Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 173
Filtrar
1.
Int. j. med. surg. sci. (Print) ; 9(2): 1-9, June 2022. ilus, graf
Artigo em Inglês | LILACS | ID: biblio-1512600

RESUMO

Cisplatin, the first platinum compound approved for cancer treatment, is widely used in the treatment of various cancers including hepatocellular carcinoma (HCC). HCC incidence rates rise globally. Epithelial mesenchymal transition (EMT) is implicated in cancer invasion and metastasis, which are associated with increased mortality. Cisplatin dose might influence cancer invasion and metastatic behavior of the cells. The aim of the study was to investigate the effect of low-dose cisplatin treatment on EMT- related changes in HepG2 cells. Following treatment with 4 µM cisplatin, HepG2 cells were evaluated morphologically. Gene expression of E-cadherin, Vimentin, Snail1 was assessed by quantitative PCR. Immunofluorescence analyses of NA-K ATPase were performed. Although the low-dose cisplatin treated cells exhibited a more stretched morphology, no statistical difference was detected in gene expression of E-cadherin, Vimentin, Snail1 and immunofluorescence of NA-K ATPase. Findings on low-dose cisplatin effects in HepG2 might contribute to the knowledge of antineoplastic inefficacy by further understanding the molecular mechanisms of drug action.


El cisplatino, el primer compuesto de platino aprobado para el tratamiento del cáncer, es ampliamente utilizado en el tratamiento de varios tipos de cáncer, incluido el carcinoma hepatocelular (CHC). Las tasas de incidencia de CHC aumentan a nivel mundial. La transición mesenquimal epitelial (EMT) está implicada en la invasión del cáncer y la metástasis, que se asocian con un aumento de la mortalidad. La dosis de cisplatino podría influir en la invasión del cáncer y el comportamiento metastásico de las células. El objetivo del estudio fue investigar el efecto del tratamiento con dosis bajas de cisplatino en los cambios relacionados con la EMT en las células HepG2. Tras el tratamiento con cisplatino de 4 µM, se evaluaron morfológicamente las células HepG2. La expresión génica de E-cadherina, vimentina, caracol1 se evaluó mediante PCR cuantitativa. Se realizaron análisis de inmunofluorescencia de NA-K ATPasa . Aunque las células tratadas con cisplatino en dosis bajas exhibieron una morfología más estirada, no se detectaron diferencias estadísticas en la expresión génica de E-cadherina, vimentina, Snail1 e inmunofluorescencia de NA-K ATPasa. Los hallazgos sobre los efectos del cisplatino en dosis bajas en HepG2 podrían contribuir al conocimiento de la ineficacia antineoplásica al comprender mejor los mecanismos moleculares de la acción del fármaco.


Assuntos
Humanos , Cisplatino/administração & dosagem , Antineoplásicos/administração & dosagem , Vimentina/efeitos dos fármacos , Vimentina/genética , Vimentina/metabolismo , Caderinas/efeitos dos fármacos , Caderinas/genética , Caderinas/metabolismo , Células Cultivadas , Imunofluorescência , Microscopia Confocal , Células Hep G2 , Transição Epitelial-Mesenquimal , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição da Família Snail/efeitos dos fármacos , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Invasividade Neoplásica
2.
Anticancer Drugs ; 32(8): 786-792, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-33675608

RESUMO

Stachydrine is a bioactive alkaloid that has been found to exert tumor-suppressive potential. However, the effect of stachydrine on hepatocellular carcinoma (HCC) has not been previously investigated. In the present study, we investigated the effect of transforming growth factor-ß1 (TGF-ß1)-induced epithelial-mesenchymal transition (EMT) in HepG2 cells. Our results showed that stachydrine significantly suppressed TGF-ß1-induced HepG2 cell migration and invasion in a dose-dependent manner. Stachydrine prevented TGF-ß1-induced EMT in HepG2 cells, as proved by the increased expression level of E-cadherin and decreased expression levels of N-cadherin and vimentin. In addition, stachydrine attenuated TGF-ß1-induced upregulation of TGF-ß receptor I (TßRI) in both protein and mRNA levels. Further mechanism investigations proved that stachydrine prevented TGF-ß1-induced activation of Smad2/3 and phosphoinositol-3-kinase (PI3K)/Akt/mTOR signaling pathways in HepG2 cells. In conclusion, these findings demonstrated that stachydrine prevented TGF-ß1-induced EMT in HCC cells through Smad2/3 and PI3K/Akt/mTOR signaling pathways. Thus, stachydrine might be a potential therapeutic agent for the treatment of HCC.


Assuntos
Carcinoma Hepatocelular/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Neoplasias Hepáticas/patologia , Prolina/análogos & derivados , Fator de Crescimento Transformador beta1/efeitos dos fármacos , Caderinas/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular , Relação Dose-Resposta a Droga , Células Hep G2 , Humanos , Fosfatidilinositol 3-Quinases , Prolina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/efeitos dos fármacos , Serina-Treonina Quinases TOR/efeitos dos fármacos , Vimentina/efeitos dos fármacos
3.
Cell Stress Chaperones ; 26(3): 505-513, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33598875

RESUMO

Heat shock protein 70 (HSP70) is a key member of the HSP family that contributes to a pre-cancerous environment; however, its role in lung cancer remains poorly understood. The present study used geranylgeranylacetone (GGA) to induce HSP70 expression, and transforming growth factor-ß (TGF-ß) was used to construct an epithelial-mesenchymal transition (EMT) model by stimulating A549 cells in vitro. Western Blot was performed to detect protein levels of NADPH oxidase 4 (NOX4) and the EMT-associated proteins E-cadherin and vimentin both before and after HSP70 expression. Cell morphological changes were observed, and the effect of HSP70 on cell migration ability was detected via the wound healing. The results demonstrated that GGA at 50 and 200 µmol/L could significantly induce HSP70 expression in A549 cells (P < 0.05). Furthermore, HSP70 induced by 200 µmol/L GGA significantly inhibited the changes of E-cadherin, vimentin, and cell morphology induced by TGF-ß (P < 0.05), while HSP70 induced by 50 µmol/L GGA did not. The results of the wound healing assay indicated that 200 µmol/L GGA significantly inhibited A549 cell migration induced by TGF-ß. Taken together, the results of the present study demonstrated that overexpression of HSP70 inhibited the TGF-ß induced EMT process and changed the cell morphology and migratory ability induced by TGF-ß in A549 cells.


Assuntos
Movimento Celular/fisiologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Proteínas de Choque Térmico HSP70/efeitos dos fármacos , Proteínas de Choque Térmico HSP70/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Células A549 , Caderinas/efeitos dos fármacos , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/fisiologia , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo
4.
Anticancer Drugs ; 32(3): 314-322, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33394687

RESUMO

Evodiamine (Evo), a quinazoline alkaloid and one of the most typical polycyclic heterocycles, is mainly isolated from Evodia rugulosa. Vasculogenic mimicry (VM) is a newly identified way of angiogenesis during tumor neovascularization, which is prevalent in a variety of highly invasive tumors. The purpose of this study was to investigate the effect and mechanism of Evo on VM in human colorectal cancer (CRC) cells. The number of VM structures was calculated by the three-dimensional culture of human CRC cells. Wound-healing was used to detect the migration of HCT116 cells. Gene expression was detected by reverse transcription-quantitative PCR assay. CD31/PAS staining was used to identify VM. Western blotting and immunofluorescence were used to detect protein levels. The results showed that Evo inhibited the migration of HCT116 cells, as well as the formation of VM. Furthermore, Evo reduced the expression of hypoxia-inducible factor 1-alpha (HIF-1α), VE-cadherin, VEGF, MMP2, and MMP9. In a model of subcutaneous xenotransplantation, Evo also inhibited tumor growth and VM formation. Our study demonstrates that Evo could inhibit VM in CRC cells HCT116 and reduce the expression of HIF-1α, VE-cadherin, VEGF, MMP2, and MMP9.


Assuntos
Neovascularização Patológica/tratamento farmacológico , Quinazolinas/farmacologia , Animais , Antígenos CD/efeitos dos fármacos , Caderinas/efeitos dos fármacos , Movimento Celular , Sobrevivência Celular , Transição Epitelial-Mesenquimal , Feminino , Células HCT116 , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Neovascularização Patológica/patologia , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Cicatrização/efeitos dos fármacos
5.
Curr Res Transl Med ; 69(1): 103268, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33069641

RESUMO

AIM: Drug delivery is crucial for therapeutic efficacy and gap junction communication channels (GJIC) facilitate movement within the tumour. Pro-drug activation, a modality of cancer therapy leads to Ganciclovir triphosphate (GCV-TP) incorporation into newly synthesized DNA resulting in cell death. The objective was to enhance, with Histone deacetylase inhibitors (HDACi) and All Trans Retinoic Acid (ATRA), GJIC, crucial for drug delivery, and with combination, abrogate the observed detrimental effect of Dexamethasone (DXM). METHODS: Cell lines (NT8E, and HeLa) were pre-treated with Valproic Acid (VPA) (1 mM), 4 Phenyl Butyrate (4PB) (2 mM), ATRA (10 µM) and Dexamethasone (1 µM). Protein quantitated with the Bicinchoninic (BCA) assay for cell lysates, membrane and soluble fractions was assessed with Western blotting for Connexins (43, 26 and 32) and E-Cadherin. A qRT-PCR was done for CX 43-GJA1, CX 26-GJB2, CX 32-GJB1 and E-Cadherin, and normalized with Glyceraldehyde Phosphate dehydrogenase (GAPDH). Further, localization of Connexins (CX) and E-Cadherin, GJIC competence, pre-clinical in-vitro studies and the mechanism of cell death were evaluated. RESULTS: There was no toxicity or change in growth patterns observed with the drugs. In both the cell lines CX 43 localized to the membrane whereas CX 32 and CX 26 were present but not membrane bound. E-Cadherin was present on the membrane in NT8E and completely absent in HeLa cells. Effects of HDACi, DXM and ATRA were seen on the expression of Connexins and E-Cadherin in both the cell lines. NT8E and HeLa cell lines showed enhanced GJIC with 4PB [30 %], VPA [36 %] and ATRA [54 %] with a 60 % increase in cytotoxicity and an abrogation of Dexamethasone inhibition on combination with VPA or ATRA. CONCLUSION: An enhancement of GJIC function by HDACi and ATRA increased cytotoxicity and could be effective in the presence of Dexamethasone, when combined with ATRA or VPA.


Assuntos
Antineoplásicos/farmacologia , Junções Comunicantes/efeitos dos fármacos , Terapia de Alvo Molecular/métodos , Neoplasias/tratamento farmacológico , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Caderinas/efeitos dos fármacos , Caderinas/genética , Caderinas/metabolismo , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/fisiologia , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Conexinas/efeitos dos fármacos , Conexinas/genética , Conexinas/metabolismo , Dexametasona/administração & dosagem , Dexametasona/farmacologia , Ganciclovir/administração & dosagem , Ganciclovir/análogos & derivados , Ganciclovir/farmacologia , Junções Comunicantes/fisiologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Inibidores de Histona Desacetilases/administração & dosagem , Inibidores de Histona Desacetilases/farmacologia , Humanos , Neoplasias/genética , Neoplasias/patologia , Tretinoína/administração & dosagem , Tretinoína/farmacologia , Ácido Valproico/administração & dosagem , Ácido Valproico/farmacologia
6.
Arthritis Rheumatol ; 73(3): 520-529, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33001586

RESUMO

OBJECTIVE: Intravenous iloprost improves Raynaud's phenomenon (RP) and promotes healing of digital ulcers in systemic sclerosis (SSc; scleroderma). Despite a short half-life, its clinical efficacy lasts weeks. Endothelial adherens junctions, which are formed by VE-cadherin clustering between endothelial cells (ECs), regulate endothelial properties including barrier function, endothelial-to-mesenchymal transition (EndoMT), and angiogenesis. We undertook this study to investigate the hypothesis that junctional disruption contributes to vascular dysfunction in SSc, and that the protective effect of iloprost is mediated by strengthening of those junctions. METHODS: Dermal ECs from SSc patients and healthy controls were isolated. The effect of iloprost on ECs was examined using immunofluorescence, permeability assays, Matrigel tube formation, and quantitative polymerase chain reaction. RESULTS: Adherens junctions in SSc were disrupted compared to normal ECs, as indicated by reduced levels of VE-cadherin and increased permeability in SSc ECs (P < 0.05). Iloprost increased VE-cadherin clustering at junctions and restored junctional levels of VE-cadherin in SSc ECs (mean ± SD 37.3 ± 4.3 fluorescence units) compared to normal ECs (mean ± SD 29.7 ± 3.4 fluorescence units; P < 0.05), after 2 hours of iloprost incubation. In addition, iloprost reduced permeability of monolayers, increased tubulogenesis, and blocked EndoMT in both normal and SSc ECs (n ≥ 3; P < 0.05). The effects in normal ECs were inhibited by a function-blocking antibody that prevents junctional clustering of VE-cadherin. CONCLUSION: Our data suggest that the long-lasting effects of iloprost reflect its ability to stabilize adherens junctions, resulting in increased tubulogenesis and barrier function and reduced EndoMT. These findings provide a mechanistic basis for the use of iloprost in treating SSc patients with RP and digital ulcers.


Assuntos
Junções Aderentes/efeitos dos fármacos , Antígenos CD/efeitos dos fármacos , Caderinas/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Iloprosta/farmacologia , Doença de Raynaud/tratamento farmacológico , Esclerodermia Difusa/tratamento farmacológico , Vasodilatadores/farmacologia , Junções Aderentes/metabolismo , Antígenos CD/metabolismo , Caderinas/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Estudos de Casos e Controles , Células Cultivadas , Células Endoteliais/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Humanos , Iloprosta/uso terapêutico , Masculino , Pessoa de Meia-Idade , Neovascularização Fisiológica/efeitos dos fármacos , Doença de Raynaud/etiologia , Doença de Raynaud/fisiopatologia , Esclerodermia Difusa/complicações , Esclerodermia Difusa/fisiopatologia , Vasodilatadores/uso terapêutico
7.
Drug Des Devel Ther ; 14: 4205-4214, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33116408

RESUMO

AIM: To examine the effects of platelet-activating factor (PAF) on the barrier functions of cultured retinal pigment epithelial (RPE) cells. METHODS: A human RPE cell line (ARPE-19) was cultured on microporous filter supports and treated with PAF and WEB 2086, a specific PAF-receptor (PAF-R) antagonist. The permeability of the RPE monolayer was measured using transepithelial electrical resistance (TER) and sodium fluorescein flux. The expression of the tight junction protein zonula occludens (ZO)-1 and the adherens junction protein N-cadherin was assessed using immunohistochemistry and Western blotting. We also measured the vascular endothelial growth factor (VEGF) concentrations in PAF-treated cultures and re-measured RPE monolayer permeability in the presence of VEGF-neutralizing antibodies. RESULTS: PAF significantly decreased the TER and enhanced the sodium fluorescein flux of the RPE monolayer and downregulated the expression of ZO-1 and N-cadherin. These effects were abolished by WEB 2086-mediated blockage of the PAF-R. PAF stimulation increased VEGF expression in RPE cells, and the antibody-mediated neutralization of VEGF caused a partial recovery of the barrier properties. CONCLUSION: The barrier functions of ARPE-19 cells were altered by PAF, and these effects were partly mediated by an upregulation of VEGF expression in these cells. Our results contribute to the growing body of evidence supporting the role of PAF in choroidal neovascularization. Our findings suggest that PAF is a novel target in the development of therapies for increased permeability of the RPE monolayer.


Assuntos
Fator de Ativação de Plaquetas/farmacologia , Epitélio Pigmentado da Retina/efeitos dos fármacos , Azepinas/farmacologia , Caderinas/efeitos dos fármacos , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Impedância Elétrica , Humanos , Neovascularização Fisiológica/efeitos dos fármacos , Fator de Ativação de Plaquetas/antagonistas & inibidores , Inibidores da Agregação Plaquetária/farmacologia , Epitélio Pigmentado da Retina/citologia , Junções Íntimas/efeitos dos fármacos , Triazóis/farmacologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Proteína da Zônula de Oclusão-1/efeitos dos fármacos
8.
Respir Physiol Neurobiol ; 276: 103402, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32006666

RESUMO

BACKGROUND: Epithelial-mesenchymal transformation (EMT) is a central mechanism for the occurrence and development of pulmonary fibrosis. Therefore, to identify the key target molecules regulating the EMT process is considered as an important direction for the prevention and treatment of pulmonary fibrosis. Transglutaminase 2 (TG2) has been recently found to play an important role in the regulation of inflammation and the generation of extracellular matrix. Here, our study focuses on the roles of TG2 in pulmonary fibrosis and EMT. METHODS: at first, the expression of TG2 and the EMT-related markers like E-cadherin, Vimentin, and α-SMA were detected with Western Blotting, immunohistochemistry and other methods in the mice with pulmonary fibrosis induced by bleomycin. Further, MLE 12 cells were used to study the effects on EMT of the inhibition of TG2 in vitro. Finally, GK921, an inhibitor against TG2, was used to show its function in both prevention and treatment of pulmonary fibrosis induced by bleomycin in mice. RESULTS: bleomycin succeeded to induce pulmonary fibrosis in mice, with increased TG2 expression, EMT and Akt activation. Knock-down of TG2 by siRNA technique in MLE 12 cell (a mouse alveolar epithelial cell line) and GK921 (an inhibitor of TG2) all inhibited the EMT process, however SC79, an activator of Akt rescued above inhibition. Finally, GK921 alleviated pulmonary fibrosis in mice induced by bleomycin. CONCLUSION: Blocking TG2 reduces bleomycin-induced pulmonary fibrosis in mice via inhibiting EMT.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Proteínas de Ligação ao GTP/genética , Fibrose Pulmonar/genética , Transglutaminases/genética , Acetatos/farmacologia , Actinas/efeitos dos fármacos , Actinas/metabolismo , Animais , Antibióticos Antineoplásicos/toxicidade , Benzopiranos/farmacologia , Bleomicina/toxicidade , Caderinas/efeitos dos fármacos , Caderinas/metabolismo , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Proteínas de Ligação ao GTP/antagonistas & inibidores , Proteínas de Ligação ao GTP/metabolismo , Técnicas de Silenciamento de Genes , Técnicas In Vitro , Camundongos , Proteína 2 Glutamina gama-Glutamiltransferase , Fibrose Pulmonar/metabolismo , Pirazinas/farmacologia , Mucosa Respiratória/citologia , Transglutaminases/antagonistas & inibidores , Transglutaminases/metabolismo , Vimentina/efeitos dos fármacos , Vimentina/metabolismo
9.
Biotech Histochem ; 95(5): 341-348, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31937145

RESUMO

Expression of cyclo-oxygenase-2 (COX-2) and protein phosphatase 2A (PP2A) deactivation occurs frequently in oral squamous cell carcinoma (OSCC). We initially assessed COX-2 and PP2A protein expression in OSCC specimens using immunohistochemical (IHC) staining and western blot analysis. We found strong COX-2 and phosphorylated PP2A (p-PP2A) expression in OSCC samples. No significant difference in total PP2A expression was observed between cancer and nontumor tissues. The effect of combining COX-2 inhibitor and celecoxib (CXB) with the PP2A inhibitor, calyculin-A (CLA) on the OSCC cell line, HSC3, was evaluated in vitro. We found that a combination of 1 nM CLA and 50 µM CXB significantly inhibited cell viability, and migration and invasion of HSC3 cells. Western blots for AKT, p-AKT, ERK, p-ERK, E-cadherin, vimentin and ß-catenin were conducted after treatment with CXB and/or CLA. Increased E-cadherin and decreased ß-catenin expression were found in CXB or CLA treated hsc-3 cells, whereas the combined CXB and CLA treatment showed no difference in E-cadherin or ß-catenin expression. Our findings suggest that CLA alone was more effective than CXB alone, but not in the combined drug treatment.


Assuntos
Carcinoma de Células Escamosas/tratamento farmacológico , Celecoxib/farmacologia , Proliferação de Células/efeitos dos fármacos , Neoplasias Bucais/patologia , Caderinas/efeitos dos fármacos , Caderinas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Neoplasias Bucais/tratamento farmacológico , Carcinoma de Células Escamosas de Cabeça e Pescoço , Vimentina/metabolismo
10.
Endocr J ; 67(4): 379-386, 2020 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-31839623

RESUMO

Rikkunshito, a traditional Japanese herbal medicine, improves appetite via activation of gastrointestinal hormone ghrelin pathway. The function of ghrelin is mediated by growth hormone secretagogue receptor (GHSR1a), and ghrelin has been known to possess diverse physiological functions including growth suppression of some cancer cells. Considering that increased ghrelin signaling by Rikkunshito could enhance sirtuin1 (SIRT1) activity in nervous system, we aimed to investigate the effect of Rikkunshito in ovarian cancer cells. Ovarian cancer cell lines were treated with Rikkunshito, and cellular viability, gene expressions and epithelial-mesenchymal transition (EMT) status were investigated. To investigate the involvement of SIRT1 by Rikkunshito in SKOV3 cancer cells, endogenous expression of SIRT1 was depleted using small interfering RNA (siRNA). Treatment with Rikkunshito elevated ghrelin, GHSR1a and SIRT1, while cellular viability was decreased. The treatment of Rikkunshito also inhibited cellular migration and invasion status in a dose-dependent manner, and these effects were translated to the enhanced EMT status, although the role of SIRT1 was not determined. Our study revealed a novel function of Rikkunshito in enhancing EMT status of ovarian cancer cells. Therefore, we would like to propose that Rikkunshito may be used as a novel adjunctive therapy in chemotherapy of ovarian cancer because platinum-based chemotherapy frequently used for the treatment of ovarian cancer inevitably impairs appetite.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Neoplasias Ovarianas/metabolismo , Receptores de Grelina/efeitos dos fármacos , Sirtuína 1/efeitos dos fármacos , Antígenos CD/efeitos dos fármacos , Antígenos CD/metabolismo , Caderinas/efeitos dos fármacos , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Feminino , Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Grelina/efeitos dos fármacos , Grelina/metabolismo , Humanos , Neoplasias Ovarianas/genética , Receptores de Grelina/genética , Receptores de Grelina/metabolismo , Sirtuína 1/genética , Sirtuína 1/metabolismo , Vimentina/efeitos dos fármacos , Vimentina/metabolismo
11.
World Neurosurg ; 132: e178-e184, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31505280

RESUMO

OBJECTIVE: We aimed to evaluate the combined effect of ellagic acid (EA) and temozolomide (TEM) on the cadherin switch and angiogenesis in the C6 glioma cell line. METHODS: A total of 100 µM EA and 100 µM TEM were applied to rat C6 glioma cells for 24, 48, and 72 hours. Cell proliferation was detected by 5-bromo-2'-deoxyuridine immunohistochemistry. The messenger RNA and protein levels of E-cadherin, N-cadherin, and vascular endothelial growth factor (VEGF) were determined by real-time polymerase chain reaction and their immunohistochemistry, respectively, subsequent to EA treatment combined with TEM. RESULTS: EA in combination with TEM conspicuously reduced the viability of C6 glioma cells at all incubation times (P < 0.001). EA upregulated the expression of E-cadherin at the gene and protein levels in a time-independent manner (P < 0.05 and P < 0.001, respectively). By the presence of TEM, the increase was exaggerated at 24-hour incubation (P < 0.01). Conversely, EA reduced N-cadherin expression and immunoreactivity in a time-independent manner (P < 0.05 and P < 0.001, respectively), and combination with TEM enhanced this effect at the 24th hour (P < 0.001). Combination also downregulated the gene expression (P < 0.001) and immunoreactivity of VEGF only at 72 hours (P < 0.001). CONCLUSIONS: A successful therapeutic efficacy of EA combined with TEM is suggested probably by inhibiting the cadherin switch and angiogenesis.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Caderinas/efeitos dos fármacos , Glioblastoma/patologia , Neovascularização Patológica/patologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Ácido Elágico/farmacologia , Ratos , Temozolomida/farmacologia
12.
Int J Biol Macromol ; 133: 51-57, 2019 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-30981772

RESUMO

Active films based on chitosan incorporated Herba Lophatheri extract (HLE) with different concentrations were developed. Physicochemical properties of the chitosan films incorporated HLE, including density, opacity, moisture content, color, water solubility, swelling, water vapor permeability and oil resistance were measured. Biological activities of the films include antioxidant activity and antimicrobial properties, which were characterized in terms of 2,2-diphenyl-1-picrylhydrazyl (DPPH) free radical scavenging activity and Oxford cup method, respectively. The potential interactions between chitosan and HLE in the films were investigated by Attenuated total reflectance-Fourier transform infrared analysis and X-ray diffraction. The results indicated that the moisture content, water solubility, swelling degree, water vapor permeability and oil absorption rate of chitosan/HLE films decreased up to 14.81%, 38.97%, 48.03%, 69.23% and 80% in comparison with the control chitosan film. The DPPH free radical scavenging activity of chitosan/HLE films increased by nearly 3.5 folds, and the diameter of inhibitory zone of the chitosan/HLE films extract solution against Staphylococcus aureus and Escherichia coli increased 17.02% and 19.28%, respectively. Additionally, the incorporation of HLE caused interactions between chitosan and HLE and gave rise to the chitosan/HLE films more opacity, darker, redness and yellowness appearance. In summary, the addition of HLE enhanced the moisture and oil resistance, antioxidant activity which declined with time, and antimicrobial activity of the chitosan film, which were all desirable for food packaging.


Assuntos
Fenômenos Químicos , Quitosana/química , Quitosana/farmacologia , Extratos Vegetais/química , Poaceae/química , Antibacterianos/química , Antibacterianos/farmacologia , Compostos de Bifenilo/química , Caderinas/efeitos dos fármacos , Cor , Sequestradores de Radicais Livres/química , Sequestradores de Radicais Livres/farmacologia , Fenômenos Ópticos , Permeabilidade , Picratos/química , Solubilidade , Staphylococcus aureus/efeitos dos fármacos , Vapor
13.
Med Sci Monit ; 24: 9488-9496, 2018 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-30594943

RESUMO

BACKGROUND Ginkgo biloba extract EGb 761 is a putative antioxidant and has been used for thousands of years to treat a variety of ailments, including cancer. While it is known that cell behavior can be modulated by long non-coding RNAs (lncRNAs), the contributions of lncRNAs in EGb 761-induced anti-cancer effects are largely unknown. MATERIAL AND METHODS Colon cancer cell lines HT29 and HCT116 were used in this study. RT-qPCR was used to detect the relative expression of lincRNA-p21 in colon cancer cells. Wound-healing assay and Matrigel Transwell assay were performed to investigate the migration and invasion of colon cancer cells. Immunoprecipitation and Western blot experiments were used to verify ubiquitination and the interaction between lincRNA-p21 and E-cadherin, or E-cadherin and b-transducin repeat containing (BTRC) E3 ubiquitin protein ligase. RESULTS Cell function assay verified that treatment with EGb 761 suppressed the migratory and invasive abilities of colon cancer cells in a dose-dependent manner via the suppression of E-cadherin expression level. lincRNA-p21 was upregulated in colon cancer cells after treatment with EGb 761, and knockdown of lincRNA-p21 reversed the EGb 761-induced anti-metastatic effect. Furthermore, lincRNA-p21 was localized in cytoplasm of colon cells and regulated E-cadherin expression at a post-transcriptional level. Specifically, lincRNA-p21 promotes E-cadherin stability by preventing the interaction between BTRC and E-cadherin, which leads to the inhibition of E-cadherin ubiquitination. CONCLUSIONS We demonstrated that lincRNA-p21 mediates the anti-cancer effect of Ginkgo biloba extract EGb 761 by stabilizing E-cadherin protein in colon cancer, which may help define the functional role of EGb 761 in cancer treatment.


Assuntos
Neoplasias do Colo/genética , Extratos Vegetais/uso terapêutico , RNA Longo não Codificante/genética , Antioxidantes , Caderinas/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Ginkgo biloba , Células HT29/efeitos dos fármacos , Humanos , Invasividade Neoplásica/genética , RNA Longo não Codificante/metabolismo
14.
J Am Heart Assoc ; 7(17): e008776, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30371149

RESUMO

Background Dysfunctional endothelium may contribute to the development of cardiovascular complications in chronic kidney disease ( CKD ). Supplementation with active vitamin D has been proposed to have vasoprotective potential in CKD , not only by direct effects on the endothelium but also by an increment of α-Klotho. Here, we explored the capacity of the active vitamin D analogue paricalcitol to protect against uremia-induced endothelial damage and the extent to which this was dependent on increased α-Klotho concentrations. Methods and Results In a combined rat model of CKD with vitamin D deficiency, renal failure induced vascular permeability and endothelial-gap formation in thoracic aorta irrespective of baseline vitamin D, and this was attenuated by paricalcitol. Downregulation of renal and serum α-Klotho was found in the CKD model, which was not restored by paricalcitol. By measuring the real-time changes of the human endothelial barrier function, we found that paricalcitol effectively improved the recovery of endothelial integrity following the addition of the pro-permeability factor thrombin and the induction of a wound. Furthermore, immunofluorescence staining revealed that paricalcitol promoted vascular endothelial-cadherin-based cell-cell junctions and diminished F-actin stress fiber organization, preventing the formation of endothelial intracellular gaps. Conclusions Our results demonstrate that paricalcitol attenuates the CKD -induced endothelial damage in the thoracic aorta and directly mediates endothelial stability in vitro by enforcing cell-cell interactions.


Assuntos
Aorta Torácica/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Ergocalciferóis/farmacologia , Insuficiência Renal Crônica/metabolismo , Uremia/metabolismo , Deficiência de Vitamina D/metabolismo , Actinas/efeitos dos fármacos , Actinas/metabolismo , Animais , Aorta Torácica/metabolismo , Caderinas/efeitos dos fármacos , Caderinas/metabolismo , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Glucuronidase/efeitos dos fármacos , Glucuronidase/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Junções Intercelulares/efeitos dos fármacos , Junções Intercelulares/metabolismo , Proteínas Klotho , Ratos , Fibras de Estresse/efeitos dos fármacos
15.
Stroke ; 49(9): 2211-2219, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30354988

RESUMO

Background and Purpose- tPA (tissue-type plasminogen activator) is the only recommended intravenous thrombolytic agent for ischemic stroke. However, its application is limited because of increased risk of hemorrhagic transformation beyond the time window. T541 is a Chinese compound medicine with potential to attenuate ischemia and reperfusion injury. This study was to explore whether T541-benefited subjects underwent tPA thrombolysis extending the time window. Methods- Male C57BL/6 N mice were subjected to carotid artery thrombosis by stimulation with 10% FeCl3 followed by 10 mg/kg tPA with/without 20 mg/kg T541 intervention at 4.5 hours. Thrombolysis and cerebral blood flow were observed dynamically until 24 hours after drug treatment. Neurological deficit scores, brain edema and hemorrhage, cerebral microvascular junctions and basement membrane proteins, and energy metabolism in cortex were assessed then. An in vitro hypoxia/reoxygenation model using human cerebral microvascular endothelial cells was used to evaluate effect of T541 on tight junctions and F-actin in the presence of tPA. Results- tPA administered at 4.5 hours after carotid thrombosis resulted in a decrease in thrombus area and survival rate, whereas no benefit on cerebral blood flow. Study at 24 hours after tPA administration revealed a significant angioedema and hemorrhage in the ischemia hemisphere, a decreased expression of junction proteins claudin-5, zonula occludens-1, occludin, junctional adhesion molecule-1 and vascular endothelial cadherin, and collagen IV and laminin. Meanwhile, ADP/ATP, AMP/ATP, and ATP5D (ATP synthase subunit) expression and activities of mitochondria complex I, II, and IV declined, whereas malondialdehyde and 8-Oxo-2'-deoxyguanosine increased and F-actin arrangement disordered. All the insults after tPA treatment were attenuated by addition of T541 dose dependently. Conclusions- The results suggest T541 as a potential remedy to attenuate delayed tPA-related angioedema and hemorrhage and extend time window for tPA treatment. The potential of T541 to upregulate energy metabolism and protect blood-brain barrier is likely attributable to its effects observed.


Assuntos
Alcenos/farmacologia , Edema Encefálico , Trombose das Artérias Carótidas , Circulação Cerebrovascular/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Hemorragias Intracranianas , Polifenóis/farmacologia , Traumatismo por Reperfusão , Saponinas/farmacologia , Animais , Antígenos CD/efeitos dos fármacos , Antígenos CD/metabolismo , Astrágalo , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Caderinas/efeitos dos fármacos , Caderinas/metabolismo , Moléculas de Adesão Celular/efeitos dos fármacos , Moléculas de Adesão Celular/metabolismo , Claudina-5/efeitos dos fármacos , Claudina-5/metabolismo , Colágeno Tipo IV/efeitos dos fármacos , Colágeno Tipo IV/metabolismo , Modelos Animais de Doenças , Combinação de Medicamentos , Complexo I de Transporte de Elétrons , Complexo II de Transporte de Elétrons , Complexo IV da Cadeia de Transporte de Elétrons , Laminina/efeitos dos fármacos , Laminina/metabolismo , Masculino , Camundongos , Ocludina/efeitos dos fármacos , Ocludina/metabolismo , Panax notoginseng , Receptores de Superfície Celular/efeitos dos fármacos , Receptores de Superfície Celular/metabolismo , Ativador de Plasminogênio Tecidual/farmacologia , Proteína da Zônula de Oclusão-1/efeitos dos fármacos , Proteína da Zônula de Oclusão-1/metabolismo
16.
Med Sci Monit ; 24: 6405-6413, 2018 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-30208371

RESUMO

BACKGROUND Colorectal cancer is one of the leading causes of death in China, and the development of effective drugs is urgently needed. Here, we report on Paeoniflorin (PF), a product isolated from the roots of the peony plant, as a possible candidate because of its anti-tumor effects on epithelial-to-mesenchymal transition (EMT) of PF in human colorectal cancer (CRC). MATERIAL AND METHODS Cell proliferation, wound healing, and Transwell assays were used to analyze the effects of PF on in vitro cell migration and invasion of HCT116 and SW480, 2 colorectal cancer cell lines. The tumor xenograft model was used to verify the anti-metastasis effects of PF in vivo. The RNA and protein levels of epithelia-cadherin (E-cadherin), Vimentin, and histone deacetylase2 (HDAC2) were measured by qPCR and Western blot analysis to explore the mechanism involved. RESULTS Our results showed that PF inhibited colorectal cancer cell migration and invasion and suppressed the metastatic potential of the cancer cells in vivo. Moreover, PF significantly decreased the expression of HDAC2 and Vimentin, while increasing the expression of E-cadherin. CONCLUSIONS These results suggest that PF inhibits colorectal cancer cell migration and invasion ability and reverses the EMT process, through inhibiting the expression of HDAC2, and then affects the expression level of E-cadherin and Vimentin at the cell level. Our results were also verified in the tumor xenograft model. This indicates that PF may be a candidate for colorectal cancer treatment.


Assuntos
Neoplasias Colorretais/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Glucosídeos/farmacologia , Monoterpenos/farmacologia , Animais , Caderinas/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , China , Neoplasias Colorretais/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Histona Desacetilase 2/efeitos dos fármacos , Humanos , Medicina Tradicional Chinesa , Camundongos , Camundongos Nus , Transdução de Sinais/efeitos dos fármacos , Vimentina/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Braz J Med Biol Res ; 51(10): e7579, 2018 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-30088541

RESUMO

Glucocorticoid insensitivity is an important barrier to the treatment of several inflammatory diseases, including acute lung injury (ALI). Saquinavir (SQV) is an inhibitor of the human immunodeficiency virus protease, and the therapeutic effects of SQV in ALI accompanied with glucocorticoid insensitivity have not been previously investigated. In this study, the effects of SQV on lipopolysaccharide (LPS)-mediated injury in human pulmonary microvascular endothelial cells (HPMECs), human type I alveolar epithelial cells (AT I), and alveolar macrophages were determined. In addition, the effects of SQV on an LPS-induced ALI model with or without methylprednisolone (MPS) were studied. In LPS-stimulated HPMECs, SQV treatment resulted in a decrease of high mobility group box 1 (HMGB1), phospho-NF-κB (p-NF-κB), and toll-like receptor 4 (TLR4), and an increase of VE-cadherin. Compared to MPS alone, MPS plus SQV attenuated the decrease of glucocorticoid receptor alpha (GRα) and IκBα in LPS-stimulated HPMECs. HMGB1, TLR4, and p-NF-κB expression were also lessened in LPS-stimulated alveolar macrophages with SQV treatment. In addition, SQV reduced the injury in human AT I with a decrease of HMGB1 and p-NF-κB, and with an increase of aquaporin 5 (AQP 5). SQV ameliorated the lung injury caused by LPS in rats with reductions in vascular permeability, myeloperoxidase activity (MPO) and histopathological scores, and with lowered HMGB1, TLR4, and p-NF-κB expression, but with enhanced VE-cadherin expression. By comparison, SQV plus MPS increased GRα and IκBα in lung tissues of rats with ALI. This study demonstrated that SQV prevented experimental ALI and improved glucocorticoid insensitivity by modulating the HMGB1/TLR4 pathway.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Metilprednisolona/administração & dosagem , Saquinavir/administração & dosagem , Lesão Pulmonar Aguda/induzido quimicamente , Animais , Antígenos CD/efeitos dos fármacos , Antígenos CD/metabolismo , Caderinas/efeitos dos fármacos , Caderinas/metabolismo , Modelos Animais de Doenças , Proteína HMGB1/efeitos dos fármacos , Proteína HMGB1/metabolismo , Lipopolissacarídeos , Masculino , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo
18.
Exp Cell Res ; 370(2): 613-622, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30031128

RESUMO

Fucosyltransferase 2 (FUT2), the enzyme catalyzing α-1,2-fucosylation in mammals, has been implicated in cancer. The up-regulation of FUT2 has been observed in lung adenocarcinoma (LUAD), and FUT2 can enhance the cell migration and invasion of LUAD cell lines. However, the underlying mechanism of FUT2 in LUAD remains largely unknown. Abundant studies have revealed that epithelial-mesenchymal transition (EMT) played a pivotal role during lung cancer metastasis and progression. In the present study, we showed that knocking down FUT2 in LUAD cell lines increased the expression of E-cadherin and reduced the expression of Vimentin, N-cadherin, TßRII, p-Smad2, p-Smad3 and Snail, which were the makers of EMT. Meanwhile, the expression of E-cadherin was decreased, and the expression of Vimentin was increased by restoring the expression of FUT2 in RNA interference FUT2 (RNAi-FUT2) cells, suggesting that FUT2 enhanced the EMT process in LUAD. Additionally, silencing FUT2 expression can up-regulate E-cadherin and down-regulate Vimentin, significantly attenuated EMT in vivo. Treated with the SIS3, a new-type inhibitor of p-Smad3 of TGF-ß signaling, the expression of E-cadherin, Vimentin and Snail were not affected by RNAi-FUT2 cells, indicating that the effect of FUT2 on EMT depended on TGF-ß/Smad signaling. Overall, the current results indicated that FUT2 might promote LUAD metastasis through the EMT initiated by TGF-ß/Smad signaling. Therefore, FUT2 might be a prognostic factor and therapeutic target for LUAD.


Assuntos
Caderinas/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Fucosiltransferases/farmacologia , Fator de Crescimento Transformador beta/efeitos dos fármacos , Caderinas/metabolismo , Regulação para Baixo/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Regulação para Cima/efeitos dos fármacos , Vimentina/metabolismo
19.
Arthritis Rheumatol ; 70(11): 1879-1889, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29781582

RESUMO

OBJECTIVE: Immune complex (IC) deposition activates polymorphonuclear neutrophils (PMNs), increases vascular permeability, and leads to organ damage in systemic lupus erythematosus and rheumatoid arthritis. The bioactive lipid sphingosine 1-phosphate (S1P), acting via S1P receptor 1 (S1P1 ), is a key regulator of endothelial cell (EC) barrier function. This study was undertaken to investigate whether augmenting EC integrity via S1P1 signaling attenuates inflammatory injury mediated by ICs. METHODS: In vitro barrier function was assessed in human umbilical vein endothelial cells (HUVECs) by electrical cell-substrate impedance sensing. Phosphorylation of myosin light chain 2 (p-MLC-2) and VE-cadherin staining in HUVECs were assessed by immunofluorescence. A reverse Arthus reaction (RAR) was induced in the skin and lungs of mice with S1P1 deleted from ECs (S1P1 EC-knockout [ECKO] mice) and mice treated with S1P1 agonists and antagonists. RESULTS: S1P1 agonists prevented loss of barrier function in HUVECs treated with IC-activated PMNs. S1P1 ECKO and wild-type (WT) mice treated with S1P1 antagonists had amplified RAR, whereas specific S1P1 agonists attenuated skin and lung RAR in WT mice. ApoM-Fc, a novel S1P chaperone, mitigated EC cell barrier dysfunction induced by activated PMNs in vitro and attenuated lung RAR. Expression levels of p-MLC-2 and disruption of VE-cadherin, each representing manifestations of cell contraction and destabilization of adherens junctions, respectively, that were induced by activated PMNs, were markedly reduced by treatment with S1P1 agonists and ApoM-Fc. CONCLUSION: Our findings indicate that S1P1 signaling in ECs modulates vascular responses to IC deposition. S1P1 agonists and ApoM-Fc enhance the EC barrier, limit leukocyte escape from capillaries, and provide protection against inflammatory injury. The S1P/S1P1 axis is a newly identified target to attenuate tissue responses to IC deposition and mitigate end-organ damage.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Permeabilidade Capilar/genética , Células Endoteliais/metabolismo , Receptores de Lisoesfingolipídeo/genética , Junções Aderentes/efeitos dos fármacos , Junções Aderentes/metabolismo , Anilidas/farmacologia , Animais , Antígenos CD/efeitos dos fármacos , Antígenos CD/metabolismo , Apolipoproteínas M/farmacologia , Reação de Arthus , Caderinas/efeitos dos fármacos , Caderinas/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Miosinas Cardíacas/efeitos dos fármacos , Miosinas Cardíacas/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Indanos/farmacologia , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Lisofosfolipídeos/farmacologia , Camundongos , Camundongos Knockout , Cadeias Leves de Miosina/efeitos dos fármacos , Cadeias Leves de Miosina/metabolismo , Organofosfonatos/farmacologia , Oxidiazóis/farmacologia , Receptores de Lisoesfingolipídeo/agonistas , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/metabolismo , Pele/irrigação sanguínea , Pele/efeitos dos fármacos , Pele/metabolismo , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Receptores de Esfingosina-1-Fosfato , Tiofenos/farmacologia
20.
Biotech Histochem ; 93(5): 328-335, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29652183

RESUMO

Diabetes mellitus (DM) affects many organs including kidney. Tyrosine kinase can cause hypoglycemia and sunitinib is an inhibitor of tyrosine kinase. We investigated the possible effects of sunitinib on the kidney of streptozotocin (STZ) induced type 1 diabetic mice. We used 28 CD 1 type male mice divided into four groups of seven. Type 1 diabetes was induced by injection of STZ. Group 1 was the untreated control. Group 2 comprised non-diabetic mice + sunitinib. Both groups 1 and 2 exhibited normal blood glucose levels. Group 3 comprised STZ treated diabetic mice + saline. Group 4 were diabetic mice + sunitinib treatment. Kidneys were removed after 8 weeks. The immunoreactivities of vimentin, E-cadherin and S100 were assessed. Immunostaining of vimentin, E-cadherin and S100 was located in both the glomeruli and tubules of the kidney. We found that the number of vimentin and E-cadherin positive glomeruli and tubules were increased after sunitinib treatment compared to saline treated diabetic mice. The number of vimentin labeled tubules was decreased in the sunitinib treated group compared to diabetic + saline groups. Differences in the number of S100 positive tubules and glomeruli between groups 3 and 4 were not statistically significant. The effect of sunitinib on experimental diabetic mice appears to be related to levels of vimentin, E-cadherin and S100 in the glomeruli and tubules of the kidney, and sunitinib may protect against renal damage from DM.


Assuntos
Caderinas/efeitos dos fármacos , Nefropatias Diabéticas/metabolismo , Proteínas S100/efeitos dos fármacos , Sunitinibe/farmacologia , Vimentina/efeitos dos fármacos , Animais , Caderinas/metabolismo , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Tipo 1/metabolismo , Nefropatias Diabéticas/tratamento farmacológico , Rim/metabolismo , Glomérulos Renais/metabolismo , Masculino , Camundongos , Proteínas S100/metabolismo , Estreptozocina/farmacologia , Vimentina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...