Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Arthritis Res Ther ; 26(1): 94, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38702742

RESUMO

BACKGROUND: Systemic sclerosis (SSc) is an autoimmune disease characterized by vascular injury and inflammation, followed by excessive fibrosis of the skin and other internal organs, including the lungs. CX3CL1 (fractalkine), a chemokine expressed on endothelial cells, supports the migration of macrophages and T cells that express its specific receptor CX3CR1 into targeted tissues. We previously reported that anti-CX3CL1 monoclonal antibody (mAb) treatment significantly inhibited transforming growth factor (TGF)-ß1-induced expression of type I collagen and fibronectin 1 in human dermal fibroblasts. Additionally, anti-mouse CX3CL1 mAb efficiently suppressed skin inflammation and fibrosis in bleomycin- and growth factor-induced SSc mouse models. However, further studies using different mouse models of the complex immunopathology of SSc are required before the initiation of a clinical trial of CX3CL1 inhibitors for human SSc. METHODS: To assess the preclinical utility and functional mechanism of anti-CX3CL1 mAb therapy in skin and lung fibrosis, a sclerodermatous chronic graft-versus-host disease (Scl-cGVHD) mouse model was analyzed with immunohistochemical staining for characteristic infiltrating cells and RNA sequencing assays. RESULTS: On day 42 after bone marrow transplantation, Scl-cGVHD mice showed increased serum CX3CL1 level. Intraperitoneal administration of anti-CX3CL1 mAb inhibited the development of fibrosis in the skin and lungs of Scl-cGVHD model, and did not result in any apparent adverse events. The therapeutic effects were correlated with the number of tissue-infiltrating inflammatory cells and α-smooth muscle actin (α-SMA)-positive myofibroblasts. RNA sequencing analysis of the fibrotic skin demonstrated that cGVHD-dependent induction of gene sets associated with macrophage-related inflammation and fibrosis was significantly downregulated by mAb treatment. In the process of fibrosis, mAb treatment reduced cGVHD-induced infiltration of macrophages and T cells in the skin and lungs, especially those expressing CX3CR1. CONCLUSIONS: Together with our previous findings in other SSc mouse models, the current results indicated that anti-CX3CL1 mAb therapy could be a rational therapeutic approach for fibrotic disorders, such as human SSc and Scl-cGVHD.


Assuntos
Anticorpos Monoclonais , Quimiocina CX3CL1 , Modelos Animais de Doenças , Doença Enxerto-Hospedeiro , Fibrose Pulmonar , Escleroderma Sistêmico , Pele , Animais , Doença Enxerto-Hospedeiro/tratamento farmacológico , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/patologia , Escleroderma Sistêmico/tratamento farmacológico , Escleroderma Sistêmico/patologia , Escleroderma Sistêmico/imunologia , Camundongos , Quimiocina CX3CL1/metabolismo , Quimiocina CX3CL1/antagonistas & inibidores , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Fibrose Pulmonar/imunologia , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/patologia , Fibrose Pulmonar/prevenção & controle , Pele/patologia , Pele/efeitos dos fármacos , Pele/metabolismo , Pele/imunologia , Fibrose , Feminino , Camundongos Endogâmicos C57BL , Humanos , Pulmão/patologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/imunologia
2.
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
3.
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
4.
Mod Rheumatol ; 31(4): 783-789, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33427546

RESUMO

OBJECTIVES: To evaluate the safety and efficacy of E6011, a novel humanized anti-fractalkine monoclonal antibody, in patients with active rheumatoid arthritis (RA) with an inadequate response to biological disease-modifying antirheumatic drugs (DMARDs). METHODS: Active RA patients inadequately responding to biological DMARDs were randomly assigned to placebo or E6011 400-mg group at a 1:1 ratio, and administered E6011 at weeks 0, 1, 2, and subsequently every 2 weeks. Primary endpoint was American College of Rheumatology (ACR)20 response at week 12. RESULTS: Of 64, 33 received placebo, 31 received E6011 400-mg. The ACR20 response rate at week 12 (non-responder imputation) was 27.3% and 22.6% in the placebo and E6011 groups, respectively. ACR50, ACR70 response rates at week 12 were 3.0%, 0% in the placebo and 9.7%, 3.2% in the E6011 group. Exploratory PK exposure analysis revealed that the effect of E6011 tended to be clearer in patients with higher serum trough E6011 concentration. E6011 was well tolerated with no notable safety concerns. CONCLUSIONS: E6011 400-mg was well tolerated but had no clear efficacy at week 12 in RA patients with inadequate response to biologics. Further investigations are warranted to determine the optimal clinical dose and evaluation period for E6011.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Antirreumáticos/uso terapêutico , Artrite Reumatoide/tratamento farmacológico , Quimiocina CX3CL1/antagonistas & inibidores , Adulto , Anticorpos Monoclonais Humanizados/efeitos adversos , Método Duplo-Cego , Feminino , Humanos , Testes Imunológicos , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento
5.
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
6.
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
7.
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
8.
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
9.
Int Immunol ; 31(5): 287-302, 2019 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-30668715

RESUMO

Chemokine systems modulate inflammatory and immune responses in inflammatory bowel disease (IBD). The colons of IBD patients show increased levels of fractalkine (FKN) and high numbers of FKN receptor-positive (CX3CR1+) cells; however, the FKN-CX3CR1 axis's role in intestinal inflammation, especially in intravascular leukocyte behaviors, still remains unclear. Here, we show that interruption of the FKN-CX3CR1 axis by anti-FKN monoclonal antibody (mAb) ameliorates murine colitis through regulation of intravascular monocyte behaviors in murine colitis models. FKN expression was detectable in vascular endothelium and CX3CR1+ macrophages accumulated in the mucosal lamina propria and submucosa of the inflamed colons. CD115+ monocytes tethered to the venous endothelium and expressed pro-inflammatory mediators. The anti-FKN mAb improved colitis symptoms, markedly reduced pro-inflammatory factors in the colon, maintained blood vessel integrity and reduced tethered monocytes in the inflamed veins. Intravital imaging revealed that CD115+Gr-1low/- monocytes crawled on the apical surfaces of venous endothelium, and anti-FKN mAb rapidly dislodged the crawling monocytes and inhibited their patrolling behavior. These findings suggest that the FKN-CX3CR1 axis triggers the patrolling behavior of crawling monocytes on the venous endothelium of inflamed colons, and accelerates the subsequent leukocyte activation and infiltration by locally producing inflammatory cytokines and chemokines. The mAb also ameliorated symptoms in another IBD model, T-cell-transferred colitis. Blocking the FKN-CX3CR1 axis with an anti-FKN mAb considerably inhibits the colitis-triggered inflammatory cascades, which may be an alternative strategy to treat IBD.


Assuntos
Anticorpos Monoclonais/farmacologia , Receptor 1 de Quimiocina CX3C/antagonistas & inibidores , Quimiocina CX3CL1/antagonistas & inibidores , Colite/tratamento farmacológico , Monócitos/efeitos dos fármacos , Administração Retal , Animais , Anticorpos Monoclonais/imunologia , Receptor 1 de Quimiocina CX3C/imunologia , Quimiocina CX3CL1/imunologia , Colite/induzido quimicamente , Colite/imunologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/imunologia , Oxazóis
10.
Arthritis Rheumatol ; 71(2): 222-231, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30079992

RESUMO

OBJECTIVE: To elucidate the role of the fractalkine (FKN)/CX3 CR1 pathway in joint destruction in rheumatoid arthritis. METHODS: We examined the effect of treatment with anti-mouse FKN (anti-mFKN) monoclonal antibody (mAb) on joint destruction and the migration of osteoclast precursors (OCPs) into the joint, using the collagen-induced arthritis (CIA) model. DBA/1 mice were immunized with bovine type II collagen to induce arthritis, and then treated with anti-mFKN mAb. Disease severity was monitored by arthritis score, and joint destruction was evaluated by soft x-ray and histologic analyses. Plasma levels of joint destruction markers were assessed by enzyme-linked immunosorbent assay. FKN expression on endothelial cells was detected by immunohistochemistry. Bone marrow-derived OCPs were labeled with fluorescein and transferred to mice with CIA, and the migration of the OCPs to the joints was then analyzed. RESULTS: Both prophylactic and therapeutic treatment with anti-mFKN mAb significantly decreased the arthritis and soft x-ray scores. Plasma levels of cartilage oligomeric matrix protein and matrix metalloproteinase 3 decreased after treatment with anti-mFKN mAb. Histologic analysis revealed that anti-mFKN mAb inhibited synovitis, pannus formation, and cartilage destruction, as well as suppressed bone damage, with a marked reduction in the number of tartrate-resistant acid phosphatase-positive osteoclasts. Anti-mFKN mAb strongly inhibited the migration of bone marrow-derived OCPs into the affected synovium. CONCLUSION: Anti-mFKN mAb notably ameliorates arthritis and joint destruction in the CIA model, as well as inhibits migration of OCPs into the synovium. These results suggest that inhibition of the FKN/CX3 CR1 pathway could be a novel strategy for treatment of both synovitis and joint destruction in rheumatoid arthritis.


Assuntos
Anticorpos Monoclonais/farmacologia , Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Receptor 1 de Quimiocina CX3C/imunologia , Movimento Celular/efeitos dos fármacos , Quimiocina CX3CL1/antagonistas & inibidores , Osteoclastos/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Animais , Proteína de Matriz Oligomérica de Cartilagem/efeitos dos fármacos , Proteína de Matriz Oligomérica de Cartilagem/metabolismo , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/imunologia , Cartilagem Articular/patologia , Quimiocina CX3CL1/imunologia , Metaloproteinase 3 da Matriz/efeitos dos fármacos , Metaloproteinase 3 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos DBA , Osteoclastos/metabolismo , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/imunologia , Membrana Sinovial/patologia , Sinovite/patologia , Fosfatase Ácida Resistente a Tartarato/metabolismo
11.
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
12.
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
13.
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
14.
Mol Med Rep ; 16(5): 7548-7552, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28944873

RESUMO

Infections by pathogens may lead to cardiovascular diseases, including acute/chronic myocarditis. (Coxsackieviruses B3) CVB3 is considered to be the most common causative agent in m­yocarditis, which can lead to dilated cardiomyopathy. The present study aimed to investigate the mechanism of CVB3­infected myocardial microvascular endothelial cells. The CVB3 infection was detected by 50% tissue culture infective dose (TCID50). The role of fractalkine (FKN) in the infection was detected using western blotting and RNA interference. To assess mitogen­activated protein kinase signaling activity, levels of total and phosphorylated extracellular signal­regulated kinase (ERK)1/2, c­Jun N­terminal kinase, and p38 were measured at 0, 20, 40, and 60 min after CVB3 infection by western blot analysis. The results showed that infection activated FKN via the ERK1/2 signaling pathway. Furthermore, the TCID50 of CVB3 in infected cells was lower compared with that in myocardial microvascular endothelial cells following ERK1/2 inhibition and FKN silencing. CVB3 infection of myocardial microvascular endothelial cells activates FKN via the ERK1/2 signaling pathway. These findings represent a foundation for the development of novel methods of treating CVB3­induced myocarditis.


Assuntos
Quimiocina CX3CL1/metabolismo , Enterovirus Humano B/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Butadienos/farmacologia , Células Cultivadas , Quimiocina CX3CL1/antagonistas & inibidores , Quimiocina CX3CL1/genética , Regulação para Baixo/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Microvasos/citologia , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Miocárdio/citologia , Nitrilas/farmacologia , Fosforilação/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
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
16.
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
17.
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
18.
J Exp Med ; 211(6): 1185-96, 2014 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-24821910

RESUMO

Atopic dermatitis (AD) is a chronic allergic dermatosis characterized by epidermal thickening and dermal inflammatory infiltrates with a dominant Th2 profile during the acute phase, whereas a Th1 profile is characteristic of the chronic stage. Among chemokines and chemokine receptors associated with inflammation, increased levels of CX3CL1 (fractalkine) and its unique receptor, CX3CR1, have been observed in human AD. We have thus investigated their role and mechanism of action in experimental models of AD and psoriasis. AD pathology and immune responses, but not psoriasis, were profoundly decreased in CX3CR1-deficient mice and upon blocking CX3CL1-CX3CR1 interactions in wild-type mice. CX3CR1 deficiency affected neither antigen presentation nor T cell proliferation in vivo upon skin sensitization, but CX3CR1 expression by both Th2 and Th1 cells was required to induce AD. Surprisingly, unlike in allergic asthma, where CX3CL1 and CX3CR1 regulate the pathology by controlling effector CD4(+) T cell survival within inflamed tissues, adoptive transfer experiments established CX3CR1 as a key regulator of CD4(+) T cell retention in inflamed skin, indicating a new function for this chemokine receptor. Therefore, although CX3CR1 and CX3CL1 act through distinct mechanisms in different pathologies, our results further indicate their interest as promising therapeutic targets in allergic diseases.


Assuntos
Quimiocina CX3CL1/imunologia , Dermatite Atópica/imunologia , Receptores de Quimiocinas/imunologia , Pele/imunologia , Linfócitos T/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Receptor 1 de Quimiocina CX3C , Proliferação de Células , Células Cultivadas , Quimiocina CX3CL1/antagonistas & inibidores , Quimiocina CX3CL1/genética , Dermatite Atópica/genética , Citometria de Fluxo , Humanos , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Oligonucleotídeos/genética , Oligonucleotídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/imunologia , Receptores de Quimiocinas/genética , Pele/metabolismo , Pele/patologia , Linfócitos T/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo
19.
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
20.
Crit Care Med ; 40(11): 3026-33, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22926332

RESUMO

OBJECTIVES: To test the hypothesis that resveratrol would improve cardiac remodeling by inhibiting the detrimental effects of fractalkine. We previously reported that fractalkine exacerbates heart failure. Furthermore, this study sought to determine whether resveratrol targets fractalkine to improve myocardial ischemia and cardiac remodeling. DESIGN: Randomized and controlled laboratory investigation. SETTING: Research laboratory. SUBJECTS: Neonatal rat cardiac cells and C57BL/6 mice. INTERVENTIONS: Cardiac cells were treated with recombinant mouse soluble fractalkine for 24 hrs or pretreated with 25 µM resveratrol. Cardiomyocytes were exposed to anoxia/reoxygenation, H2O2, or pretreatment with resveratrol. Ex vivo murine hearts were perfusioned with soluble fractalkine or pretreated with resveratrol after global ischemia. Mice were subjected to the left coronary artery ligation to induce myocardial infarction and randomized to treatment with resveratrol or vehicle alone for 42 days. MEASUREMENTS AND MAIN RESULTS: In a murine myocardial infarction model, we found that resveratrol increased survival and delayed the progression of cardiac remodeling evaluated by serial echocardiography. At 6 wks, the heart weight/body weight ratio, lung weight/body weight ratio, and old infarct size were significantly smaller in resveratrol-treated mice than in untreated myocardial infarction mice. In cultures of neonatal rat cells, exposure to soluble fractalkine increased the atrial natriuretic peptide expression by cardiomyocytes, matrix metalloproteinase-9 and procollagen expression by fibroblasts, and intercellular adhesion molecule-1 expression by microvascular endothelial cells, while it decreased autophagy in cardiomyocytes. All these effects were blocked by coculture with resveratrol. The methyl thiazolyl tetrazolium assay showed that soluble fractalkine reduced the viability of cultured cardiomyocytes during exposure to anoxia/reoxygenation or H2O2, while pretreatment with resveratrol blocked this effect. Perfusion of ex vivo murine hearts with soluble fractalkine after global ischemia led to an increase of infarct size, which was prevented by pretreatment with resveratrol. CONCLUSION: Resveratrol alleviates the deleterious effects of fractalkine on myocardial ischemia and thus reduces subsequent cardiac remodeling.


Assuntos
Quimiocina CX3CL1/efeitos adversos , Insuficiência Cardíaca/tratamento farmacológico , Isquemia Miocárdica/tratamento farmacológico , Inibidores da Agregação Plaquetária/farmacologia , Estilbenos/uso terapêutico , Animais , Animais Recém-Nascidos , Quimiocina CX3CL1/antagonistas & inibidores , Modelos Animais de Doenças , Eletrocardiografia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Isquemia Miocárdica/fisiopatologia , Distribuição Aleatória , Ratos , Resveratrol , Remodelação Ventricular/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...