Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Ethnopharmacol ; 275: 114175, 2021 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-33933571

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Circulating tumor cells (CTCs) play an important role in tumor metastasis and may be a target for metastasis prevention. The traditional Chinese medicine Jinfukang functions to improve immunity, prevent metastasis, and prolong lung cancer patient survival periods. Yet, whether Jinfukang prevents metastasis by regulating immune cells to clearance CTCs is still unknown. AIM OF THE STUDY: To explore the anti-metastasis mechanism of Jinfukang from the perspective of regulating NK cells to clear CTCs. MATERIALS AND METHODS: CTC-TJH-01 cell was treated with Jinfukang. Cytokine chip was used to detect cytokines in cell culture supernatant. Lymphocyte recruitment assay was detected by Transwell and flow cytometry. Protein expression was analysis by Western blot. LDH kit was used to detect cytotoxicity. NOD-SCID mice used for tail vein injection to study lung metastasis. RESULTS: Jinfukang could promote the expression and secretion of the chemokine CX3CL1 by CTCs. In addition, Jinfukang could promote the recruitment of natural killer (NK) cells by CTCs and significantly increase the cytotoxic effect of NK cells on CTCs. Moreover, Jinfukang could upregulate the expression of FasL and promote the secretion of TNF-α by NK cells and that NK cells could induce the apoptosis of CTCs through the Fas/FasL signaling pathway. Finally, we confirmed that Jinfukang could promote NK cells to kill CTCs and then inhibit lung cancer metastasis in vivo. The above effects of Jinfukang could be partially reversed by an anti-CX3CL1 mAb. CONCLUSIONS: These results suggest that Jinfukang may prevent lung cancer metastasis by enhancing the clearance of CTCs in the peripheral blood by NK cells, providing evidence for the anti-metastasis effect of Jinfukang.


Assuntos
Antineoplásicos/farmacologia , Quimiocina CX3CL1/genética , Medicamentos de Ervas Chinesas/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Metástase Neoplásica/prevenção & controle , Células Neoplásicas Circulantes/efeitos dos fármacos , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Quimiocina CX3CL1/antagonistas & inibidores , Quimiocina CX3CL1/metabolismo , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/uso terapêutico , Proteínas Ligadas por GPI/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células Matadoras Naturais/imunologia , Neoplasias Pulmonares/complicações , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Metástase Neoplásica/imunologia , Células Neoplásicas Circulantes/imunologia , Células Neoplásicas Circulantes/patologia , Receptores de Morte Celular/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima/efeitos dos fármacos , Receptor fas/metabolismo
2.
Cell Death Dis ; 12(2): 135, 2021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33542207

RESUMO

Despite the progress to understand inflammatory reactions, mechanisms causing their resolution remain poorly understood. Prostanoids, especially prostaglandin E2 (PGE2), are well-characterized mediators of inflammation. PGE2 is produced in an inducible manner in macrophages (Mϕ) by microsomal PGE2-synthase-1 (mPGES-1), with the notion that it also conveys pro-resolving properties. We aimed to characterize the role of mPGES-1 during resolution of acute, zymosan-induced peritonitis. Experimentally, we applied the mPGES-1 inhibitor compound III (CIII) once the inflammatory response was established and confirmed its potent PGE2-blocking efficacy. mPGES-1 inhibition resulted in an incomplete removal of neutrophils and a concomitant increase in monocytes and Mϕ during the resolution process. The mRNA-seq analysis identified enhanced C-X3-C motif receptor 1 (CX3CR1) expression in resident and infiltrating Mϕ upon mPGES-1 inhibition. Besides elevated Cx3cr1 expression, its ligand CX3CL1 was enriched in the peritoneal lavage of the mice, produced by epithelial cells upon mPGES-1 inhibition. CX3CL1 not only increased adhesion and survival of Mϕ but its neutralization also completely reversed elevated inflammatory cell numbers, thereby normalizing the cellular, peritoneal composition during resolution. Our data suggest that mPGES-1-derived PGE2 contributes to the resolution of inflammation by preventing CX3CL1-mediated retention of activated myeloid cells at sites of injury.


Assuntos
Quimiocina CX3CL1/metabolismo , Dinoprostona/metabolismo , Inibidores Enzimáticos/farmacologia , Células Epiteliais/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Peritonite/enzimologia , Prostaglandina-E Sintases/antagonistas & inibidores , Animais , Anticorpos Neutralizantes/farmacologia , Receptor 1 de Quimiocina CX3C/genética , Receptor 1 de Quimiocina CX3C/metabolismo , Adesão Celular , Sobrevivência Celular , Células Cultivadas , Quimiocina CX3CL1/antagonistas & inibidores , Quimiocina CX3CL1/genética , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Feminino , Macrófagos Peritoneais/enzimologia , Macrófagos Peritoneais/imunologia , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos , Peritonite/genética , Peritonite/imunologia , Fenótipo , Prostaglandina-E Sintases/metabolismo , Regulação para Cima
3.
Int J Oncol ; 57(1): 249-263, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32319605

RESUMO

Spinal metastasis occurs in 50­75% of bone metastases caused by hepatocellular carcinoma (HCC), and HCC­derived spinal metastasis can lead to a less favorable prognosis. Recently, several studies have demonstrated that C­X3­C motif chemokine ligand 1 (CX3CL1) is closely associated with cancer metastasis, and its secretion is modulated by a disintegrin and metalloproteinase 17 (ADAM17). Bone marrow endothelial cells (BMECs) are an essential component of bone marrow. However, little is known about the roles in and effects of BMECs on HCC spinal metastasis. The present study demonstrated that CX3CL1 and C­X­C motif chemokine receptor 3 (CXCR3) expression was upregulated in HCC spinal metastases, and that CX3CL1 promoted the migration and invasion of HCC cells to the spine. Western blot analysis revealed that the Src/protein tyrosine kinase 2 (PTK2) axis participated in CX3CL1­induced HCC cell invasion and migration. CX3CL1 also increased the expression of M2 macrophage markers in THP­1 monocytes. BMECs promoted the migration and invasion of Hep3B and MHCC97H cells by secreting soluble CX3CL1, whereas the neutralization of CX3CL1 inhibited this enhancement. CX3CL1 enhanced the activation of the phosphatidylinositol­4,5­bisphosphate 3­kinase catalytic subunit alpha (PIK3CA)/AKT serine/threonine kinase 1 (AKT1) and Ras homolog family member A (RHOA)/Rho associated coiled­coil containing protein kinase 2 (ROCK2) signaling pathways through the Src/PTK2 signaling pathway. Furthermore, ADAM17 was activated by mitogen­activated protein kinase (MAPK)z14 in BMECs and significantly promoted the secretion of CX3CL1. HCC cells enhanced the recruitment and proliferation of BMECs. The overexpression of CX3CR1 facilitated the spinal metastasis of HCC in a mouse model in vivo. In addition, in vivo experiments revealed that BMECs promoted the growth of HCC in the spine. The present study demonstrated that CX3CL1 participates in HCC spinal metastasis, and that BMECs play an important role in the regulation of CX3CL1 in the spinal metastatic environment.


Assuntos
Proteína ADAM17/metabolismo , Carcinoma Hepatocelular/secundário , Quimiocina CX3CL1/metabolismo , Células Progenitoras Endoteliais/metabolismo , Neoplasias Hepáticas/patologia , Neoplasias da Coluna Vertebral/secundário , Proteína ADAM17/genética , Adulto , Idoso , Animais , Medula Óssea/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Quimiocina CX3CL1/antagonistas & inibidores , Quinase 1 de Adesão Focal/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Imidazóis , Masculino , Camundongos , Pessoa de Meia-Idade , Proteína Quinase 14 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Piridinas , RNA Interferente Pequeno/metabolismo , Receptores CXCR3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Coluna Vertebral/patologia , Microambiente Tumoral
4.
Arthritis Rheumatol ; 71(11): 1923-1934, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31173491

RESUMO

OBJECTIVE: To assess the preclinical efficacy and mechanism of action of an anti-CX3 CL1 monoclonal antibody (mAb) in systemic sclerosis (SSc). METHODS: Cultured human dermal fibroblasts were used to evaluate the direct effect of anti-CX3 CL1 mAb on fibroblasts. In addition, bleomycin-induced and growth factor-induced models of SSc were used to investigate the effect of anti-CX3 CL1 mAb on leukocyte infiltration, collagen deposition, and vascular damage in the skin. RESULTS: Anti-CX3 CL1 mAb treatment significantly inhibited Smad3 phosphorylation (P < 0.05) and expression of type I collagen and fibronectin 1 (P < 0.01) in dermal fibroblasts stimulated with transforming growth factor ß1 (TGFß1). In the bleomycin model, daily subcutaneous bleomycin injection increased serum CX3 CL1 levels (P < 0.05) and augmented lesional CX3 CL1 expression. Simultaneous administration of anti-CX3 CL1 mAb or CX3 CR1 deficiency significantly suppressed the dermal thickness, collagen content, and capillary loss caused by bleomycin (P < 0.05). Injection of bleomycin induced expression of pSmad3 and TGFß1 in the skin, which was inhibited by anti-CX3 CL1 mAb. Further, the dermal infiltration of CX3 CR1+ cells, macrophages (inflammatory and alternatively activated [M2-like] subsets), and CD3+ cells significantly decreased following anti-CX3 CL1 mAb therapy (P < 0.05), as did the enhanced skin expression of fibrogenic molecules, such as thymic stromal lymphopoietin and secreted phosphoprotein 1 (P < 0.05). However, the treatment did not significantly reduce established skin fibrosis. In the second model, simultaneous anti-mCX3 CL1 mAb therapy significantly diminished the skin fibrosis induced by serial subcutaneous injection of TGFß and connective tissue growth factor (P < 0.01). CONCLUSION: Anti-CX3 CL1 mAb therapy may be a novel approach for treating early skin fibrosis in inflammation-driven fibrotic skin disorders such as SSc.


Assuntos
Anticorpos Monoclonais/farmacologia , Receptor 1 de Quimiocina CX3C/imunologia , Capilares/efeitos dos fármacos , Quimiocina CX3CL1/antagonistas & inibidores , Colágeno/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Escleroderma Sistêmico/imunologia , Pele/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/toxicidade , Bleomicina/toxicidade , Capilares/patologia , Quimiocina CX3CL1/imunologia , Colágeno/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Fibroblastos/patologia , Fibrose/induzido quimicamente , Humanos , Técnicas In Vitro , Inflamação , Camundongos , Escleroderma Sistêmico/patologia , Transdução de Sinais , Pele/imunologia , Pele/patologia , Proteína Smad3/efeitos dos fármacos , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta3/toxicidade
5.
Front Immunol ; 10: 784, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31134047

RESUMO

Fractalkine (FKN), also known as chemokine (C-X3-C motif) ligand 1, constitutes an intriguing chemokine with a documented role in the development of numerous inflammatory diseases including autoimmune disease. Specifically, it has been reported that FKN is involved in the disease progression of lupus nephritis (LN). The epithelial-mesenchymal transition (EMT) plays a significant role in the formation of tubulointerstitial lesions (TIL), which are increasingly recognized as a hallmark of tissue fibrogenesis after injury. However, the correlation between FKN and EMT or TIL in LN has not been determined. To investigate the potential role of FKN in EMT and TIL, MRL lymphoproliferation (MRL/lpr) strain mice were treated with an anti-FKN antibody, recombinant-FKN chemokine domain, or isotype antibody. Our results revealed that treatment with the anti-FKN antibody improved EMT, TIL, and renal function in MRL/lpr mice, along with inhibiting activation of the Wnt/ß-catenin signaling pathway. In contrast, administration of the recombinant-FKN chemokine domain had the opposite effect. Furthermore, to further explore the roles of FKN in EMT, we assessed the levels of EMT markers in FKN-depleted or overexpressing human proximal tubule epithelial HK-2 cells. Our results provide the first evidence that the E-cadherin level was upregulated, whereas α-SMA and vimentin expression was downregulated in FKN-depleted HK-2 cells. In contrast, overexpression of FKN in HK-2 cells enhanced EMT. In addition, inhibition of the Wnt/ß-catenin pathway by XAV939 negated the effect of FKN overexpression, whereas activation of the Wnt/ß-catenin pathway by Ang II impaired the effect of the FKN knockout on EMT in HK-2 cells. Together, our data indicate that FKN plays essential roles in the EMT progression and development of TIL in MRL/lpr mice, most likely through activation of the Wnt/ß-catenin signaling pathway.


Assuntos
Quimiocina CX3CL1/imunologia , Transição Epitelial-Mesenquimal/imunologia , Túbulos Renais Proximais/imunologia , Nefrite Lúpica/imunologia , Via de Sinalização Wnt/imunologia , Animais , Anticorpos/farmacologia , Linhagem Celular , Quimiocina CX3CL1/antagonistas & inibidores , Quimiocina CX3CL1/genética , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Feminino , Técnicas de Silenciamento de Genes , Túbulos Renais Proximais/patologia , Nefrite Lúpica/genética , Nefrite Lúpica/patologia , Camundongos , Domínios Proteicos , Proteínas Recombinantes/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/genética
6.
Lasers Med Sci ; 34(9): 1841-1847, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30969378

RESUMO

Temporomandibular disorder (TMD) is a collective term that encompasses a set of clinical problems that affect the masticatory muscles, the temporomandibular joint, and associated structures. Despite their high clinical prevalence, the mechanisms of chronic craniofacial muscle pain are not yet well understood. Treatments for TMD pain relief and control should be minimally invasive, reversible, and conservative. Photobiomodulation (PBM) is a promising option once it is known to inhibit inflammatory response and to relief painful symptoms. Herein, the effects of PBM (660 nm, 30 mW, 16 J/cm2, 0.2 cm2, 15 s in a continuous frequency) on the pain sensitivity of rats submitted to an experimental model of TMD induced by CFA was evaluated. Experimental TMD was induced in rats by the injection of complete Freund's adjuvant (CFA) injection into the masseter muscle. Nociceptive behavior was evaluated by electronic von Frey before CFA and after 1 h, 3 h, 6 h, and 24 h and 7, 14, and 21 days after PBM treatment. Inflammatory infiltrate was evaluated by histology of the masseter muscle and fractalkine expression was evaluated by immunohistochemistry of the trigeminal ganglia. PBM reversed the mechanical hypersensitivity of the animals by inhibiting the local inflammatory response, observed by the decrease of the inflammatory infiltrate in the masseter muscle of rats and by a central inhibition of fractalkine observed in the trigeminal ganglion. These data provide new insights into the mechanisms involved in the effects of photobiomodulation therapy emphasizing its therapeutic potential in the treatment of TMD.


Assuntos
Analgesia , Quimiocina CX3CL1/antagonistas & inibidores , Terapia com Luz de Baixa Intensidade , Transtornos da Articulação Temporomandibular/radioterapia , Animais , Quimiocina CX3CL1/metabolismo , Adjuvante de Freund , Masculino , Músculo Masseter/fisiopatologia , Microglia/patologia , Dor/patologia , Manejo da Dor , Ratos Sprague-Dawley , Regeneração , Gânglio Trigeminal/patologia
7.
J Clin Pharmacol ; 59(5): 688-701, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30575978

RESUMO

E6011 is a novel humanized antifractalkine (FKN) monoclonal antibody being developed as a therapeutic target for Crohn's disease, rheumatoid arthritis, and primary biliary cholangitis. This study was a randomized, double-blind, placebo-controlled single-ascending-dose study of intravenous administration of E6011 (0.0006-10 mg/kg) in healthy Japanese adult men (n = 64). The starting dose was the minimum anticipated biological effect level (MABEL). MABEL was estimated by extrapolating results of a pharmacokinetic/pharmacodynamic (PK/PD) model relating E6011 exposure and suppression of free soluble FKN using data obtained from cynomolgus monkeys. Safety assessments consisted of monitoring and recording adverse events, laboratory tests, vital signs, intensive electrocardiograms, and chest x-rays. Blood samples to determine PK, PD (serum total FKN concentration), and serum anti-E6011 antibody were collected. Noncompartmental analysis was used to derive PK parameters. Single intravenous infusions of E6011 were safe and well tolerated in healthy subjects. Serum E6011 concentrations showed triphasic elimination. An increase in serum total FKN concentration was observed, confirming target engagement. The dose strategy for patient studies is to select regimens that will attain a minimum serum E6011 exposure of 10 µg/mL, identified as the minimum concentration needed to saturate the target-mediated elimination pathway. Model-based drug development from preclinical stage was successful in identifying dose regimens for clinical testing.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/farmacocinética , Administração Intravenosa , Adulto , Animais , Anticorpos Monoclonais Humanizados/efeitos adversos , Artrite Reumatoide/tratamento farmacológico , Quimiocina CX3CL1/antagonistas & inibidores , Quimiocina CX3CL1/imunologia , Doença de Crohn/tratamento farmacológico , Relação Dose-Resposta a Droga , Método Duplo-Cego , Humanos , Cirrose Hepática Biliar/tratamento farmacológico , Macaca fascicularis , Masculino , Modelos Biológicos , Placebos , Adulto Jovem
8.
Clin Liver Dis ; 22(3): 501-515, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30259849

RESUMO

Primary biliary cholangitis is an archetypal autoimmune disease that causes cholestasis, fibrosis, and liver failure. Ursodeoxycholic acid and obeticholic acid are approved for its treatment. Not all patients respond, some are intolerant, many have ongoing symptoms, and new therapies are required. Herein we describe drugs in development and potential future biological targets. We consider compounds acting on the farnesoid X receptor/fibroblast growth factor 19 pathway, fibrates and other agonists of the peroxisome proliferator-activated receptor family, transmembrane-G-protein-receptor-5 agonists, and several immunological agents. We also consider the roles of bile acid reuptake inhibitors, nalfurafine, and fibrates in pruritus management.


Assuntos
Ácido Quenodesoxicólico/análogos & derivados , Ácidos Cólicos/uso terapêutico , Fatores de Crescimento de Fibroblastos/análogos & derivados , Imunossupressores/uso terapêutico , Cirrose Hepática Biliar/tratamento farmacológico , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Receptores Citoplasmáticos e Nucleares/agonistas , Abatacepte/uso terapêutico , Acetatos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Antirreumáticos/uso terapêutico , Bezafibrato/uso terapêutico , Antígenos CD40/antagonistas & inibidores , Ligante de CD40/antagonistas & inibidores , Chalconas/uso terapêutico , Quimiocina CX3CL1/antagonistas & inibidores , Ácido Quenodesoxicólico/uso terapêutico , Desenvolvimento de Medicamentos , Fenofibrato/uso terapêutico , Humanos , Hipolipemiantes/uso terapêutico , Inibidores de Janus Quinases/uso terapêutico , Cirrose Hepática Biliar/complicações , Metilaminas/uso terapêutico , PPAR alfa/agonistas , PPAR delta/agonistas , PPAR gama/agonistas , Propionatos/uso terapêutico , Prurido/tratamento farmacológico , Prurido/etiologia , Receptores Acoplados a Proteínas G/agonistas , Tiazepinas/uso terapêutico , Triazóis/uso terapêutico , Ustekinumab/uso terapêutico
9.
Biochem Biophys Res Commun ; 493(4): 1510-1517, 2017 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-28986258

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is a fatal malignancy with an estimated 5 year survival rate of approximately 5% of all stages combined. High potential of PDAC metastasis is a leading cause for high mortality and poor prognosis. The majority of patients present with distant metastasis at diagnosis. Fractalkine (FKN) is recognized as a chemokine and a specific ligand of CX3CR1. It has been reported that FKN/CX3CR1 system was upregulated in many types of solid tumors. However, role of FKN/CX3CR1 in PDAC development remains unclear. In the current investigation, we found that FKN and CX3CR1 expression was significantly increased in PDAC tissues, especially in the metastatic samples, and was highly-correlated with severity of PDAC. Ectopic expression of FKN promoted the proliferation and migration of PDAC, while knockdown of CX3CR1 reversed the function of FKN. In addition, PDAC cells with FKN-deficiency showed impaired proliferation and migration activity. The underlying mechanism is that FKN/CX3CR1 activated JAK/STAT signaling, which in turn regulated cell growth. Consistently, in vivo tumorigenesis assay validated the regulatory role of FKN/CX3CR1 in PDAC growth. Our investigation helped understanding the pathogenesis of PDAC occurrence, and demonstrated critical role of FKN/CX3CR1 in PDAC development.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Quimiocina CX3CL1/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores de Quimiocinas/metabolismo , Animais , Receptor 1 de Quimiocina CX3C , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/secundário , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Quimiocina CX3CL1/antagonistas & inibidores , Quimiocina CX3CL1/genética , Técnicas de Silenciamento de Genes , Humanos , Janus Quinases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Pancreáticas/etiologia , Neoplasias Pancreáticas/genética , Interferência de RNA , Receptores de Quimiocinas/antagonistas & inibidores , Receptores de Quimiocinas/genética , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais
10.
Cell Biochem Funct ; 35(6): 315-326, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28845524

RESUMO

Fractalkine (FKN, CX3CL1) is highly expressed in a majority of malignant solid tumours. Fractalkine is the only known ligand for CX3CR1. In this study, we performed an analysis to determine the effects of fractalkine/CX3CR1 on modulating apoptosis and explored the related mechanisms. The expression of fractalkine/CX3CR1 was detected by immunohistochemistry and western blotting. The levels of AKT/p-AKT, BCL-xl, and BCL-2 were detected by western blotting. Then, the effects of exogenous and endogenous fractalkine on the regulation of tumour apoptosis and proliferation were investigated. The mechanism of fractalkine/CX3CR1 on modulating apoptosis in cancer cells through the activation of AKT/NF-κB/p65 signals was evaluated. The effect of fractalkine on regulating cell cycle distribution was also tested. Fractalkine, AKT/p-AKT, and apoptotic regulatory proteins BCL-xl and BCL-2 were highly expressed in human pancreatic cancer tissues. In vitro, fractalkine/CX3CR1 promoted proliferation and mediated resistance to apoptosis in pancreatic cancer cells. The antiapoptotic effect of fractalkine was induced by the activation of AKT/NF-κB/p65 signalling in pancreatic cancer cells. The NF-κB/p65 contributes to promote the expressions of BCL-xl and BCL-2 and reduce caspase activity, thereby inhibiting apoptotic processes. Treatment with fractalkine resulted in the enrichment of pancreatic cancer cells in S phase with a concomitant decrease in the number of cells in G1 phase. The present study demonstrated the function of fractalkine in the activation of the AKT/NF-κB/p65 signalling cascade and mediation of apoptosis resistance in pancreatic cancer cells. Fractalkine/CX3CR1 could serve as a diagnostic marker and as a potential target for chemotherapy in early stage pancreatic cancer. Pancreatic cancer is characterized by local recurrence, neural invasion, or distant metastasis. The present study demonstrated the overexpression of fractalkine/CX3CR1 in pancreatic cancer tissues, indicating its important role in the tumourigenesis of pancreatic cancer, and suggested that the overexpression of fractalkine/CX3CR1 could serve as a diagnostic marker for pancreatic cancer. Moreover, we reveal the mechanism that fractalkine functions on the activation of the AKT/NF-κB/p65 signalling cascade and regulation of the antiapoptosis process in pancreatic cancer cells. Fractalkine/CX3CR1 could serve as an effective therapeutic target of chemotherapeutic and biologic agents in early stage pancreatic cancer.


Assuntos
Apoptose , Proliferação de Células , Quimiocina CX3CL1/metabolismo , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição RelA/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quimiocina CX3CL1/antagonistas & inibidores , Quimiocina CX3CL1/genética , Quimiocina CX3CL1/farmacologia , Humanos , Imuno-Histoquímica , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteína bcl-X/metabolismo
11.
Mol Med Rep ; 13(4): 3627-38, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26955849

RESUMO

Protectin D1 (PD1) is a bioactive product generated from docosahexaenoic acid, which may exert anti-inflammatory effects in various inflammatory diseases. However, the underlying molecular mechanism of its anti­inflammatory activity on concanavalin A (Con A)-induced hepatitis remains unknown. The aim of the present study was to investigate the protective effects of PD1 against Con A­induced liver injury and the underlying mechanisms via intravenous injection of PD1 prior to Con A administration. C57BL/6 mice were randomly divided into four experimental groups as follows: Control group, Con A group (30 mg/kg), 20 µg/kg PD1 + Con A (30 mg/kg) group and 10 µg/kg PD1 + Con A (30 mg/kg) group. PD1 pretreatment was demonstrated to significantly inhibit elevated plasma aminotransferase levels, high mobility group box 1 and liver necrosis, which were observed in Con A­induced hepatitis. Furthermore, compared with the Con A group, PD1 pretreatment prevented the production of pro­inflammatory cytokines, including tumor necrosis factor­α, interferon­Î³ and interleukin­2, ­1ß and ­6. In addition, pretreatment with PD1 markedly downregulated cluster of differentiation (CD)4+, CD8+ and natural killer T (NKT) cell infiltration in the liver. PD1 pretreatment was observed to suppress the messenger RNA and protein expression levels of NLR family, pyrin domain containing 3 and Toll­like receptor (TLR) 4 in liver tissue samples. Further data indicated that PD1 pretreatment inhibited the activation of the nuclear factor κ­light­chain­enhancer of activated B cells (NF­κB) signaling pathway and chemokine (C­X3­C motif) ligand 1 (CX3CL1)/chemokine (C-X3-C motif) receptor 1 (CX3CR1) axis by preventing phosphorylation of nuclear factor of κ light polypeptide gene enhancer in B-cells inhibitor, α and NF­κB in Con A­induced liver injury. Therefore, these results suggest that PD1 administration protects mice against Con A­induced liver injury via inhibition of various inflammatory cytokines and, in part, by suppressing CD4+, CD8+ and NKT cell infiltration in the liver and the NF­κB­activated CX3CL1/CX3CR1 signaling pathway. The beneficial effect of PD1 may be associated with the inhibition of TLR4 expression and the downregulation of NF­κB activation. In conclusion, PD1 appears to be a potential natural bioproduct, and provide a promising strategy, for the prevention of hepatic injury in patients with chronic or acute liver disease.


Assuntos
Quimiocina CX3CL1/metabolismo , Ácidos Docosa-Hexaenoicos/farmacologia , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Receptores de Quimiocinas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Receptor 1 de Quimiocina CX3C , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Quimiocina CX3CL1/antagonistas & inibidores , Concanavalina A/toxicidade , Citocinas/análise , Ácidos Docosa-Hexaenoicos/uso terapêutico , Inflamassomos/metabolismo , Fígado/citologia , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Receptores de Quimiocinas/antagonistas & inibidores , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Transaminases/sangue
12.
Exp Physiol ; 100(7): 805-17, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25943588

RESUMO

NEW FINDINGS: What is the central question of this study? What is the cardioprotective role of fractalkine neutralization in heart failure and what are the mechanisms responsible? What is the main finding and its importance? The concentration of fractalkine is increased in the left ventricle of mice with myocardial infarction, similar to the increases in plasma from heart failure patients. The present study shows a clear beneficial effect of neutralizing fractalkine in a model of myocardial infarction, which results in increased survival. Such an approach may be worthwhile in human patients. Concentrations of the chemokine fractalkine (FKN) are increased in patients with chronic heart failure, and our previous studies show that aged mice lacking the prostaglandin E2 EP4 receptor subtype (EP4-KO) have increased cardiac FKN, with a phenotype of dilated cardiomyopathy. However, how FKN participates in the pathogenesis of heart failure has rarely been studied. We hypothesized that FKN contributes to the pathogenesis of heart failure and that anti-FKN treatment prevents heart failure induced by myocardial infarction (MI) more effectively in EP4-KO mice. Male EP4-KO mice and wild-type littermates underwent sham or MI surgery and were treated with an anti-FKN antibody or control IgG. At 2 weeks post-MI, echocardiography was performed and hearts were excised for determination of infarct size, immunohistochemistry and Western blot of signalling molecules. Given that FKN protein levels in the left ventricle were increased to a similar extent in both strains after MI and that anti-FKN treatment improved survival and cardiac function in both strains, we subsequently used only wild-type mice to examine the mechanisms whereby anti-FKN is cardioprotective. Myocyte cross-sectional area and interstitial collagen fraction were reduced after anti-FKN treatment, as were macrophage migration and gelatinase activity. Activation of ERK1/2 and p38 MAPK were reduced after neutralization of FKN. In vitro, FKN increased fibroblast proliferation. In conclusion, increased FKN contributes to heart failure after MI. This effect is not exacerbated in EP4-KO mice, suggesting that there is no link between FKN and lack of EP4. Overall, inhibition of FKN may be important to preserve cardiac function post-MI.


Assuntos
Quimiocina CX3CL1/antagonistas & inibidores , Infarto do Miocárdio/tratamento farmacológico , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Quimiocina CX3CL1/imunologia , Modelos Animais de Doenças , Insuficiência Cardíaca/fisiopatologia , Camundongos Transgênicos , Infarto do Miocárdio/fisiopatologia , Remodelação Ventricular/efeitos dos fármacos
13.
Stem Cell Res Ther ; 5(2): 40, 2014 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-24646750

RESUMO

INTRODUCTION: Studies have demonstrated that mesenchymal stromal cells (MSCs) could reverse acute and chronic kidney injury by a paracrine or endocrine mechanism, and microvesicles (MVs) have been regarded as a crucial means of intercellular communication. In the current study, we focused on the therapeutic effects of human Wharton-Jelly MSCs derived microvesicles (hWJMSC-MVs) in renal ischemia/reperfusion injury and its potential mechanisms. METHODS: MVs isolated from conditioned medium were injected intravenously in rats immediately after ischemia of the left kidney for 60 minutes. The animals were sacrificed at 24 hours, 48 hours and 2 weeks after reperfusion. The infiltration of inflammatory cells was identified by the immunostaining of CD68+ cells. ELISA was employed to determine the inflammatory factors in the kidney and serum von Willebrand Factor (VWF). Tubular cell proliferation and apoptosis were identified by immunostaining. Renal fibrosis was assessed by Masson's tri-chrome straining and alpha-smooth muscle actin (α-SMA) staining. The CX3CL1 expression in the kidney was measured by immunostaining and Western blot, respectively. In vitro, human umbilical vein endothelial cells were treated with or without MVs for 24 or 48 hours under hypoxia injury to test the CX3CL1 by immunostaining and Western blot. RESULTS: After administration of hWJMSC-MVs in acute kidney injury (AKI) rats, renal cell apoptosis was mitigated and proliferation was enhanced, inflammation was also alleviated in the first 48 hours. MVs also could suppress the expression of CX3CL1 and decrease the number of CD68+ macrophages in the kidney. In the late period, improvement of renal function and abrogation of renal fibrosis were observed. In vitro, MVs could down-regulate the expression of CX3CL1 in human umbilical vein endothelial cells under hypoxia injury at 24 or 48 hours. CONCLUSIONS: A single administration of MVs immediately after ischemic AKI could ameliorate renal injury in both the acute and chronic stage, and the anti-inflammatory property of MVs through suppression of CX3CL1 may be a potential mechanism. This establishes a substantial foundation for future research and treatment.


Assuntos
Injúria Renal Aguda/terapia , Quimiocina CX3CL1/antagonistas & inibidores , Rim/irrigação sanguínea , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Traumatismo por Reperfusão/prevenção & controle , Geleia de Wharton/citologia , Injúria Renal Aguda/metabolismo , Animais , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Geleia de Wharton/metabolismo
14.
Biochim Biophys Acta ; 1813(12): 2026-35, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21888931

RESUMO

OBJECTIVE: In atherosclerotic lesions, fractalkine (CX3CL1) and its receptor (CX3CR1) expressed by smooth muscle cells (SMC) and monocytes/macrophages, mediate the heterotypic anchorage and chemotaxis of these cells. We questioned whether, during the close interaction of monocytes with SMC, the CX3CL1/CX3CR1 pair modulates the expression of pro-atherogenic molecules in these cells. METHODS AND RESULTS: SMC were co-cultured with monocytes or LPS-activated monocytes (18h) and then the cells were separated and individually investigated for the gene and protein expression of TNFα, IL-1ß, IL-6, CX3CR1 and metalloproteinases (MMP-2, MMP-9). We found that SMC-monocyte interaction induced, in each cell type, an increased mRNA and protein expression of TNFα, IL-1ß, IL-6, CX3CR1, MMP-2 and MMP-9. Blocking the binding of fractalkine to CX3CR1 (by pre-incubation of monocytes with anti-CX3CR1 or by CX3CR1 siRNA transfection) before cell co-culture decreased the production of TNFα, CX3CR1 and MMP-9. Monocyte-SMC interaction induced the phosphorylation of p38MAPK and activation of AP-1 transcription factor. Silencing the p65 (NF-kB subunit) inhibited the IL-1ß and IL-6 and silencing c-jun inhibited the TNFα, CX3CR1 and MMP-9 induced by SMC-monocyte interaction. CONCLUSIONS: The cross-talk between SMC and monocytes augments the inflammatory response in both cell types as revealed by the increased expression of TNFα, IL-1ß, IL-6, CX3CR1 and MMPs. Up-regulation of TNFα, CX3CR1 and MMP-9 is further increased upon interaction of SMC with activated monocytes and is dependent on fractalkine/CXRCR1 pair. These data imply that the fractalkine/CX3RCR1 axis may represent a therapeutic target to impede the inflammatory process associated with atherosclerosis.


Assuntos
Aorta/metabolismo , Quimiocina CX3CL1/metabolismo , Monócitos/metabolismo , Músculo Liso Vascular/metabolismo , Receptores de Quimiocinas/metabolismo , Aorta/citologia , Aorta/efeitos dos fármacos , Western Blotting , Receptor 1 de Quimiocina CX3C , Células Cultivadas , Quimiocina CX3CL1/antagonistas & inibidores , Quimiocina CX3CL1/genética , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Lipopolissacarídeos/farmacologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Monócitos/citologia , Monócitos/efeitos dos fármacos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , NF-kappa B/genética , NF-kappa B/metabolismo , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Quimiocinas/antagonistas & inibidores , Receptores de Quimiocinas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
15.
Expert Opin Ther Targets ; 14(2): 207-19, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20055718

RESUMO

IMPORTANCE OF THE FIELD: Fractalkine, also known as CX3CL1, is the unique member of the fourth class of chemokines and mediates both chemotaxis and adhesion of inflammatory cells via its highly selective receptor CX3CR1. Fractalkine mediates inflammatory responses and pain sensation and is involved in the pathogenesis and progression of numerous inflammatory disorders and malignancies. AREAS COVERED IN THIS REVIEW: We performed a Medline/PubMed search to detect all published studies that explored the role of fractalkine and CX3CR1 and the possibilities of therapeutic intervention in the fractalkine/CX3CR1 axis in a wide range of clinical disorders, using CX3CR1 blocking antibodies, different fractalkine antagonists, CX3CR1 depletion or transfection of fractalkine expression vectors. WHAT THE READER WILL GAIN: This review summarizes the role of fractalkine and its receptor CX3CR1 in various diseases, focusing on their high potential as novel therapeutic targets, with special emphasis on pancreatic diseases. TAKE HOME MESSAGE: The reviewed studies provide promising results demonstrating fractalkine and CX3CR1 as potential target molecules for future therapeutics that may attenuate pain, inflammation and furthermore serve as an anti-cancer therapy. However, to date, no therapeutics targeting fractalkine or CX3CR1 are in clinical use.


Assuntos
Quimiocina CX3CL1/fisiologia , Animais , Quimiocina CX3CL1/antagonistas & inibidores , Humanos , Inflamação/etiologia , Neoplasias/tratamento farmacológico , Neoplasias/etiologia , Dor/tratamento farmacológico , Dor/etiologia , Pancreatopatias/tratamento farmacológico , Pancreatopatias/etiologia
16.
Atherosclerosis ; 209(1): 89-95, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19781706

RESUMO

OBJECTIVE: A major site of action for the atheroprotective drug nicotinic acid (NA) is adipose tissue, via the G-protein-coupled receptor, GPR109A. Since, adipose tissue is an active secretory organ that contributes both positively and negatively to systemic inflammatory processes associated with cardiovascular disease, we hypothesized that NA would act directly upon adipocytes to alter the expression of pro-inflammatory chemokines, and the anti-inflammatory adipokine adiponectin. METHODS AND RESULTS: TNF-alpha treatment (1.0ng/mL) of 3T3-L1 adipocytes resulted in an increase in gene expression of fractalkine (9+/-3.3-fold, P<0.01); monocyte chemoattractant protein-1 (MCP-1) (24+/-1.2-fold, P<0.001), 'regulated upon activation, normal T cell expressed and secreted' (RANTES) (500+/-55-fold, P<0.001) and inducible nitric oxide synthase (iNOS) (200+/-70-fold, P<0.05). The addition of NA (10(-4)M) to TNF-alpha-treated adipocytes attenuated expression of fractalkine (50+/-12%, P<0.01); MCP-1 (50+/-6%, P<0.01), RANTES (70+/-3%, P<0.01) and iNOS (60+/-16%). This pattern was mirrored in protein released from the adipocytes into the surrounding media. The effect on gene expression was neutralised by pre-treatment with pertussis toxin. NA attenuated macrophage chemotaxis (by 27+/-3.5%, P<0.001) towards adipocyte conditioned media. By contrast, NA, (10(-6)-10(-3)M) increased, in a dose-dependent manner, mRNA of the atheroprotective hormone adiponectin (3-5-fold n=6, P<0.01). CONCLUSIONS: NA suppresses pro-atherogenic chemokines and upregulates the atheroprotective adiponectin through a G-protein-coupled pathway. Since adipose tissue has the potential to contribute to both systemic and local (perivascular) inflammation associated with atherosclerosis our results suggest a new "pleiotropic" role for NA.


Assuntos
Adipócitos/efeitos dos fármacos , Anti-Inflamatórios não Esteroides/farmacologia , Aterosclerose/prevenção & controle , Niacina/farmacologia , Células 3T3 , Adipócitos/metabolismo , Adiponectina/antagonistas & inibidores , Adiponectina/metabolismo , Animais , Aterosclerose/metabolismo , Quimiocina CCL2/antagonistas & inibidores , Quimiocina CCL2/metabolismo , Quimiocina CCL5/antagonistas & inibidores , Quimiocina CCL5/metabolismo , Quimiocina CX3CL1/antagonistas & inibidores , Quimiocina CX3CL1/metabolismo , Meios de Cultivo Condicionados/metabolismo , Camundongos , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima
17.
Z Rheumatol ; 67(5): 424-8, 2008 Sep.
Artigo em Alemão | MEDLINE | ID: mdl-18633630

RESUMO

Fractalkine (CX3CL1), so far the only member of the CX3C class of chemokines, and its receptor, CX3CR1, are strongly expressed in the chronically inflamed synovial tissue of patients with rheumatoid arthritis (RA). Due to the specific binding of Fractalkine to its receptor, many proinflammatory reactions involved in the pathogenesis of RA are triggered. Functionally, fractalkine plays an important proinflammatory role in RA pathogenesis as characterized by induction of synovial angiogenesis, chemotaxis, activation of monocytes and T cells as well as the stimulation of proliferation and synthesis of matrix degrading enzymes (matrix metalloproteinases, MMP) in synovial fibroblasts. Fractalkine thus may represent a novel target molecule for therapeutic intervention in RA.


Assuntos
Artrite Reumatoide/imunologia , Quimiocina CX3CL1/sangue , Antirreumáticos/uso terapêutico , Artrite Reumatoide/tratamento farmacológico , Receptor 1 de Quimiocina CX3C , Quimiocina CX3CL1/antagonistas & inibidores , Fibroblastos/imunologia , Humanos , Macrófagos/imunologia , Receptores de Quimiocinas/sangue , Membrana Sinovial/imunologia , Linfócitos T/imunologia
18.
FASEB J ; 22(6): 1807-16, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18230685

RESUMO

Glucocorticoids are an important anti-inflammatory treatment of many inflammatory diseases including asthma. However, the mechanisms by which they mediate their suppressive effects are not fully understood. Respiratory epithelial cells are a source of CX(3)CL1 (fractalkine), which mediates cell adhesion and acts as a chemoattractant for monocytes, T cells, and mast cells. We show, in lung A549 epithelial cells, that the tumor necrosis factor-alpha (TNF-alpha) and IFNgamma synergistically induced protein release and mRNA expression of CX(3)CL1 is inhibited by dexamethasone, without interfering with cytokine-induced nuclear translocation of NF-kappaB, and by an inhibitor of IkappaB kinase 2, AS602868. DNA binding assays confirmed the ability of NF-kappaB to bind to the proximal CX(3)CL1 promoter. Chromatin immunoprecipitation assays showed a 5-fold increase in the recruitment of NF-kappaB to the CX(3)CL1 gene promoter in response to IFNgamma/TNF-alpha; this too was reversed by dexamethasone. In contrast, dexamethasone did not displace NF-kappaB from the granulocyte-macrophage colony-stimulating factor gene promoter. We conclude that CX(3)CL1 expression is regulated through the NF-kappaB pathway and that dexamethasone inhibits CX(3)CL1 expression through a glucocorticoid receptor-dependent (RU486 sensitive) mechanism. This study also provides support for the action of glucocorticoids mediating their suppressive effects on expression by interfering with the binding of transcriptional activators at native gene promoters.


Assuntos
Quimiocina CX3CL1/genética , Células Epiteliais/citologia , Glucocorticoides/farmacologia , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Linhagem Celular , Quimiocina CX3CL1/antagonistas & inibidores , Dexametasona/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Receptores de Glucocorticoides/metabolismo , Mucosa Respiratória
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA