, because they are more closely related to each other than to any other chemokine. The aforementioned chemokines are especially involved in Th1-type response and in various diseases, as their expression correlates with the tissue infiltration of T cells. Their production is strongly induced by interferon gamma (IFN-υ), the most typical Th1 cytokine. They act by binding to the CXC3 receptor. Knowledge about the action mechanism of CXCR3 and its ligands may be useful in the treatment of CNS diseases. However, data in the literature concerning the evaluation of CXCL9, CXCL10, CXCL11, and their receptor with the use of the enzyme-linked immunosorbent assay (ELISA) method is limited.


Assuntos
Doenças do Sistema Nervoso Central/imunologia , Quimiocinas/biossíntese , Degeneração Neural/imunologia , Doenças Neurodegenerativas/imunologia , Quimiocina CXCL10/biossíntese , Quimiocina CXCL11/biossíntese , Quimiocina CXCL9/biossíntese , Humanos , Receptores CXCR3/biossíntese
18.
Inflammation ; 41(4): 1172-1181, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29549479

RESUMO

CXCL10, a CXC chemokine induced by interferon-gamma [IFN-γ], has been observed in a wide variety of chronic inflammatory disorders and autoimmune conditions. Although CXCL10 is known to be overexpressed in the salivary glands of individuals with primary Sjögren's syndrome (pSS), it is unclear which cells produce CXCL10 under what types of stimulations. Here, we investigated the precise molecular mechanisms by which CXCL10 was produced in human salivary gland ductal (NS-SV-DC) and acinar (NS-SV-AC) cell lines. Our results demonstrated that NS-SV-DC cells produced higher levels of CXCL10 compared to NS-SV-AC cells. In addition, our findings demonstrated that the regulator of the enhancement of CXCL10 was different between NS-SV-DC and NS-SV-AC cells, i.e., interferon-gamma (IFN-γ) had more potential than interferon-alpha (IFN-α), tumor necrosis factor (TNF)-α, and interleukin (IL)1-ß in the induction of CXCL10 production in NS-SV-DC cells, whereas TNF-α had potential to induce CXCL10 production in NS-SV-AC cells. A Western blot analysis demonstrated that IFN-γ enhanced the production of CXCL10 via both the JAK/STAT1 pathway and the NF-κB pathway in NS-SV-DC cells, whereas TNF-α enhanced the production of CXCL10 via the NF-κB pathway in NS-SV-AC cells. The results of study suggest that the CXCL10 overexpression in the salivary glands is caused mainly by IFN-γ-stimulated salivary gland ductal cells. The enhanced production of CXCL10 by IFN-γ from ductal cells may result in the inflammation of pSS lesions.


Assuntos
Células Acinares/citologia , Quimiocina CXCL10/biossíntese , Glândulas Salivares/citologia , Linhagem Celular , Quimiocina CXCL10/efeitos dos fármacos , Citocinas , Humanos , Interferon gama/farmacologia , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
19.
Front Immunol ; 9: 2, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29403489

RESUMO

Phenolic glycolipids (PGLs) are cell wall components of a subset of pathogenic mycobacteria, with immunomodulatory properties. Here, we show that in addition, PGLs exert antibactericidal activity by limiting the production of nitric oxide synthase (iNOS) in mycobacteria-infected macrophages. PGL-mediated downregulation of iNOS was complement receptor 3-dependent and comparably induced by bacterial and purified PGLs. Using Mycobacterium leprae PGL-1 as a model, we found that PGLs dampen the toll-like receptor (TLR)4 signaling pathway, with macrophage exposure to PGLs leading to significant reduction in TIR-domain-containing adapter-inducing interferon-ß (TRIF) protein level. PGL-driven decrease in TRIF operated posttranscriptionally and independently of Src-family tyrosine kinases, lysosomal and proteasomal degradation. It resulted in the defective production of TRIF-dependent IFN-ß and CXCL10 in TLR4-stimulated macrophages, in addition to iNOS. Our results unravel a mechanism by which PGLs hijack both the bactericidal and inflammatory responses of host macrophages. Moreover, they identify TRIF as a critical node in the crosstalk between CR3 and TLR4.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Antígenos de Bactérias/metabolismo , Glicolipídeos/metabolismo , Macrófagos/imunologia , Mycobacterium leprae/imunologia , Óxido Nítrico Sintase Tipo II/biossíntese , Receptor 4 Toll-Like/metabolismo , Animais , Parede Celular/metabolismo , Células Cultivadas , Quimiocina CXCL10/biossíntese , Interferon beta/biossíntese , Hanseníase/imunologia , Hanseníase/microbiologia , Hanseníase/patologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais
20.
Eur Rev Med Pharmacol Sci ; 21(19): 4423-4430, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29077150

RESUMO

OBJECTIVE: To investigate the therapeutic effect of drug therapy with cyclophosphamide and leflunomide on the joint function damage of patients with systemic lupus erythematosus (SLE) and its regulatory effects on expression levels of programmed death receptor 1, Notch signaling pathway genes and interferon-inducible protein 10 in peripheral blood mononuclear cells. PATIENTS AND METHODS: A total of 60 patients with SLE were randomly divided into two groups. They were treated with cyclophosphamide and leflunomide, respectively. The number of painful joints, joint tenderness index, joint swelling index and erythrocyte sedimentation rate of patients before and after treatment were evaluated, and the peripheral blood was collected from patients in the two groups; the peripheral blood mononuclear cells were extracted. RESULTS: We observed that the number of painful joints, joint tenderness index and joint swelling index in cyclophosphamide group were decreased after treatment (p<0.05), and the erythrocyte sedimentation rate was significantly decreased (p<0.05). The number of painful joints, joint tenderness index and joint swelling index in leflunomide group were decreased after treatment (p<0.05), and the erythrocyte sedimentation rate was significantly decreased (p<0.05). The comparisons of changes in joint functions and erythrocyte sedimentation rates between cyclophosphamide group and leflunomide group after drug therapy showed that the curative effect in leflunomide group was superior to that in cyclophosphamide group (p<0.05). The positive expression rate of peripheral blood mononuclear cell Notch1 in leflunomide group after treatment was significantly decreased, and the curative effect was superior to that in cyclophosphamide group (p<0.05). The comparisons of changes in programmed death receptor 1 of lymphocytes and interferon-inducible protein 10 between cyclophosphamide group and leflunomide group after drug therapy showed that the curative effect in leflunomide group was superior to that in cyclophosphamide group (p<0.05). The comparison of positive expression rate of nuclear factor-κB (NF-κB) in peripheral blood mononuclear cells between the two groups after treatment showed that the curative effect in leflunomide group was superior to that in cyclophosphamide group (p<0.05). There were positive correlations of the expression level of programmed death receptor 1 of peripheral blood lymphocytes in SLE patients with double-stranded DNA (ds-DNA) and SLE disease activity index (p<0.05). There were positive correlations of the expression level of peripheral interferon-inducible protein 10 in SLE patients with ds-DNA and SLE disease activity index (p<0.05). CONCLUSIONS: This study proved that both leflunomide and cyclophosphamide have therapeutic effects on the joint functions and immune dysfunction of peripheral blood mononuclear cells of SLE patients; however, and the effect of leflunomide is better. There are positive correlations of SLE disease activity index with the Notch signaling pathway genes, programmed death receptor 1 and interferon-inducible protein 10 in peripheral blood mononuclear cells, suggesting that these factors are related to the immune dysfunction of peripheral blood mononuclear cells.


Assuntos
Articulações/fisiopatologia , Leucócitos Mononucleares/efeitos dos fármacos , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Lúpus Eritematoso Sistêmico/fisiopatologia , Adulto , Idoso , Quimiocina CXCL10/biossíntese , Quimiocina CXCL10/genética , Ciclofosfamida/efeitos adversos , Ciclofosfamida/uso terapêutico , Feminino , Humanos , Imunossupressores/efeitos adversos , Imunossupressores/uso terapêutico , Isoxazóis/efeitos adversos , Isoxazóis/uso terapêutico , Leflunomida , Contagem de Leucócitos , Masculino , Pessoa de Meia-Idade , Receptor Notch1/biossíntese , Receptor Notch1/genética , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA