Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
Invest Ophthalmol Vis Sci ; 62(14): 14, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34784411

RESUMO

Purpose: The purpose of this study was to explore the C-X-C chemokines CXCL2 and CXCL10 as potential anti-inflammatory targets for Bacillus endophthalmitis. Methods: Bacillus endophthalmitis was induced in C57BL/6J, CXCL2-/-, and CXCL10-/- mice. At specific times postinfection, eyes were analyzed for Bacillus, retinal function, and inflammation. The efficacies of intravitreal anti-CXCL2 and anti-CXCL10 with or without gatifloxacin in B. cereus endophthalmitis were also assessed using the same techniques. Results: Despite similar Bacillus growth in eyes of C57BL/6J, CXCL2-/-, and CXCL10-/- mice, retinal function retention was greater in eyes of CXCL2-/- and CXCL10-/- mice compared to that of C57BL/6J mice. Neutrophil migration into eyes of CXCL2-/- and CXCL10-/- mice was reduced to a greater degree compared to that of eyes of C57BL/6J mice. Infected CXCL2-/- and CXCL10-/- mouse eyes had significantly less inflammation compared to that of C57BL/6J eyes. Retinal structures in infected eyes of CXCL2-/- mice were preserved for a longer time than in CXCL10-/- eyes. Compared to untreated eyes, there was less inflammation and significant retention of retinal function in eyes treated with anti-CXCL2 and anti-CXCL10 with or without gatifloxacin. Conclusions: For Bacillus endophthalmitis, the absence of CXCL2 or CXCL10 in mice resulted in retained retinal function and less inflammation. The absence of CXCL2 led to a better clinical outcome than the absence of CXCL10. The use of anti-CXCL2 and anti-CXCL10 limited inflammation during B. cereus endophthalmitis. These results highlight the utility of CXCL2 and CXCL10 as potential targets for anti-inflammatory therapy that can be tested in conjunction with antibiotics for improving treating Bacillus endophthalmitis.


Assuntos
Bacillus cereus/crescimento & desenvolvimento , Quimiocina CXCL10/fisiologia , Quimiocina CXCL2/fisiologia , Endoftalmite/fisiopatologia , Infecções Oculares Bacterianas/fisiopatologia , Infecções por Bactérias Gram-Positivas/fisiopatologia , Inflamação/fisiopatologia , Animais , Antibacterianos/uso terapêutico , Anticorpos Monoclonais/farmacologia , Bacillus cereus/isolamento & purificação , Quimiocinas CXC/fisiologia , Contagem de Colônia Microbiana , Modelos Animais de Doenças , Eletrorretinografia , Endoftalmite/tratamento farmacológico , Endoftalmite/microbiologia , Infecções Oculares Bacterianas/tratamento farmacológico , Infecções Oculares Bacterianas/microbiologia , Feminino , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Infecções por Bactérias Gram-Positivas/microbiologia , Inflamação/tratamento farmacológico , Inflamação/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/imunologia , Retina/fisiopatologia
2.
Cancer Lett ; 506: 95-106, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-33689775

RESUMO

Interferon regulatory factor 1 (IRF-1) is a tumor suppressor gene in cancer biology with anti-proliferative and pro-apoptotic effect on cancer cells, however mechanisms of IRF-1 regulating tumor microenvironment (TME) in hepatocellular carcinoma (HCC) remain only partially characterized. Here, we investigated that IRF-1 regulates C-X-C motif chemokine 10 (CXCL10) and chemokine receptor 3 (CXCR3) to activate anti-tumor immunity in HCC. We found that IRF-1 mRNA expression was positively correlated with CXCL10 and CXCR3 through qRT-PCR assay in HCC tumors and in analysis of the TCGA database. IRF-1 response elements were identified in the CXCL10 promoter region, and ChIP-qPCR confirmed IRF-1 binding to promote CXCL10 transcription. IRF-2 is a competitive antagonist for IRF-1 mediated transcriptional effects, and overexpression of IRF-2 decreased basal and IFN-γ induced CXCL10 expression. Although IRF-1 upregulated CXCR3 expression in HCC cells, it inhibited proliferation and exerted pro-apoptotic effects, which overcome proliferation partly mediated by activating the CXCL10/CXCR3 autocrine axis. In vitro and in vivo studies showed that IRF-1 increased CD8+ T cells, NK and NKT cells migration, and activated IFN-γ secretion in NK and NKT cells to induce tumor apoptosis through the CXCL10/CXCR3 paracrine axis. Conversely, this effect was markedly abrogated in HCC tumor bearing mice deficient in CXCR3. Therefore, the IRF-1/CXCL10/CXCR3 axis contributes to the anti-tumor microenvironment in HCC.


Assuntos
Carcinoma Hepatocelular/imunologia , Quimiocina CXCL10/fisiologia , Fator Regulador 1 de Interferon/fisiologia , Neoplasias Hepáticas/imunologia , Receptores CXCR3/fisiologia , Animais , Apoptose , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Quimiocina CXCL10/genética , Feminino , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Linfócitos T/imunologia
3.
J Hematol Oncol ; 13(1): 68, 2020 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-32503584

RESUMO

BACKGROUND: CD8+ T cell trafficking to the tumor site is essential for effective colorectal cancer (CRC) immunotherapy. However, the mechanism underlying CD8+ T cell infiltration in colorectal tumor tissues is not fully understood. In the present study, we investigated CD8+ T cell infiltration in CRC tissues and the role of chemokine-chemokine receptor signaling in regulation of T cell recruitment. METHODS: We screened chemokines and cytokines in healthy donor and CRC tissues from early- and advanced-stage patients using multiplex assays and PCR screening. We also utilized transcription factor activation profiling arrays and established a xenograft mouse model. RESULTS: Compared with tumor tissues of early-stage CRC patients, CD8+ T cell density was lower in advanced-stage tumor tissues. PCR screening showed that CXCL10 levels were significantly increased in advanced-stage tumor tissues. CXCR3 (the receptor of CXCL10) expression on CD8+ T cells was lower in the peripheral blood of advanced-stage patients. The migratory ability of CD8+ T cells to CXCL10 depended on CXCR3 expression. Multiplex arrays showed that IL-17A was increased in advanced-stage patient sera, which markedly downregulated CXCR3 expression via activating STAT3 signaling and reduced CD8+ T cell migration. Similar results were found after CD8+ T cells were treated with Th17 cell supernatant. Adding anti-IL-17A or the STAT3 inhibitor, Stattic, rescued these effects in vitro and in vivo. Moreover, survival analysis showed that patients with low CD8 and CXCR3 expression and high IL-17A levels had significantly worse prognosis. CONCLUSIONS: CD8+ T cell infiltration in advanced-stage tumor was systematically inhibited by Th17 cells via IL-17A/STAT3/CXCR3 axis. Our findings indicate that the T cell infiltration in the tumor microenvironment may be improved by inhibiting STAT3 signaling.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Neoplasias Colorretais/imunologia , Interleucina-17/fisiologia , Linfócitos do Interstício Tumoral/imunologia , Proteínas de Neoplasias/fisiologia , Receptores CXCR3/fisiologia , Fator de Transcrição STAT3/fisiologia , Células Th17/fisiologia , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Adulto , Idoso , Animais , Antígenos CD8/biossíntese , Antígenos CD8/genética , Movimento Celular , Quimiocina CXCL10/fisiologia , Neoplasias Colorretais/patologia , Óxidos S-Cíclicos/farmacologia , Regulação para Baixo , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Distribuição Aleatória , Receptores CXCR3/biossíntese , Receptores CXCR3/genética , Fator de Transcrição STAT3/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Mol Cell Endocrinol ; 512: 110866, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32417506

RESUMO

Although 70% of estrogen receptor (ER)-positive breast cancer patients can benefit from tamoxifen therapy, the rapid development of tamoxifen resistance hampers the treatment advantage. In this investigation, we found that the serum level of CXCL10 in breast cancer patients was positively correlated with tumor size and ER status. Furthermore, GSE22220 dataset analysis demonstrated that CXCL10 expression in the tumor was correlated with tumor grade and lymphatic metastasis status, and Kaplan-Meier analysis indicated that patients with high CXCL10 expression had a poor prognosis. Estrogen-deprived MCF7 cells were transfected with CXCL10 luciferase reporter plasmid and treated with 10 nM estrogen. Luciferase reporter assay confirmed that CXCL10 was regulated by estrogen. CXCL10 promoted the proliferation of both parental MCF7 cells and tamoxifen-resistant (TamR) MCF7 cells through the AKT pathway, while CXCL10 inhibition restored the sensitivity of TamR cells to tamoxifen. All of these data indicate that CXCL10 could be utilized as a biomarker to predict the prognosis of breast cancer and as a therapeutic target to treat tamoxifen resistant cases.


Assuntos
Neoplasias da Mama , Quimiocina CXCL10/fisiologia , Resistencia a Medicamentos Antineoplásicos/genética , Tamoxifeno/uso terapêutico , Adulto , Idoso , Antineoplásicos Hormonais/farmacologia , Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Quimiocina CXCL10/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Estrogênios/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Pessoa de Meia-Idade , Estudos Retrospectivos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Tamoxifeno/farmacologia
5.
J Clin Invest ; 129(12): 5518-5536, 2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31710308

RESUMO

microRNA-21 (miR-21) is the most commonly upregulated miRNA in solid tumors. This cancer-associated microRNA (oncomiR) regulates various downstream effectors associated with tumor pathogenesis during all stages of carcinogenesis. In this study, we analyzed the function of miR-21 in noncancer cells of the tumor microenvironment to further evaluate its contribution to tumor progression. We report that the expression of miR-21 in cells of the tumor immune infiltrate, and in particular in macrophages, was responsible for promoting tumor growth. Absence of miR-21 expression in tumor- associated macrophages (TAMs), caused a global rewiring of their transcriptional regulatory network that was skewed toward a proinflammatory angiostatic phenotype. This promoted an antitumoral immune response characterized by a macrophage-mediated improvement of cytotoxic T-cell responses through the induction of cytokines and chemokines, including IL-12 and C-X-C motif chemokine 10. These effects translated to a reduction in tumor neovascularization and an induction of tumor cell death that led to decreased tumor growth. Additionally, using the carrier peptide pH (low) insertion peptide, we were able to target miR-21 in TAMs, which decreased tumor growth even under conditions where miR-21 expression was deficient in cancer cells. Consequently, miR-21 inhibition in TAMs induced an angiostatic and immunostimulatory activation with potential therapeutic implications.


Assuntos
Macrófagos/imunologia , MicroRNAs/genética , Neoplasias/imunologia , Animais , Quimiocina CXCL10/fisiologia , Citotoxicidade Imunológica , Interleucina-12/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/irrigação sanguínea , Microambiente Tumoral
6.
Osteoarthritis Cartilage ; 26(8): 1045-1054, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29782915

RESUMO

OBJECTIVE: This study was conducted to identify cytokine profiles associated with radiographic phenotypes of knee osteoarthritis (rKOA) with a focus on early stage of the disease. METHODS: The pilot population study involved 60 middle-aged patients (mean age 50 ± 7.3y.). Standardized weight-bearing anteroposterior and axial radiographs were used to assess rKOA severity in tibiofemoral (TFJ) of patellofemoral joint (PFJ) by grading system (grades 0-3). Luminex (xMAP®) technology was used to simultaneously assess 60 biomarkers (BMs). RESULTS: Several pathways of angiogenic (CXCL10/IP-10, FGF1/2, PDGF-AA/BB, ANG1, RANTES), tissue remodeling/fibrosis (MMP1/3, TIMP2/3/4, TGFß), and fat tissue (leptin) BMs associated with rKOA severity already in very early phase (grade 1). We identified several sets of cytokines as key markers of early knee osteoarthritis (KOA) predicting radiographic features in logistic-regression models (AUC = 0.80-0.97). Marked sex-specificity of rKOA course was detected: upregulation of angiogenesis dominated in females, whereas the activation of tissue remodeling was dominant in males. Several of these shifts, e.g., decrease of CXCL10/IP-10, took place only in grade 1 KOA and disappeared or reversed in later stages. OA of different knee-joint compartments has distinct profiles of cytokines. A broad list of BMs (TIMP2/3/4, MMP1/3, TGFß1/2, vWF-A2, sE-selectin and leptin) associated with OA in the PFJ. CONCLUSION: Our results demonstrate that substantial and time-limited shifts in the angiogenic and TIMP/MMP systems occur in the early stage of KOA. Our study findings highlight the sex-, grade- and compartment-dependent shifts in above processes. The data may contribute to the individualized prevention of KOA in the future.


Assuntos
Citocinas/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Neovascularização Patológica/patologia , Osteoartrite do Joelho/patologia , Biomarcadores/metabolismo , Quimiocina CXCL10/metabolismo , Quimiocina CXCL10/fisiologia , Citocinas/metabolismo , Progressão da Doença , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fator 1 de Crescimento de Fibroblastos/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 1 da Matriz/fisiologia , Pessoa de Meia-Idade , Neovascularização Patológica/metabolismo , Osteoartrite do Joelho/metabolismo , Projetos Piloto , Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator de Crescimento Derivado de Plaquetas/fisiologia , Fatores Sexuais , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Inibidor Tecidual de Metaloproteinase-2/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/fisiologia
7.
Biosci Biotechnol Biochem ; 82(1): 97-105, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29235416

RESUMO

Accumulating evidence indicates that skeletal muscle secrets proteins referred to as myokines and that exercise contributes to their regulation. In this study, we propose that chemokine (C-X-C motif) ligand 10 (CXCL10) functions as a novel myokine. Initially, we stimulated differentiated C2C12 myotubes with or without electrical pulse stimulation (EPS) to identify novel myokines. Cytokine array analysis revealed that CXCL10 secretion was significantly reduced by EPS, which was further confirmed by enzyme-linked immunosorbent assay and quantitative polymerase chain reaction analysis. Treadmill experiments in mice identified significant reduction of Cxcl10 gene expression in the soleus muscle. Additionally, contraction-dependent p38 MAPK activation appeared to be involved in this reduction. Furthermore, C2C12 conditioned medium obtained after applying EPS could induce survival of MSS31, a vascular endothelial cell model, which was partially attenuated by the addition of recombinant CXCL10. Overall, our findings suggest CXCL10 as a novel exercise-reducible myokine, to control endothelial cell viability.


Assuntos
Quimiocina CXCL10/fisiologia , Teste de Esforço , Fibras Musculares Esqueléticas/fisiologia , Indutores da Angiogênese , Animais , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Sistema de Sinalização das MAP Quinases , Camundongos , Contração Muscular , Condicionamento Físico Animal , Reação em Cadeia da Polimerase
8.
J Invest Dermatol ; 138(5): 1126-1136, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29248547

RESUMO

Primary cutaneous anaplastic T-cell lymphoma, characterized by the CD30+ anaplastic large T cells, comprises the second most common group of cutaneous T-cell lymphoma. Little is known about the mechanisms of disease progression. Here we report that enhancer of zeste homolog 2 (EZH2), the catalytic subunit of polycomb repressive complex 2 that mediates histone H3 lysine 27 trimethylation, is overexpressed in CD30+ anaplastic cells in primary cutaneous anaplastic T-cell lymphoma and large-cell transformed cutaneous T-cell lymphoma. Silencing EZH2 or inhibiting its histone methyltransferase activity conferred increased apoptosis and G1 cell-cycle arrest in primary cutaneous anaplastic T-cell lymphoma cells in vitro and a xenograft model in vivo. This was mediated by the de-repression of thioredoxin-interacting protein, a major redox control molecule, and consequent formation of reactive oxygen species. Silencing thioredoxin-interacting protein abrogated reactive oxygen species accumulation in EZH2 suppressed cells and rescued cell growth disadvantage. Moreover, EZH2 suppression de-repressed C-X-C motif chemokine ligand 10 and facilitated the recruitment of effector CD4+ and CD8+ T cells into the tumor microenvironment via a C-X-C motif chemokine ligand 10/receptor 3 interaction. These results demonstrate a dual role for polycomb repressive complex 2-mediated epigenetic silencing in tumor progression and antitumor immunity in primary cutaneous anaplastic T-cell lymphoma, and provide a rationale for the pharmacologic inhibition of EZH2 activity in large-cell transformed cutaneous T-cell lymphoma.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste/fisiologia , Linfoma Anaplásico de Células Grandes/etiologia , Neoplasias Cutâneas/etiologia , Microambiente Tumoral , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Sobrevivência Celular , Quimiocina CXCL10/genética , Quimiocina CXCL10/fisiologia , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Histona Metiltransferases/metabolismo , Humanos , Linfoma Anaplásico de Células Grandes/tratamento farmacológico , Linfoma Anaplásico de Células Grandes/patologia , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Receptores CXCR3/fisiologia , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia
9.
J Immunol ; 199(12): 4078-4090, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29109123

RESUMO

Acute liver injury can be secondary to a variety of causes, including infections, intoxication, and ischemia. All of these insults induce hepatocyte death and subsequent inflammation, which can make acute liver injury a life-threatening event. IL-22 is a dual natured cytokine which has context-dependent protective and pathogenic properties during tissue damage. Accordingly, IL-22 was shown to promote liver regeneration upon acute liver damage. However, other studies suggest pathogenic properties of IL-22 during chronic liver injury. IL-22 binding protein (IL-22BP, IL-22Ra2) is a soluble inhibitor of IL-22 that regulates IL-22 activity. However, the significance of endogenous IL-22BP in acute liver injury is unknown. We hypothesized that IL-22BP may play a role in acute liver injury. To test this hypothesis, we used Il22bp-deficient mice and murine models of acute liver damage induced by ischemia reperfusion and N-acetyl-p-aminophenol (acetaminophen) administration. We found that Il22bp-deficient mice were more susceptible to acute liver damage in both models. We used Il22 × Il22bp double-deficient mice to show that this effect is indeed due to uncontrolled IL-22 activity. We could demonstrate mechanistically increased expression of Cxcl10 by hepatocytes, and consequently increased infiltration of inflammatory CD11b+Ly6C+ monocytes into the liver in Il22bp-deficient mice upon liver damage. Accordingly, neutralization of CXCL10 reversed the increased disease susceptibility of Il22bp-deficient mice. In conclusion, our data indicate that IL-22BP plays a protective role in acute liver damage, via controlling IL-22-induced Cxcl10 expression.


Assuntos
Acetaminofen/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/fisiopatologia , Fígado/irrigação sanguínea , Receptores de Interleucina/fisiologia , Traumatismo por Reperfusão/fisiopatologia , Animais , Movimento Celular , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Quimiocina CXCL10/antagonistas & inibidores , Quimiocina CXCL10/fisiologia , Constrição , Hepatectomia , Hepatócitos/metabolismo , Interleucinas/deficiência , Interleucinas/metabolismo , Isquemia/fisiopatologia , Fígado/fisiologia , Falência Hepática Aguda/etiologia , Falência Hepática Aguda/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/fisiologia , Receptores de Interleucina/deficiência , Receptores de Interleucina/genética , Regeneração , Traumatismo por Reperfusão/prevenção & controle , Interleucina 22
11.
Cell Biochem Biophys ; 75(1): 139-147, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28111710

RESUMO

Excessive proliferation of vascular smooth muscle cells is one of the main pathological processes leading to atherosclerosis and intimal hyperplasia after vascular interventional therapy. Our previous study has shown that interferon-γ inducible protein-10 contributes to the proliferation of vascular smooth muscle cell. However, the underlying mechanisms remain unclear. Extracellular signal-regulated kinase 1/2, serine/threonine kinase Akt, and cAMP response element binding protein are signaling pathways, which are considered to play important roles in the processes of vascular smooth muscle cell proliferation. Moreover, chemokine receptor 3 and Toll-like receptor 4 are potential receptors of inducible protein-10 in this process. In the present study, IP-10 was found to directly induce vascular smooth muscle cell proliferation, and exposure to inducible protein-10 activated extracellular signal-regulated kinase 1/2, serine/threonine kinase, and cAMP response element binding protein signaling. Inhibitor of extracellular signal-regulated kinase 1/2, rather than inhibitor of serine/threonine kinase, inhibited the phosphorylation of cAMP response element binding protein and reduced inducible protein-10-stimulated vascular smooth muscle cell proliferation. Knockdown of cAMP response element binding protein by siRNA inhibited inducible protein-10-induced vascular smooth muscle cell proliferation. Moreover, anti-CXCR3 IgG, instead of anti-Toll-like receptor 4 IgG, reduced inducible protein-10-induced vascular smooth muscle cell proliferation and inducible protein-10-stimulated extracellular signal-regulated kinase 1/2 and cAMP response element binding protein activation. Together, these results indicate that inducible protein-10 promotes vascular smooth muscle cell proliferation via chemokine receptor 3 and activation of extracellular signal-regulated kinase 1/2 inducible protein-10-induced vascular smooth muscle cell proliferation. These data provide important targets for future studies to modulate atherosclerosis and restenosis after vascular interventional therapy.


Assuntos
Proliferação de Células , Quimiocina CXCL10/fisiologia , Sistema de Sinalização das MAP Quinases , Miócitos de Músculo Liso/fisiologia , Receptores CXCR3/fisiologia , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Músculo Liso Vascular/citologia , Ligação Proteica , Mapeamento de Interação de Proteínas
12.
Mol Neurobiol ; 54(3): 1631-1641, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-26873848

RESUMO

This study was aimed to explore the role of estrogen in inducing inflammatory infiltration of macrophages in polymyositis (PM) through downregulation of miR-21, which could further inhibit the expression of C-X-C motif chemokine 10 (CXCL10). Biopsies were collected from 20 PM patients before and after treatment of glucocorticoid. Additionally, peritoneal macrophages were isolated from male SD model rats (n = 40). Creatine kinase (CK) and CXCL10 and nuclear factor-kappa (NF-κB) expressions were tested using immunosorbent and immunocytochemical assays. We also conducted transwell assay to observe invasive abilities of cells; RT-PCR and western blot were intended to semi-quantify miR-21 and CXCL10 expressions in vitro and in vivo. Compared with the control group, serum creatine kinase (S-CK) was upregulated in PM subjects, but its content decreased after treatment of immunosuppressive substances (e.g., glucocorticoids). Moreover, hormone treatment can significantly increase miR-21 expressions in PM patients (P < 0.05). However, CXCL10 expressions had an opposite tendency compared to miR-21expressions. Results drawn from rat model were consistent with those discovered in PM patients. Moreover, miR-21 transfection could significantly decrease the relative luciferase activity when it was integrated with CXCL10 3'-untranslated region (3'-UTR) in macrophage. Estrogen treatment can also upregulate the expression of NF-κB in macrophage nucleus. Nonetheless, the upregulated tendency was inhibited by either miR-21 mimics or anti-CXCL10 mAb (P < 0.05). Both macrophage migration and CXCL10 expressions were significantly decreased after applying miR-21 treatments compared with the control group, yet estrogen could enhance macrophage migration and increase CXCL10 expressions (P < 0.05). Immune inhibitors such as glucocorticoids can significantly downregulate miR-21 and upregulate CXCL10, ultimately eliciting the inflammatory infiltration of macrophage.


Assuntos
Quimiocina CXCL10/fisiologia , Regulação para Baixo/fisiologia , Estrogênios/farmacologia , Macrófagos/metabolismo , MicroRNAs/biossíntese , Polimiosite/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Feminino , Expressão Gênica , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Masculino , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Pessoa de Meia-Idade , Polimiosite/patologia , Ratos , Ratos Sprague-Dawley
13.
J Invest Dermatol ; 137(2): 350-358, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27686391

RESUMO

Vitiligo is an autoimmune disease of the skin that results in the destruction of melanocytes and the clinical appearance of white spots. Disease pathogenesis depends on IFN-γ and IFN-γ-induced chemokines to promote T-cell recruitment to the epidermis where melanocytes reside. The skin is a complex organ, with a variety of resident cell types. We sought to better define the microenvironment and distinct cellular contributions during autoimmunity in vitiligo, and we found that the epidermis is a chemokine-high niche in both a mouse model and human vitiligo. Analysis of chemokine expression in mouse skin showed that CXCL9 and CXCL10 expression strongly correlate with disease activity, whereas CXCL10 alone correlates with severity, supporting them as potential biomarkers for following disease progression. Further studies in both our mouse model and human patients showed that keratinocytes were the major chemokine producers throughout the course of disease, and functional studies using a conditional signal transducer and activator of transcription (STAT)-1 knockout mouse showed that IFN-γ signaling in keratinocytes was critical for disease progression and proper autoreactive T-cell homing to the epidermis. In contrast, epidermal immune cell populations including endogenous T cells, Langerhans cells, and γδ T cells were not required. These results have important clinical implications, because topical therapies that target IFN-γ signaling in keratinocytes could be safe and effective new treatments, and skin expression of these chemokines could be used to monitor disease activity and treatment responses.


Assuntos
Quimiocinas/fisiologia , Epiderme/imunologia , Linfócitos T/fisiologia , Vitiligo/imunologia , Animais , Biomarcadores/análise , Quimiocina CXCL10/análise , Quimiocina CXCL10/fisiologia , Quimiocina CXCL9/análise , Quimiocina CXCL9/fisiologia , Humanos , Interferon gama/fisiologia , Queratinócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Índice de Gravidade de Doença , Vitiligo/tratamento farmacológico
14.
Oncotarget ; 7(44): 71112-71122, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27764779

RESUMO

Prostate cancer (PCa) has become the second leading cause of male cancer-related mortality in the United States. Mesenchymal stem cells (MSCs) are able to migrate to tumor tissues, and are thus considered to be novel antitumor carriers. However, due to their immunosuppressive nature, the application of MSCs in PCa therapy remains limited. In this study, we investigated the effect of MSCs overexpressing an NAD-dependent deacetylase sirtuin 1 (MSCs-Sirt1) on prostate tumor growth, and we analyzed the underlying mechanisms. Our results show that MSCs accelerate prostate tumor growth, whereas MSCs-Sirt1 significantly suppresses tumor growth. Natural killer (NK) cells and macrophages are the prominent antitumor effectors of the MSCs-Sirt1-induced antitumor activity. IFN-γ and C-X-C motif chemokine ligand 10 (CXCL10) are highly expressed in MSCs-Sirt1 mice. The antitumor effect of MSCs-Sirt1 is weakened when CXCL10 and IFN-γ are inhibited. These results show that MSCs-Sirt1 can effectively inhibit prostate cancer growthrecruiting NK cells and macrophages in a tumor inflammatory microenvironment.


Assuntos
Células Matadoras Naturais/fisiologia , Macrófagos/fisiologia , Células-Tronco Mesenquimais/fisiologia , Neoplasias da Próstata/prevenção & controle , Sirtuína 1/fisiologia , Animais , Movimento Celular , Proliferação de Células , Quimiocina CXCL10/fisiologia , Interferon gama/fisiologia , Ativação de Macrófagos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias da Próstata/patologia , Sirtuína 1/análise
15.
Eur J Oral Sci ; 124(5): 433-439, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27671889

RESUMO

Chemokine (C-X-C motif) ligand (CXCL)9 and CXCL10 are dysregulated in oral inflammatory conditions, and it is not known if these chemokines target microorganisms that form oral biofilm. The aim of this study was to investigate the antimicrobial activity of CXCL9 and CXCL10 on oral microflora and their expression profiles in oral keratinocytes following exposure to inflammatory and infectious stimuli. Streptococcus sanguinis was used as a model and Escherichia coli as a positive control. The antimicrobial effect of CXCL9/CXCL10 was tested using a radial diffusion assay. mRNA transcripts were isolated from lipopolysaccharide (LPS)-treated and untreated (control) oral keratinocyte cell lines at 2-, 4-, 6-, and 8-h time-points of culture. The CXCL9/10 expression profile in the presence or absence of interferon-γ (IFN-γ) was assessed using semiquantitative PCR. Although both chemokines demonstrated antimicrobial activity, CXCL9 was the most effective chemokine against both S. sanguinis and E coli. mRNA for CXCL10 was expressed in control cells and its production was enhanced at all time-points following stimulation with LPS. Conversely, CXCL9 mRNA was not expressed in control or LPS-stimulated cells. Finally, stimulation with IFN-γ enhanced basal expression of both CXCL9 and CXCL10 in oral keratinocytes. Chemokines derived from oral epithelium, particularly CXCL9, demonstrate antimicrobial properties. Bacterial and inflammatory-stimulated up-regulation of CXCL9/10 could represent a key element in oral bacterial colonization homeostasis and host-defense mechanisms.


Assuntos
Quimiocina CXCL10/fisiologia , Quimiocina CXCL9/fisiologia , Escherichia coli , Queratinócitos/fisiologia , Anti-Infecciosos , Humanos , Interferon gama , Boca/microbiologia , Streptococcus sanguis
16.
Cancer Res ; 76(20): 5970-5982, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27569211

RESUMO

The FDA-approved mTOR inhibitor rapamycin mediates important immune effects, but its contributions to the anticancer effects of the drug are unclear. Here we report evidence that rapamycin-mediated cancer protection relies upon stimulation of γδ T cells. In a well-established mouse model of carcinogen and inflammation-driven skin carcinogenesis, IFNγ recruited γδ TCRmid T cells to the epidermis where rapamycin boosted their perforin-dependent antitumor properties. These antitumor cells were mostly Vγ5-Vγ4-Vγ1- in phenotype. IFNγ signals were required in both hematopoietic and nonhematopoietic cells for rapamycin to optimally promote epidermal infiltration of γδ TCRmid T cells, as mediated by CXCR3-CXCL10 interactions, along with the antitumor effects of these cells. In mouse xenograft models of human squamous cell carcinoma, rapamycin improved human γδ T-cell-mediated cancer cell killing. Our results identify immune mechanisms for the cancer prevention and treatment properties of rapamycin, challenging the paradigm that mTOR inhibition acts primarily by direct action on tumor cells. Cancer Res; 76(20); 5970-82. ©2016 AACR.


Assuntos
Adjuvantes Imunológicos/farmacologia , Citotoxicidade Imunológica/efeitos dos fármacos , Receptores de Antígenos de Linfócitos T gama-delta/análise , Sirolimo/farmacologia , Neoplasias Cutâneas/prevenção & controle , Subpopulações de Linfócitos T/efeitos dos fármacos , Serina-Treonina Quinases TOR/antagonistas & inibidores , 9,10-Dimetil-1,2-benzantraceno , Animais , Movimento Celular , Quimiocina CXCL10/fisiologia , Feminino , Humanos , Interferon gama/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores CXCR3/fisiologia , Subpopulações de Linfócitos T/imunologia
17.
Br J Cancer ; 115(5): 553-63, 2016 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-27490802

RESUMO

BACKGROUND: Tumour-infiltrating lymphocytes (TILs) are associated with improved survival in several epithelial cancers. The two chemokines CXCL9 and CXCL10 facilitate chemotactic recruitment of TILs, and their intratumoral accumulation is a conceivable way to improve TIL-dependent immune intervention in cancer. However, the prognostic impact of CXCL9 and CXCL10 in high-grade serous ovarian cancer (HGSC) is largely unknown. METHODS: One hundred and eighty four cases of HGSC were immunohistochemically analyzed for CXCL9, CXCL10. TILs were assessed using CD3, CD56 and FOXP3 staining. Chemokine regulation was investigated using the ovarian cancer cell lines OV-MZ-6 and SKOV-3. RESULTS: High expression of CXCL9 and CXCL10 was associated with an approximately doubled overall survival (n=70, CXCL9: HR 0.41; P=0.006; CXCL10: HR 0.46; P=0.010) which was confirmed in an independent validation set (n=114; CXCL9: HR 0.60; P=0.019; CXCL10: HR 0.52; P=0.005). Expression of CXCR3 ligands significantly correlated with TILs. In human ovarian cancer cell lines the cyclooxygenase (COX) metabolite Prostaglandin E2 was identified as negative regulator of chemokine secretion, whereas COX inhibition by indomethacin significantly upregulated CXCL9 and CXCL10. In contrast, celecoxib, the only COX inhibitor prospectively evaluated for therapy of ovarian cancer, suppressed NF-κB activation and inhibited chemokine release. CONCLUSION: Our results support the notion that CXCL9 and CXCL10 exert tumour-suppressive function by TIL recruitment in human ovarian cancer. COX inhibition by indomethacin, not by celecoxib, may be a promising approach to concomitantly improve immunotherapies.


Assuntos
Quimiocina CXCL10/fisiologia , Quimiocina CXCL9/fisiologia , Inibidores de Ciclo-Oxigenase/farmacologia , Neoplasias Ovarianas/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Indometacina/farmacologia , Ligantes , Neoplasias Ovarianas/patologia , Prognóstico , Análise de Sobrevida
18.
Adv Exp Med Biol ; 904: 23-32, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26900060

RESUMO

This chapter is an overview of published observations from our laboratory on the psychophysics and neurobiology of the persistent itch and pain of allergic contact dermatitis (ACD). ACD is a clinically significant problem with many features characteristic of other pruritic disorders. Our approach was to produce ACD experimentally in humans and in the mouse. The goal was to use the mouse as an animal model for investigating the peripheral neural mechanisms of itch and pain of ACD in humans. Humans and mice were each sensitized by cutaneous topical application of squaric acid dibutyl ester, a hapten not encountered in the environment. Subsequent challenge at another cutaneous site produced local inflammation ("ACD") with humans reporting persistent itch (lasting up to a week) and mice exhibiting persistent itch- and pain-like behaviors directed toward the ACD site. Enhanced mechanically evoked itch and pain in surrounding skin in humans were reversibly blocked by numbing the ACD site with cold, suggesting dependence on ongoing activity from the site. In mice, in vivo recordings revealed spontaneous activity in a subset of pruriceptive, mechanoheat-sensitive nociceptors with unmyelinated axons innervating the ACD site. These and a larger subpopulation of acutely dissociated small-diameter neurons innervating the ACD site exhibited an upregulation of the receptor CXCR3 and excitatory responses to one of its ligands, the chemokine CXCL10 (IP-10) that contributes to the pathogenesis of ACD. Preliminary findings point to possible therapeutic targets that could be investigated in inflammatory itch disorders in humans.


Assuntos
Dermatite Alérgica de Contato/fisiopatologia , Modelos Animais , Nociceptividade/fisiologia , Prurido/fisiopatologia , Animais , Quimiocina CXCL10/fisiologia , Crioterapia , Ciclobutanos/toxicidade , Dermatite Alérgica de Contato/etiologia , Dermatite Alérgica de Contato/psicologia , Dermatite Alérgica de Contato/terapia , Feminino , Temperatura Alta/efeitos adversos , Humanos , Inflamação , Masculino , Camundongos , Fibras Nervosas Amielínicas/fisiologia , Nociceptores/efeitos dos fármacos , Nociceptores/fisiologia , Prurido/induzido quimicamente , Prurido/psicologia , Receptores CXCR3/fisiologia , Especificidade da Espécie
19.
J Transl Med ; 13: 369, 2015 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-26602091

RESUMO

BACKGROUND: Plasmodium falciparum malaria in India is characterized by high rates of severe disease, with multiple organ dysfunction (MOD)-mainly associated with acute renal failure (ARF)-and increased mortality. The objective of this study is to identify cytokine signatures differentiating severe malaria patients with MOD, cerebral malaria (CM), and cerebral malaria with MOD (CM-MOD) in India. We have previously shown that two cytokines clusters differentiated CM from mild malaria in Maharashtra. Hence, we also aimed to determine if these cytokines could discriminate malaria subphenotypes in Odisha. METHODS: P. falciparum malaria patients from the SCB Medical College Cuttack in the Odisha state in India were enrolled along with three sets of controls: healthy individuals, patients with sepsis and encephalitis (n = 222). We determined plasma concentrations of pro- and anti-inflammatory cytokines and chemokines for all individuals using a multiplex assay. We then used an ensemble of statistical analytical methods to ascertain whether particular sets of cytokines/chemokines were predictors of severity or signatures of a disease category. RESULTS: Of the 26 cytokines/chemokines tested, 19 increased significantly during malaria and clearly distinguished malaria patients from controls, as well as sepsis and encephalitis patients. High amounts of IL-17, IP-10, and IL-10 predicted MOD, decreased IL-17 and MIP-1α segregated CM-MOD from MOD, and increased IL-12p40 differentiated CM from CM-MOD. Most severe malaria patients with ARF exhibited high levels of IL-17. CONCLUSION: We report distinct differences in cytokine production correlating with malarial disease severity in Odisha and Maharashtra populations in India. We show that CM, CM-MOD and MOD are clearly distinct malaria-associated pathologies. High amounts of IL-17, IP-10, and IL-10 were predictors of MOD; decreased IL-17 and MIP-1α separated CM-MOD from MOD; and increased IL-12p40 differentiated CM from CM-MOD. Data also suggest that the IL-17 pathway may contribute to malaria pathogenesis via different regulatory mechanisms and may represent an interesting target to mitigate the pathological processes in malaria-associated ARF.


Assuntos
Injúria Renal Aguda/fisiopatologia , Quimiocina CXCL10/fisiologia , Interleucina-10/fisiologia , Interleucina-17/fisiologia , Malária Falciparum/fisiopatologia , Insuficiência de Múltiplos Órgãos/fisiopatologia , Injúria Renal Aguda/patologia , Quimiocina CXCL10/sangue , Humanos , Interleucina-10/sangue , Interleucina-17/sangue , Malária Falciparum/patologia , Insuficiência de Múltiplos Órgãos/patologia
20.
PLoS One ; 10(8): e0136089, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26291724

RESUMO

Transplantation of glioblastoma patient biopsy spheroids to the brain of T cell-compromised Rowett (nude) rats has been established as a representative animal model for human GBMs, with a tumor take rate close to 100%. In immunocompetent littermates however, primary human GBM tissue is invariably rejected. Here we show that after repeated passaging cycles in nude rats, human GBM spheroids are enabled to grow in the brain of immunocompetent rats. In case of engraftment, xenografts in immunocompetent rats grow progressively and host leukocytes fail to enter the tumor bed, similar to what is seen in nude animals. In contrast, rejection is associated with massive infiltration of the tumor bed by leukocytes, predominantly ED1+ microglia/macrophages, CD4+ T helper cells and CD8+ effector cells, and correlates with elevated serum levels of pro-inflammatory cytokines IL-1α, IL-18 and TNF-α [corrected]. We observed that in nude rat brains, an adaptation to the host occurs after several in vivo passaging cycles, characterized by striking attenuation of microglial infiltration. Furthermore, tumor-derived chemokines that promote leukocyte migration and their entry into the CNS such as CXCL-10 and CXCL-12 are down-regulated, and the levels of TGF-ß2 increase. We propose that through serial in vivo passaging in nude rats, human GBM cells learn to avoid and or/ suppress host immunity. Such adapted GBM cells are in turn able to engraft in immunocompetent rats without signs of an inflammatory response.


Assuntos
Neoplasias Encefálicas/imunologia , Glioblastoma/imunologia , Rejeição de Enxerto/prevenção & controle , Tolerância Imunológica/imunologia , Transplante de Neoplasias/métodos , Animais , Quimiocina CXCL10/fisiologia , Quimiocina CXCL12/fisiologia , Modelos Animais de Doenças , Feminino , Rejeição de Enxerto/imunologia , Humanos , Imunocompetência/imunologia , Interleucina-18/sangue , Interleucina-1beta/sangue , Masculino , Reação em Cadeia da Polimerase , Ratos , Ratos Nus , Esferoides Celulares , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA