Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
ACS Omega ; 8(7): 6945-6958, 2023 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-36844596

RESUMO

A series of (8-hydroxyquinoline) gallium(III) complexes (CP-1-4) was synthesized and characterized by single X-ray crystallography and density functional theory (DFT) calculation. The cytotoxicity of the four gallium complexes toward a human nonsmall cell lung cancer cell line (A549), human colon cancer cell line (HCT116), and human normal hepatocyte cell line (LO2) was evaluated using MTT assays. CP-4 exhibited excellent cytotoxicity against HCT116 cancer cells (IC50 = 1.2 ± 0.3 µM) and lower toxicity than cisplatin and oxaliplatin. We also evaluated the anticancer mechanism studies in cell uptake, reactive oxygen species analysis, cell cycle, wound-healing, and Western blotting assays. The results showed that CP-4 affected the expression of DNA-related proteins, which led to the apoptosis of cancer cells. Moreover, molecular docking tests of CP-4 were performed to predict other binding sites and to confirm its higher binding force to disulfide isomerase (PDI) proteins. The emissive properties of CP-4 suggest that this complex can be used for colon cancer diagnosis and treatment, as well as in vivo imaging. These results also provide a foundation for the development of gallium complexes as potent anticancer agents.

2.
Int J Nanomedicine ; 18: 225-241, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36660337

RESUMO

Background: Gallium (III) metal-organic complexes have been shown to have the ability to inhibit tumor growth, but the poor water solubility of many of the complexes precludes further application. The use of materials with high biocompatibility as drug delivery carriers for metal-organic complexes to enhance the bioavailability of the drug is a feasible approach. Methods: Here, we modified the ligands of gallium 8-hydroxyquinolinate complex with good clinical anticancer activity by replacing the 8-hydroxyquinoline ligands with 5-bromo-8-hydroxyquinoline (HBrQ), and the resulting Ga(III) + HBrQ complex had poor water solubility. Two biocompatible materials, bovine serum albumin (BSA) and graphene oxide (GO), were used to synthesize the corresponding Ga(III) + HBrQ complex nanoparticles (NPs) BSA/Ga/HBrQ NPs and GO/Ga/HBrQ NPs in different ways to enhance the drug delivery of the metal complex. Results: Both of BSA/Ga/HBrQ NPs and GO/Ga/HBrQ NPs can maintain stable existence in different solution states. In vitro cytotoxicity test showed that two nanomedicines had excellent anti-proliferation effect on HCT116 cells, which shown higher level of intracellular ROS and apoptosis ratio than that of cisplatin and oxaliplatin. In addition, the superior emissive properties of BSA/Ga/HBrQ NPs and GO/Ga/HBrQ NPs allow their use for in vivo imaging showing highly effective therapy in HCT116 tumor-bearing mouse models. Conclusion: The use of biocompatible materials for the preparation of NPs against poorly biocompatible metal-organic complexes to construct drug delivery systems is a promising strategy that can further improve drug delivery and therapeutic efficacy.


Assuntos
Antineoplásicos , Portadores de Fármacos , Gálio , Grafite , Nanopartículas Metálicas , Oxiquinolina , Animais , Humanos , Camundongos , Materiais Biocompatíveis , Linhagem Celular Tumoral , Portadores de Fármacos/síntese química , Gálio/química , Grafite/química , Células HCT116 , Nanopartículas Metálicas/análise , Nanopartículas/análise , Oxiquinolina/química , Tamanho da Partícula , Soroalbumina Bovina/farmacologia , Água , Antineoplásicos/síntese química , Antineoplásicos/química
3.
Open Biol ; 11(6): 200340, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34102079

RESUMO

Immune cell infiltration plays a key role in acute kidney injury (AKI) to chronic kidney disease (CKD) progression. T lymphocytes, neutrophils, monocytes/macrophages and other immune cells regulate inflammation, tissue remodelling and repair. To determine the kinetics of accumulation of various immune cell populations, we established an animal model combining parabiosis and separation surgery to explore the fate and lifespan of peripheral leucocytes that migrate to the kidney. We found that peripheral T lymphocytes could survive for a long time (more than 14 days), whereas peripheral neutrophils survived for a short time in both healthy and ischaemia-induced damaged kidneys. Nearly half of the peripheral-derived macrophages disappeared after 14 days in normal kidneys, while their existing time in the inflammatory kidneys was prolonged. A fraction of F4/80high macrophages were renewed from the circulating monocyte pool. In addition, we found that after renal ischaemia reperfusion, neutrophils increased significantly in the early phase, and T lymphocytes mainly accumulated in the late stage, whereas macrophages infiltrated throughout AKI-CKD progression and were sustained longer in injured as opposed to normal kidneys. In conclusion, peripheral-derived macrophages, T lymphocytes and neutrophils exhibit different lifespans in the kidney, which may play different roles during AKI-CKD progression.


Assuntos
Injúria Renal Aguda/etiologia , Injúria Renal Aguda/metabolismo , Sistema Imunitário/citologia , Sistema Imunitário/fisiologia , Isquemia/complicações , Rim/imunologia , Longevidade , Parabiose , Injúria Renal Aguda/patologia , Animais , Biomarcadores , Modelos Animais de Doenças , Suscetibilidade a Doenças , Citometria de Fluxo , Imunofluorescência , Expressão Gênica , Imuno-Histoquímica , Linfócitos/imunologia , Linfócitos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Neutrófilos/imunologia , Neutrófilos/metabolismo
4.
Biometals ; 34(2): 277-289, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33389333

RESUMO

A new schiff base cobalt(III) complex [N,N'-bis(2'-hydroxyphenylacetone)-o-ethanediamine] cobalt(III) (M3) has been synthesized and characterized by single X-ray crystallography. The cytotoxicity of complex M3 was evaluated against HeLa, LoVo, A549, A549/cis cancer cell lines, and the normal cell lines LO2 by MTT assays. The IC50 is in the range of 6.27-22.68 µM, which is somewhat lower than cisplatin on the basis of platinum molar concentration. Furthermore, anticancer mechanistic studies showed that the complex M3 inhibited cell proliferation by blocking DNA synthesis and then acted on nuclear division of HeLa cells over time. Moreover, western blot analysis indicated M3 dramatically decreased the target protein c-Myc and KLF5 expression levels, and activated many signaling pathways including ER stress, apoptosis, cell cycle and DNA damage in HeLa. M3 did not affect proteasomal activity.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Cobalto/farmacologia , Complexos de Coordenação/farmacologia , Neoplasias do Colo do Útero/tratamento farmacológico , Antineoplásicos/síntese química , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cobalto/química , Complexos de Coordenação/síntese química , Complexos de Coordenação/química , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Bases de Schiff/química , Bases de Schiff/farmacologia , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia
5.
Theranostics ; 10(26): 12144-12157, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33204334

RESUMO

Rationale: Capillaries are composed of endothelial cells and the surrounding mural cells, pericytes. Microvascular repair after injury involves not only the proliferation of endothelial cells but also pericyte-based vessel stabilization. Exogenous bone marrow derived-putative endothelial progenitor cells (b-pEPCs) have the potential for vascular repair; however, their effect on vascular structure stabilization and pericyte-related pathobiological outcomes in the injured kidney has not been fully examined. Methods: We applied ischemia-reperfusion (IR) to induce renal vascular injury and renal fibrosis in mice. Platelet-derived growth factor receptor ß (PDGFR-ß)-DTR-positive mice were generated to deplete pericytes, and exogenous b-pEPCs and the PDGFR-ß ligand, PDGF chain B (PDGF-BB), were employed to explore the relationship among b-pEPCs, pericytes, vascular repair, and early renal fibrosis. Results: Administration of b-pEPCs reduced IR-induced pericyte-endothelial detachment, pericyte proliferation, and myofibroblast transition via a paracrine mode, which preserved not only vascular stabilization but also ameliorated IR-initiated renal fibrosis. PDGF-BB upregulated the expression of PDGFR-ß, exacerbated vascular abnormality, and pericyte-myofibroblast transition, which were ameliorated by b-pEPCs administration. The exogenous b-pEPCs and their culture medium (CM) induced vascular injury protection, and renal fibrosis was blocked by selective deletion of pericytes. Conclusion: Exogenous b-pEPCs directly protect against IR-induced vascular injury and prevent renal fibrosis by inhibiting the activation of PDGFR-ß-positive pericytes.


Assuntos
Injúria Renal Aguda/prevenção & controle , Células Progenitoras Endoteliais/transplante , Rim/patologia , Pericitos/patologia , Traumatismo por Reperfusão/prevenção & controle , Injúria Renal Aguda/patologia , Animais , Becaplermina/administração & dosagem , Becaplermina/metabolismo , Proliferação de Células , Modelos Animais de Doenças , Fibrose , Humanos , Injeções Intraperitoneais , Rim/irrigação sanguínea , Masculino , Camundongos , Miofibroblastos/patologia , Parabiose , Comunicação Parácrina , Proteínas , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Traumatismo por Reperfusão/patologia
6.
Int Immunopharmacol ; 88: 106854, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32771945

RESUMO

Acute kidney injury (AKI) to chronic kidney disease (CKD) progression has become a life-threatening disease. However, an effective therapeuticstrategyis still needed. The pathophysiology of AKI-to-CKD progression involves chronic inflammation and renal fibrosis driven by macrophage activation, which is physiologically dependent on colony-stimulating factor-1 receptor (CSF-1R) signaling. In this study, we modulated macrophage infiltration through oral administration of the CSF-1R inhibitor GW2580 in an ischemia-reperfusion (I/R)-induced AKI model to evaluate its therapeutic effects on preventing the progression of AKI to CKD. We found that GW2580 induced a significant reduction in the number of macrophages in I/R-injured kidneys and attenuated I/R-induced renal injury and subsequent interstitial fibrosis. By flow cytometry, we observed that the reduced macrophages were primarily Ly6C+ inflammatory macrophages in the GW2580-treated kidneys, while there was no significant difference in the number and percentage of Ly6C-CX3CR1+ macrophages. We further found that these reduced macrophages also demonstrated some characteristics of M2-like macrophages, which have been generally regarded as profibrotic subtypes in chronic inflammation. These results indicate the existence of phenotypic and functional crossover between Ly6C+ and M2-like macrophages in I/R kidneys, which induces AKI worsening to CKD. In conclusion, therapeutic GW2580 treatment alleviates acute renal injury and subsequent fibrosis by reducing Ly6C+ M2-like macrophage infiltration in ischemia-induced AKI.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Anisóis/farmacologia , Antígenos Ly/imunologia , Macrófagos/imunologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Receptor de Fator Estimulador de Colônias de Macrófagos/antagonistas & inibidores , Traumatismo por Reperfusão/complicações , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/imunologia , Injúria Renal Aguda/patologia , Animais , Anisóis/uso terapêutico , Antígenos Ly/efeitos dos fármacos , Antígenos Ly/metabolismo , Receptor 1 de Quimiocina CX3C/efeitos dos fármacos , Receptor 1 de Quimiocina CX3C/imunologia , Receptor 1 de Quimiocina CX3C/metabolismo , Modelos Animais de Doenças , Fibrose/tratamento farmacológico , Fibrose/etiologia , Fibrose/imunologia , Túbulos Renais/efeitos dos fármacos , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/uso terapêutico , Insuficiência Renal Crônica/tratamento farmacológico , Insuficiência Renal Crônica/etiologia , Insuficiência Renal Crônica/imunologia
7.
J Inorg Biochem ; 208: 111103, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32505045

RESUMO

A new Schiff base copper(II) complex [N,N'-bis(2'-hydroxyphenylacetone)-o-ethanediamine] copper (II) (M1) has been synthesized and characterized by single X-ray crystallography. The cytotoxicity of complex M1 was evaluated against HeLa, LoVo, A549, A549/cis cancer cell lines, and the normal cell lines LO2 and HUVEC, by MTT (3-(4,5-dimethylthiazoyl-2-yl)2,5-diphenyltetrazoliumbromide) assays. The IC50 (50% inhibition concentrations) is in the range of 5.13-11.68 µM, which is somewhat lower than cisplatin on the basis of platinum molar concentration. Furthermore, anticancer mechanistic studies showed that the complex M1 inhibited cell proliferation by blocking DNA synthesis and then acted on nuclear division of HeLa cells over time. Moreover, M1 increased intracellular ROS (Reactive oxygen species) levels in a dose-dependent manner. Western blot analysis indicated M1 dramatically decrease c-Myc transcription factor and KLF5 (Krüppel-like factor 5) protein expression levels in HeLa. M1 did not inhibit proteasomal activity. Finally, M1 induced DNA damages and activated the DNA damage repair pathways.


Assuntos
Antineoplásicos , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Complexos de Coordenação , Cobre , Neoplasias , Células A549 , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Complexos de Coordenação/química , Complexos de Coordenação/farmacocinética , Complexos de Coordenação/farmacologia , Cobre/química , Cobre/farmacocinética , Cobre/farmacologia , Células HeLa , Células Endoteliais da Veia Umbilical Humana , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Bases de Schiff/química , Bases de Schiff/farmacocinética , Bases de Schiff/farmacologia
8.
J Med Virol ; 92(10): 2159-2164, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32410245

RESUMO

Corona virus disease 2019 (COVID-19) was caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). The phenomenon of positive real time reverse transcription polymerase chain reaction (RT-PCR) result of SARS-CoV-2 in recovered patients had occurred and the research about these patients was rare. In our study, we did a retrospective review of medical records from COVID-19 patients admitted to one ward of Tongji Hospital of Hua Zhong University of Science and Technology from 10 February to 13 April 2020. From 10 February to 13 April 2020, there were 108 patients of COVID-19 admitted in the one ward of Tongji Hospital. Among them, eight cases were readmission patients because the RT-PCR result of SARS-CoV-2 was positive again after discharge. On the second admission, they had no symptoms and their chest computed tomography was almost normal. Data from laboratory tests of the readmission patients showed that all eight patients had normal white blood cell count, lymphocyte count. The inflammatory factors like procalcitonin and interleukin 6 were normal. After treatment, two patients met the standard and were discharged. The other six patients were still in the hospital because their RT-PCR of SARS-CoV-2 did not get three consecutive negative results and the course of two patients had persisted more than 90 days. We still needed to be alert that these patients could infect other people as a source of infection, and we also needed to be alert that these patients become chronic virus carriers. It also aroused our concern about the discharge standard of COVID-19.


Assuntos
COVID-19/epidemiologia , COVID-19/transmissão , Pandemias , Readmissão do Paciente , SARS-CoV-2/genética , Adulto , Idoso , Antivirais/uso terapêutico , Biomarcadores/sangue , Contagem de Células Sanguíneas , COVID-19/diagnóstico , Teste para COVID-19/métodos , Portador Sadio , China/epidemiologia , Feminino , Humanos , Interleucina-6/sangue , Masculino , Pessoa de Meia-Idade , Alta do Paciente , Pró-Calcitonina/sangue , Reação em Cadeia da Polimerase em Tempo Real , Estudos Retrospectivos , Tomografia Computadorizada por Raios X , Tratamento Farmacológico da COVID-19
9.
BMC Nephrol ; 20(1): 321, 2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31419967

RESUMO

BACKGROUND: Cardiovascular disease (CVD) events are the main cause of death in long-term hemodialysis (HD) patients. Macrophage colony- stimulating factor (M-CSF) is actively involved in the formation of atherosclerosis and causes plaque instability, thrombosis and the development of acute coronary syndromes. However, little information is available on the role of M-CSF in HD patients. We aimed to investigate the association between plasma M-CSF levels and CVD events as well as all-cause mortality in patients undergoing long-term HD. METHODS: Fifty two HD patients and 8 healthy controls were recruited in this study. HD patients were followed up from September 2014 to May 2017. The primary end point was CVD event, the secondary outcome was death from any cause. Patients were divided into two groups with low and high M-CSF levels based on the optimal cut-off value determined by the ROC curve. Cox regression analyses were used to assess the predictive value of plasma M-CSF for CVD events and all-cause mortality in HD patients. We tested the levels of plasma M-CSF and other inflammatory cytokines in surviving HD patients using ELISA or CBA kit. RESULTS: The average plasma level of M-CSF in 52 patients was approximately twice that of healthy controls (992.4 vs. 427.2 pg/mL; p <  0.05). During 32 months of follow-up, 26 patients (50.0%) had at least one CVD event and 8 patients (15.4%) died. The mean plasma M-CSF concentration increased in survivors after follow-up compared to that detected at baseline (1277.8 ± 693.3 vs. 997.2 ± 417.4 pg/mL; p <  0.05). Multivariate Cox regression analysis showed that plasma M-CSF is an independent risk factor for CVD events in HD patients (p <  0.05). In the Cox regression model after adjusting for gender and age, high M-CSF levels were related to an increased risk of all-cause death (p <  0.05). We also found that M-CSF levels were positively correlated with IL-6 and IL-18 levels (both p < 0.05), which are the major pathogentic cytokines that contribute to HD-related CVD events. CONCLUSION: M-CSF is a prognostic factor for CVD events and all-cause mortality in HD patients.


Assuntos
Doenças Cardiovasculares/etiologia , Fator Estimulador de Colônias de Macrófagos/sangue , Diálise Renal/mortalidade , Fatores Etários , Biomarcadores/sangue , Doenças Cardiovasculares/mortalidade , Estudos de Casos e Controles , Causas de Morte , Feminino , Humanos , Interleucina-18/sangue , Interleucina-6/sangue , Interleucinas/sangue , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Curva ROC , Valores de Referência , Análise de Regressão , Diálise Renal/efeitos adversos , Fatores Sexuais , Fator de Necrose Tumoral alfa/sangue
10.
PLoS One ; 12(3): e0172490, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28267753

RESUMO

Flows (Qa) of arteriovenous fistula (AVF) impact the dialysis adequacy in hemodialysis (HD) patients. However, data for different access flow levels on outcomes related to long-term dialysis patients, especially in Chinese patients, are limited. Herein, we performed an ambispective, mono-centric cohort study investigating the association between the AVF flows and inflammation, cardiovascular events and deaths in Chinese hemodialysis patients bearing a radio-cephalic fistula (AVF) from 2009 to 2015. Twenty-three patients (35.9%) developed at least one episode of cardiovascular disease (CVD) in two years after AVF creation. AVF Qa, IL-6 and hsCRP were significantly higher in patients with CVD than in patients without CVD. Multi-factorial binary logistic regression analysis found that the independent and strongest risk factor for CVD in HD patients was serum IL-6, which showed a positive association with AVF Qa levels in patients. Therefore, the linkage between AVF Qa tertiles and adverse clinical outcomes (cardiovascular events and mortality) was examined over a median follow-up of five years. IL-6 was significantly increased in the high AVF Qa (>1027.13 ml/min) group. Patients with median AVF Qa showed the lowest morbidity and mortality of CVD according to the AVF Qa tertiles, whereas higher Qa was associated with a higher risk of CVD, and lower AVF Qa (600 ml/min ≤AVF Qa <821.12 ml/min) had a higher risk of non-CVD death. Therefore, keeping the AVF Qa at an optimal level (821.12 to 1027.13 ml/min) would benefit HD patients, improve long-term clinical outcomes and lower AVF-induced inflammation.


Assuntos
Fístula Arteriovenosa/fisiopatologia , Hemodinâmica , Interleucina-6/sangue , Fluxo Sanguíneo Regional , Diálise Renal/mortalidade , Biomarcadores , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/mortalidade , Estudos de Coortes , Citocinas/sangue , Feminino , Humanos , Mediadores da Inflamação/sangue , Masculino , Morbidade , Diálise Renal/efeitos adversos , Diálise Renal/estatística & dados numéricos , Fatores de Risco , Análise de Sobrevida
11.
J Huazhong Univ Sci Technolog Med Sci ; 36(1): 48-52, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26838739

RESUMO

Biliverdin (BV) has long been thought to be a cytotoxic metabolic waste product. It has also been demonstrated to have important cytoprotective functions during oxidative stress. The present study aimed to examine the cytoprotective effect of BV on NRK-52E cells, a proximal tubular cell line derived from rat kidney. Cells were treated with 50 µmol/L cisplatin for 24 h (cisplatin group) or pre-treated with BV for 30 min, then with 50 µmol/L cisplatin for 24 h (cisplatin+BV group). Those given no treatment served as a control. Cell apoptosis was evaluated by flow cytometry and cell viability by Cell Counting Kit-8 (CCK-8). The protein expressions of cleaved caspase3, Bax and Bcl-2 were assessed by Western blotting. Reactive oxygen species (ROS) levels were measured using carboxydichlorodihydrofluorescein diacetate (H2DCF). The results showed that cisplatin induced the apoptosis of NRK-52E cells, decreased cell viability, and increased the formation of ROS by upregulating the expression of cleaved caspase3 and Bax and decreasing Bcl-2 protein expression. These effects could be significantly reversed by pretreatment with BV. It was concluded that BV can protect against cisplatin-induced cell apoptosis through the anti-oxidative effects.


Assuntos
Antioxidantes/farmacologia , Apoptose , Biliverdina/farmacologia , Células Epiteliais/efeitos dos fármacos , Animais , Linhagem Celular , Cisplatino/toxicidade , Células Epiteliais/metabolismo , Túbulos Renais/citologia , Ratos , Espécies Reativas de Oxigênio/metabolismo
12.
PLoS One ; 11(2): e0149926, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26900858

RESUMO

AMD3100 is a small molecule inhibitor of chemokine receptor type 4 (CXCR4), which is located in the cell membranes of CD34+ cells and a variety of inflammatory cells and has been reported to reduce organ fibrosis in the lung, liver and myocardium. However, the effect of AMD3100 on renal fibrosis is unknown. This study investigated the impact of AMD3100 on renal fibrosis. C57bl/6 mice were subjected to unilateral ureteral obstruction (UUO) surgery with or without AMD3100 administration. Tubular injury, collagen deposition and fibrosis were detected and analyzed by histological staining, immunocytochemistry and Western Blot. Bone marrow derived pro-angiogenic cells (CD45+, CD34+ and CD309+ cells) and capillary density (CD31+) were measured by flow cytometry (FACS) and immunofluorescence (IF). Inflammatory cells, chemotactic factors and T cell proliferation were characterized. We found that AMD3100 treatment did not alleviate renal fibrosis but, rather, increased tissue damage and renal fibrosis. Continuous AMD3100 administration did not improve bone marrow derived pro-angiogenic cells mobilization but, rather, inhibited the migration of bone marrow derived pro-angiogenic cells into the fibrotic kidney. Additionally, T cell infiltration was significantly increased in AMD3100-treated kidneys compared to un-treated kidneys. Thus, treatment of UUO mice with AMD3100 led to an increase in T cell infiltration, suggesting that AMD3100 aggravated renal fibrosis.


Assuntos
Medula Óssea/patologia , Compostos Heterocíclicos/farmacologia , Inflamação/patologia , Rim/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Linfócitos T/patologia , Animais , Benzilaminas , Complexo CD3/metabolismo , Proliferação de Células/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Colágeno Tipo IV/metabolismo , Ciclamos , Citocinas/metabolismo , Fibrose , Hipóxia/complicações , Hipóxia/patologia , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/patologia , Masculino , Camundongos Endogâmicos C57BL , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Linfócitos T/efeitos dos fármacos , Obstrução Ureteral/complicações
13.
Int J Mol Sci ; 16(9): 22621-35, 2015 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-26393580

RESUMO

Biliverdin reductase A is an enzyme, with serine/threonine/tyrosine kinase activation, converting biliverdin (BV) to bilirubin (BR) in heme degradation pathway. It has been reported to have anti-inflammatory and antioxidant effect in monocytes and human glioblastoma. However, the function of BVRA in polarized macrophage was unknown. This study aimed to investigate the effect of BVRA on macrophage activation and polarization in injured renal microenvironment. Classically activated macrophages (M1macrophages) and alternative activation of macrophages (M2 macrophages) polarization of murine bone marrow derived macrophage was induced by GM-CSF and M-CSF. M1 polarization was associated with a significant down-regulation of BVRA and Interleukin-10 (IL-10), and increased secretion of TNF-α. We also found IL-10 expression was increased in BVRA over-expressed macrophages, while it decreased in BVRA knockdown macrophages. In contrast, BVRA over-expressed or knockdown macrophages had no effect on TNF-α expression level, indicating BVRA mediated IL-10 expression in macrophages. Furthermore, we observed in macrophages infected with recombinant adenoviruses BVRA gene, which BVRA over-expressed enhanced both INOS and ARG-1 mRNA expression, resulting in a specific macrophage phenotype. Through in vivo study, we found BVRA positive macrophages largely existed in mice renal ischemia perfusion injury. With the treatment of the regular cytokines GM-CSF, M-CSF or LPS, excreted in the injured renal microenvironment, IL-10 secretion was significantly increased in BVRA over-expressed macrophages. In conclusion, the BVRA positive macrophage is a source of anti-inflammatory cytokine IL-10 in injured kidney, which may provide a potential target for treatment of kidney disease.


Assuntos
Regulação da Expressão Gênica , Interleucina-10/imunologia , Rim/patologia , Macrófagos/imunologia , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/imunologia , Insuficiência Renal/patologia , Traumatismo por Reperfusão/patologia , Animais , Linhagem Celular , Polaridade Celular , Células Cultivadas , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Interleucina-10/genética , Rim/imunologia , Rim/metabolismo , Lipopolissacarídeos/imunologia , Ativação de Macrófagos , Fator Estimulador de Colônias de Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , Insuficiência Renal/genética , Insuficiência Renal/imunologia , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/imunologia
14.
PLoS One ; 8(10): e76836, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24130796

RESUMO

Excess mesangial extracellular matrix (ECM) and mesangial cell proliferation is the major pathologic feature of diabetic nephropathy (DN). Fenofibrate, a PPARα agonist, has been shown to attenuate extracellular matrix formation in diabetic nephropathy. However, the mechanisms underlying this effect remain to be elucidated. In this study, the effect of fenofibrate on high-glucose induced cell proliferation and extracellular matrix exertion and its mechanisms were investigated in cultured rat mesangial cells by the methylthiazoletetrazolium (MTT) assay, flow cytometry and western blot. The results showed that treatment of mesangial cells (MCs) with fenofibrate repressed high-glucose induced up-regulation of extracellular matrix Collagen-IV, and inhibited entry of cell cycle into the S phase. This G1 arrest and ECM inhibition was caused by the reduction of phosphorylation and activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and AKT. On the contrary, PPARα siRNA accelerated high glucose-induced cell cycle progression by ERK1/2 and AKT activation. Taken together, fenofibrate ameliorated glucose-induced mesangial cell proliferation and matrix production via its inhibition of PI3K/AKT and ERK1/2 signaling pathways. Such mechanisms may contribute to the favorable effects of treatment using fenofibrate in diabetic nephropathy.


Assuntos
Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/enzimologia , Fenofibrato/farmacologia , Glucose/farmacologia , Células Mesangiais/citologia , Células Mesangiais/efeitos dos fármacos , Animais , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo IV/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Matriz Extracelular/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células Mesangiais/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , PPAR alfa/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Regulação para Cima/efeitos dos fármacos
15.
Int J Biol Sci ; 8(6): 859-69, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22745576

RESUMO

Cdc42-interacting protein-4 (CIP4) is an F-BAR (Fer/CIP4 and Bin, amphiphysin, Rvs) family member that regulates membrane deformation and endocytosis, playing a key role in extracellular matrix (ECM) deposition and invasion of cancer cells. These processes are analogous to those observed during the initial epithelial-mesenchymal transition (EMT) of renal tubular epithelial cells. The role of CIP4 in renal tubular EMT and renal tubulointerstitial fibrosis was investigated over the course of the current study, demonstrating that the expression of CIP4 increased in the tubular epithelia of 5/6-nephrectomized rats and TGF-ß1 treated HK-2 cells. Endogenous CIP4 evidenced punctate localization throughout the cytosol, with elevated levels observed in the perinuclear region of HK-2 cells. Subsequent to TGF-ß1 treatment, CIP4 expression increased, forming clusters at the cell periphery that gradually redistributed into the cytoplasm. Simultaneously, EMT induction in cells was confirmed by the prevalence of morphological changes, loss of E-cadherin, increase in α-SMA expression, and secretion of fibronectin. Overexpression of CIP4 promoted characteristics similar to those commonly observed in EMT, and small interfering RNA (siRNA) molecules capable of CIP4 knockdown were used to demonstrate reversed EMT. Cumulatively, results of the current study suggest that CIP4 promotes TGF-ß1-induced EMT in tubular epithelial cells. Through this mechanism, CIP4 is capable of inducing ECM deposition and exacerbating progressive fibrosis in chronic renal failure.


Assuntos
Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Matriz Extracelular/metabolismo , Túbulos Renais Proximais/citologia , Proteínas Associadas aos Microtúbulos/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Animais , Western Blotting , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/citologia , Transição Epitelial-Mesenquimal/genética , Matriz Extracelular/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Masculino , Proteínas Associadas aos Microtúbulos/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Nephrol Dial Transplant ; 26(4): 1149-56, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20959347

RESUMO

BACKGROUND: The p38 mitogen-activated protein kinase (p38 MAPK) is an important intracellular signal transduction pathway involved in TGF-ß1-induced epithelial-mesenchymal transition (EMT). Sema4C, a member of the semaphorin family, was found to be essential for the activation of p38 MAPK. However, the role of Sema4C in promoting TGF-ß1-induced EMT is unclear. METHODS: Renal fibrosis was induced by 5/6 subtotal nephrectomy rat model. In vitro, Sema4C was induced in human proximal tubular epithelial cells (HKC) by treatment with TGF-ß1, or was inhibited by siRNA or was over-expressed by Sema4C transfection. The selective p38 MAPK inhibitor, SB203580, was administered to inhibit the p38 pathway. The expression of Sema4C, the markers of EMT, p38 phosphorylation and fibronectin secretion were measured by western blotting, immunohistochemistry, immunocytochemistry or enzyme-linked immunosorbent assay. RESULTS: The expression of Sema4C increased in HKC cells that were treated with TGF-ß1. Knockdown of Sema4C potently inhibited phosphorylation of p38 MAPK and reversed TGF-ß1-induced EMT. Over-expression of Sema4C via Sema4C transfection elicited p38 MAPK phosphorylation and promoted EMT. The effects of Sema4C during EMT were blocked by a p38-specific inhibitor. In vivo, the expression of Sema4C increased in the tubular epithelia of 5/6-nephrectomized rats and human fibrotic renal tissue, and similar localization of phosphorylated p38 and Sema4C was demonstrated by immunohistochemistry on serial sections. CONCLUSIONS: Our findings suggest that Sema4C plays an important role in TGF-ß1-induced EMT through activation of p38 MAPK in proximal tubular epithelial cells.


Assuntos
Transição Epitelial-Mesenquimal , Glomerulosclerose Segmentar e Focal/metabolismo , Túbulos Renais Proximais/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Semaforinas/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Animais , Western Blotting , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Fibrose/metabolismo , Fibrose/patologia , Glomerulosclerose Segmentar e Focal/patologia , Humanos , Técnicas Imunoenzimáticas , Túbulos Renais Proximais/citologia , Masculino , Nefrectomia , Fosforilação , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Semaforinas/antagonistas & inibidores , Semaforinas/genética
17.
Yao Xue Xue Bao ; 45(9): 1103-8, 2010 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-21351565

RESUMO

This study is to explore the effect of ginkgolide B (BN52021) on the production of nitric oxide (NO), interleukin (IL)-6 and regulated upon activation normal T cell expressed and secreted (RANTES) from astrocytes induced by stimulators. Primary cultured rat astrocytes were stimulated with lipopolysaccharides (LPS), the production of NO was assayed using Griess reaction; U251 cells were stimulated with IL-1 beta, the contents of IL-6 and RANTES in the supernatant were measured using ELISA. The mRNA expressions of IL-6 and RANTES were detected using RT-PCR. LPS (10 ng mL(-1) to 10 microg mL(-1)) could stimulate rat astrocytes to produce NO in a dose-dependent manner. Ginkgolide B at the concentrations of 0.1-10 micromol L(-1) were shown to decrease NO production significantly. IL-1 beta could induce the mRNA expression and protein secretion of IL-6 from U251 cells, as well as RANTES. Ginkgolide B at concentrations of 0.1-10 micromol L(-1) were shown to inhibit RANTES secretion, and to inhibit mRNA expression of IL-6 and RANTES at concentration of 10 micromol L(-1). Ginkgolide B has inhibitory effect on the production of NO, IL-6 and RANTES from astrocytes treated with inflammatory stimulators.


Assuntos
Astrócitos/metabolismo , Quimiocina CCL5/metabolismo , Ginkgolídeos/farmacologia , Interleucina-6/metabolismo , Lactonas/farmacologia , Óxido Nítrico/metabolismo , Animais , Astrócitos/citologia , Linhagem Celular Tumoral , Células Cultivadas , Quimiocina CCL5/genética , Relação Dose-Resposta a Droga , Ginkgolídeos/administração & dosagem , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Interleucina-1beta , Interleucina-6/genética , Lactonas/administração & dosagem , Lipopolissacarídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Ativação de Plaquetas/antagonistas & inibidores , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar
18.
Yao Xue Xue Bao ; 40(4): 311-5, 2005 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-16011257

RESUMO

AIM: To investigate the inhibitory effect of ginkgolide B on angiogenesis in chronic inflammation and the possible mechanisms. METHODS: The murine chronic granulomatous air pouch model was used to observe the anti-angiogenesis effect of ginkgolide B. The vascular index was determined by colorimetry of carminic acid, and angiogenesis was observed by histology method. The interleukin-1beta (IL-1beta) levels in mice serum and in supernatants of U937 cell culture stimulated by phorbol 12-myristate 13-acetate (PMA) were detected by radioimmunoassay (RIA). The tumor necrosis factor-alpha (TNF-alpha) levels in mice serum and in supernatant of U937 cell culture were measured by cytotoxicity bioassay. The mRNA expression of IL-1beta and TNF-alpha of U937 cell culture was investigated by RT-PCR. RESULTS: Oral administration of ginkgolide B 25 and 100 mg x kg(-1) was shown to significantly inhibit the vascular index of murine chronic granulomatous air pouch model with the inhibitory rate of 22.52% and 25.29%, respectively. This result was supported by histological observation. Concomitantly, the IL-1beta levels in mice serums were also significantly decreased with the inhibitory rate of 50.61% and 58.66%; so were the TNF-alpha levels with the inhibitory rate of 28.91% and 52.41%. Ginkgolide B at concentration of 1 x 10(-5) to 1 x 10(-8) mol x L(-1) could also reduce both the IL-1beta and TNF-alpha contents in the supernatants of U937 cell culture stimulated by PMA, but the scopes of changes were much different. For IL-1beta the IC50 was 1.93 x 10(-8) mol x L(-1), while ginkgolide B at concentration of 1 x 10(-5) mol x L(-1) only decreased the release of TNF-alpha by 25.99%. Furthermore, ginkgolide B at concentrations of 1 x 10(-5) to 1 x 10(-7) mol x L(-1) was shown to significantly inhibit TNF-alpha mRNA expression of U937 cells; and at concentrations of 1 x 10(-5) and 1 x 10(-6) mol x L(-1) could inhibit IL-1beta mRNA expression. CONCLUSION: Ginkgolide B was shown to significantly inhibit angiogenesis of the murine chronic granulomatous air pouch model, reduce the IL-1beta and TNF-alpha levels in mice serums, and significantly inhibit IL-1beta and TNF-alpha mRNA expression and protein secretion in supernatants of U937 cell culture. It was suggested that reduction of proangiogenic cytokines IL-1beta and TNF-alpha secretion may contribute to the anti-angiogenesis effect of ginkgolide B in the murine chronic granulomatous air pouch model.


Assuntos
Diterpenos/farmacologia , Granuloma/patologia , Interleucina-1/biossíntese , Lactonas/farmacologia , Neovascularização Patológica/patologia , Fator de Necrose Tumoral alfa/biossíntese , Animais , Linhagem Celular , Feminino , Fibrinolíticos/farmacologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Ginkgolídeos , Granuloma/metabolismo , Humanos , Inflamação/metabolismo , Inflamação/patologia , Interleucina-1/genética , Camundongos , Camundongos Endogâmicos BALB C , Fator de Ativação de Plaquetas/antagonistas & inibidores , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Fator de Necrose Tumoral alfa/genética , Células U937/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA