Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Langmuir ; 35(20): 6752-6761, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-31030514

RESUMO

There is a great demand worldwide for bone-related implant materials. The drawbacks of chronic infections and poor bone healing of current implant materials have limited their clinical applications. Functionalizing the implant surfaces with antibacterial and osteogenic films on implant materials provides new opportunities for fabricating novel implant materials. In the present study, an ultrathin (GO/Lys)8 film of several tens of nanometers was fabricated using a layer-by-layer (LBL) technique with alternative deposition of graphene oxide (GO) and lysozyme (Lys). The deposition of the (GO/Lys) n film exhibited a successive growth as supported by ellipsometry, UV-vis, and Fourier transform infrared data, and the physical properties (morphology, roughness, and stiffness) of this film were characterized with an atomic force microscope. The ultrathin films exhibited a great effect on bacterium sterilization of Gram-positive Staphylococcus aureus and Gram-negative Escherichia coli and enhanced osteogenic differentiation efficiency, showing the potential application in bone implant coatings. We believe that this LBL assembling strategy will pave the way for fabricating dual-functional surfaces and guide the design of the implanted surfaces in the future.


Assuntos
Antibacterianos , Polpa Dentária/metabolismo , Escherichia coli/crescimento & desenvolvimento , Grafite , Membranas Artificiais , Osteogênese/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento , Células-Tronco/metabolismo , Antibacterianos/química , Antibacterianos/farmacologia , Polpa Dentária/citologia , Grafite/química , Grafite/farmacologia , Humanos , Muramidase/química , Muramidase/farmacologia , Células-Tronco/citologia
2.
EMBO Mol Med ; 9(11): 1491-1503, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28899928

RESUMO

The currently available antihypertensive agents have undesirable adverse effects due to systemically altering target activity including receptors, channels, and enzymes. These effects, such as loss of potassium ions induced by diuretics, bronchospasm by beta-blockers, constipation by Ca2+ channel blockers, and dry cough by ACEI, lead to non-compliance with therapies (Moser, 1990). Here, based on new hypertension mechanisms, we explored a new antihypertensive approach. We report that transient receptor potential vanilloid 4 (TRPV4) interacts with Ca2+-activated potassium channel 3 (KCa2.3) in endothelial cells (ECs) from small resistance arteries of normotensive humans, while ECs from hypertensive patients show a reduced interaction between TRPV4 and KCa2.3. Murine hypertension models, induced by high-salt diet, N(G)-nitro-l-arginine intake, or angiotensin II delivery, showed decreased TRPV4-KCa2.3 interaction in ECs. Perturbation of the TRPV4-KCa2.3 interaction in mouse ECs by overexpressing full-length KCa2.3 or defective KCa2.3 had hypotensive or hypertensive effects, respectively. Next, we developed a small-molecule drug, JNc-440, which showed affinity for both TRPV4 and KCa2.3. JNc-440 significantly strengthened the TRPV4-KCa2.3 interaction in ECs, enhanced vasodilation, and exerted antihypertensive effects in mice. Importantly, JNc-440 specifically targeted the impaired TRPV4-KCa2.3 interaction in ECs but did not systemically activate TRPV4 and KCa2.3. Together, our data highlight the importance of impaired endothelial TRPV4-KCa2.3 coupling in the progression of hypertension and suggest a novel approach for antihypertensive drug development.


Assuntos
Anti-Hipertensivos/uso terapêutico , Hipertensão/tratamento farmacológico , Canais de Potássio Cálcio-Ativados/metabolismo , Canais de Cátion TRPV/metabolismo , Angiotensina II/genética , Angiotensina II/metabolismo , Animais , Anti-Hipertensivos/química , Pressão Sanguínea , Células Cultivadas , Modelos Animais de Doenças , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Humanos , Hipertensão/metabolismo , Hipertensão/patologia , Artérias Mesentéricas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Mutagênese , Nitroprussiato/farmacologia , Plasmídeos/genética , Plasmídeos/metabolismo , Canais de Potássio Cálcio-Ativados/genética , Canais de Cátion TRPV/deficiência , Canais de Cátion TRPV/genética , Vasodilatação/efeitos dos fármacos
3.
Clin Sci (Lond) ; 131(3): 227-237, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27895148

RESUMO

Transient receptor potential channel 5 (TrpC5) is a member of the TrpC subgroup, and it forms a receptor-activated, non-selective Ca2+ channel. The architecture of the TrpC5 channel is poorly understood. In the present study, we report that TrpC5 is a key factor in regulating differentiation in colorectal cancer (CRC). Through a study of specimens from a large cohort of patients with CRC, we found that TrpC5 was highly expressed and its cellular level correlated with tumour grade. We showed further that up-regulated TrpC5 caused a robust rise in intracellular calcium concentration [Ca2+]i, increased Wnt5a expression and the nuclear translocation of ß-catenin, leading to a reduction in cancer differentiation and an increase in cancer cell stemness. Notably, patients with tumours that expressed high levels of TrpC5 showed significantly poorer disease-free and overall survival. Therefore, our findings suggest that TrpC5 is an independent adverse prognostic factor for death in CRC, reducing differentiation through the Ca2+/Wnt5a signalling pathway.


Assuntos
Sinalização do Cálcio , Carcinoma/metabolismo , Neoplasias Colorretais/metabolismo , Canais de Cátion TRPC/metabolismo , Proteína Wnt-5a/metabolismo , Animais , Células CACO-2 , Carcinoma/diagnóstico , Carcinoma/patologia , Diferenciação Celular , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/patologia , Células HT29 , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Gradação de Tumores , Prognóstico , beta Catenina/metabolismo
4.
Biochem Pharmacol ; 118: 18-30, 2016 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-27520484

RESUMO

It is believed that tumor cells can give rise to endothelial cells and tumor endothelium has a neoplastic origin. Yet, the stimuli and underlying mechanism remain unclear. Here, we demonstrate that adriamycin or paclitaxel, first-line chemotherapy agent, induced breast cancer cells to generate morphological, phenotypical and functional features of endothelial cells in vitro. In xenografts models, challenges from adriamycin or paclitaxel induced cancer cells to generate the majority of microvessels. Importantly, in breast cancer specimens from patients with neoadjuvant anthracycline-based or taxane-based chemotherapy, tumor-derived endothelial microvessels, lined by EGFR-amplified or/and TP53+-CD31+ endothelial cells, was significantly higher in patients with progressive or stable disease (PD/SD) than in those with a partial or complete response (PR/CR). Further, exposure to the Notch signaling inhibitor and gene silencing studies showed that Notch signaling inhibition or silencing Nothc4/Dll3 decreased endothelial markers and function of tumor-derived endothelial cells under chemotherapy treatment, which may be through VEGFR3. Thus, our findings demonstrate that chemotherapy induces functional tumor-derived endothelial microvessels by mediating Notch signaling and VEGF signaling, and may provide new targets for anti-angiogenesis therapy in breast cancer.


Assuntos
Antineoplásicos/efeitos adversos , Neoplasias da Mama/tratamento farmacológico , Endotélio Vascular/efeitos dos fármacos , Proteínas de Neoplasias/agonistas , Neovascularização Patológica/induzido quimicamente , Receptores Notch/agonistas , Transdução de Sinais/efeitos dos fármacos , Animais , Antraciclinas/efeitos adversos , Antraciclinas/uso terapêutico , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidade , Biomarcadores/metabolismo , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Hidrocarbonetos Aromáticos com Pontes/efeitos adversos , Hidrocarbonetos Aromáticos com Pontes/uso terapêutico , Doxorrubicina/efeitos adversos , Doxorrubicina/uso terapêutico , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Feminino , Humanos , Células MCF-7 , Camundongos Nus , Microvasos/efeitos dos fármacos , Microvasos/metabolismo , Microvasos/patologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Paclitaxel/efeitos adversos , Paclitaxel/uso terapêutico , Interferência de RNA , Receptores Notch/antagonistas & inibidores , Receptores Notch/genética , Receptores Notch/metabolismo , Organismos Livres de Patógenos Específicos , Taxoides/efeitos adversos , Taxoides/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Acta Pharmacol Sin ; 37(9): 1199-207, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27397542

RESUMO

AIM: TRPV4-C1 heteromeric channels contribute to store-operated Ca(2+) entry in vascular endothelial cells. However, the negative regulation of these channels is not fully understood. This study was conducted to investigate the inhibitory effect of PKG1α on TRPV4-C1 heteromeric channels. METHODS: Immuno-fluorescence resonance energy transfer (FRET) was used to explore the spatial proximity of PKG1α and TRPC1. Phosphorylation of endogenous TRPC1 was tested by phosphorylation assay. [Ca(2+)]i transients and cation current in MAECs were assessed with Fura-2 fluorescence and whole-cell recording, respectively. In addition, rat mesenteric arteries segments were prepared, and vascular relaxation was examined with wire myography. RESULTS: In immuno-FRET experiments, after exposure of these cells to 8-Br-cGMP, more PKG1α was observed in the plasma membrane, and PKG1α and TRPC1 were observed to be in closer proximity. TAT-TRPC1(S172) and TAT-TRPC1(T313) peptide fragments, which contain the PKG targeted residues Ser172 and Thr313, respectively, were introduced into isolated endothelial cells to abrogate the translocation of PKG1α. Furthermore, a phosphorylation assay demonstrated that PKG directly phosphorylates TRPC1 at Ser172 and Thr313 in endothelial cells. In addition, PKG activator 8-Br-cGMP markedly reduced the magnitude of the 4αPDD-induced and 11,12-EET-induced [Ca(2+)]i transients, the cation current and vascular relaxation. CONCLUSION: This study uncovers a novel mechanism by which PKG negatively regulates endothelial heteromeric TRPV4-C1 channels through increasing the spatial proximity of TRPV4-C1 to PKG1α via translocation and through phosphorylating Ser172 and Thr313 of TRPC1.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Proteína Quinase Dependente de GMP Cíclico Tipo I/metabolismo , Endotélio Vascular/metabolismo , Artérias Mesentéricas/metabolismo , Canais de Cátion TRPC/metabolismo , Animais , Técnicas de Cultura de Células , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Células Endoteliais/metabolismo , Transferência Ressonante de Energia de Fluorescência , Técnicas In Vitro , Potenciais da Membrana/efeitos dos fármacos , Camundongos Knockout , Técnicas de Patch-Clamp , Fragmentos de Peptídeos/farmacologia , Fosforilação , Transporte Proteico , Ratos Sprague-Dawley , Canais de Cátion TRPC/genética , Vasodilatação
6.
Pharmacol Res ; 93: 36-42, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25579062

RESUMO

Chemotherapy targeting anti-angiogenesis in tumors may have insufficient efficacy, but little is known about the underlying mechanisms. Here, we showed that the Ca(2+)-permeable channel, TrpC5, is highly expressed in human breast cancer after long-term chemotherapy drug-treatment. It mediates downstream hypoxia-inducible factor 1α accumulation in the nucleus, and then activates the transcription of vascular endothelial growth factor which promotes tumor angiogenesis, leading to a poor chemotherapeutic outcome. We verified this mechanism at both the cellular and xenograft levels. Moreover, in samples from patients, high TrpC5 expression was correlated with enhanced tumor vasculature after chemotherapy. Taken together, our research demonstrated the essential role of TrpC5 in tumor angiogenesis when facing the challenge of chemotherapy and presents a new potential target for overcoming the high vasculature of human breast cancer after chemotherapy.


Assuntos
Neoplasias da Mama/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Canais de Cátion TRPC/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Antibióticos Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Feminino , Humanos , Camundongos Nus , Neovascularização Patológica/metabolismo , RNA Interferente Pequeno/administração & dosagem , Canais de Cátion TRPC/genética , Ensaios Antitumorais Modelo de Xenoenxerto
7.
J Biol Chem ; 290(1): 448-56, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25404731

RESUMO

5-Fluorouracil (5-Fu) is commonly used in the chemotherapy of colorectal cancer (CRC), but resistance to 5-Fu occurs in most cases, allowing cancer progression. Suppressing ABCB1 (ATP-binding cassette, subfamily B, member 1), which is a pump overproduced in cancer cells to export cytotoxic drugs, is an attractive strategy to overcome drug resistance. In the present study, transient receptor potential channel TrpC5 was found to be overproduced at the mRNA and protein levels together with ABCB1 in 5-Fu-resistant human CRC HCT-8 (HCT-8/5-Fu) and LoVo (LoVo/5-Fu) cells. More nuclear-stabilized ß-catenin accumulation was found in HCT-8/5-Fu and LoVo/5-Fu cells than in HCT-8 and LoVo cells. Suppressing TrpC5 expression with TrpC5-specific siRNA inhibited the canonical Wnt/ß-catenin signal pathway, reduced the induction of ABCB1, weakened the ABCB1 efflux pump, and caused a remarkable reversal of 5-Fu resistance in HCT-8/5-Fu and LoVo/5-Fu cells. On the contrary, enforcing TrpC5 expression resulted in an activated Wnt/ß-catenin signal pathway and up-regulation of ABCB1. Taken together, we demonstrated an essential role of TrpC5 in ABCB1 induction and drug resistance in human CRC cells via promoting nuclear ß-catenin accumulation.


Assuntos
Antineoplásicos/farmacologia , Fluoruracila/farmacologia , Regulação Neoplásica da Expressão Gênica , Canais de Cátion TRPC/genética , beta Catenina/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Linhagem Celular Tumoral , Colo/efeitos dos fármacos , Colo/metabolismo , Colo/patologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo
8.
FEBS Lett ; 588(11): 2009-15, 2014 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-24786471

RESUMO

To investigate the role of microRNAs in the development of chemoresistance and related epithelial-mesenchymal transition (EMT), we examined the effect of miR-489 in adriamycin (ADM)-resistant human breast cancer cells (MCF-7/ADM). MiR-489 was significantly suppressed in MCF-7/ADM cells compared with chemosensitive parental control MCF-7/WT cells. Forced-expression of miR-489 reversed chemoresistance. Furthermore, Smad3 was identified as the target of miR-489 and is highly expressed in MCF-7/ADM cells. Forced expression of miR-489 both inhibited Smad3 expression and Smad3 related EMT properties. Finally, the interactions between Smad3, miR-489 and EMT were confirmed in chemoresistant tumor xenografts and clinical samples, indicating their potential implication for treatment of chemoresistance.


Assuntos
Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal , MicroRNAs/fisiologia , Regiões 3' não Traduzidas , Animais , Antraciclinas/farmacologia , Antraciclinas/uso terapêutico , Antibióticos Antineoplásicos/farmacologia , Antibióticos Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Quimioterapia Adjuvante , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Concentração Inibidora 50 , Células MCF-7 , Camundongos , Camundongos Nus , Terapia Neoadjuvante , Interferência de RNA , Proteína Smad3/genética , Proteína Smad3/metabolismo , Taxoides/farmacologia , Taxoides/uso terapêutico , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Proc Natl Acad Sci U S A ; 111(17): 6389-94, 2014 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-24733904

RESUMO

A critical challenge for chemotherapy is the development of chemoresistance in breast cancer. However, the underlying mechanisms and validated predictors remain unclear. Extracellular vesicles (EVs) have gained attention as potential means for cancer cells to share intracellular contents. In adriamycin-resistant human breast cancer cells (MCF-7/ADM), we analyzed the role of transient receptor potential channel 5 (TrpC5) in EV formation and transfer as well as the diagnostic implications. Up-regulated TrpC5, accumulated in EVs, is responsible for EV formation and trapping of adriamycin (ADM) in EVs. EV-mediated intercellular transfer of TrpC5 allowed recipient cells to acquire TrpC5, consequently stimulating multidrug efflux transporter P-glycoprotein production through a Ca(2+)- and activated T-cells isoform c3-mediated mechanism and thus, conferring chemoresistance on nonresistant cells. TrpC5-containing circulating EVs were detected in nude mice bearing MCF-7/ADM tumor xenografts, and the level was lower after TrpC5-siRNA treatment. In breast cancer patients who underwent chemotherapy, TrpC5 expression in the tumor was significantly higher in patients with progressive or stable disease than in patients with a partial or complete response. TrpC5-containing circulating EVs were found in peripheral blood from patients who underwent chemotherapy but not patients without chemotherapy. Taken together, we found that TrpC5-containing circulating EVs may transfer chemoresistance property to nonchemoresistant recipient cells. It may be worthwhile to further explore the potential of using TrpC5-containing EVs as a diagnostic biomarker for chemoresistant breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Vesículas Citoplasmáticas/metabolismo , Resistencia a Medicamentos Antineoplásicos , Canais de Cátion TRPC/metabolismo , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/ultraestrutura , Linhagem Celular Tumoral , Vesículas Citoplasmáticas/efeitos dos fármacos , Vesículas Citoplasmáticas/ultraestrutura , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Humanos , Camundongos , Camundongos Nus , Canais de Cátion TRPC/sangue , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Biochem Biophys Res Commun ; 446(1): 85-90, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24582564

RESUMO

Treatment of carcinoma commonly fails due to chemoresistance. Studies have shown that endothelial cells acquire resistance via the tumor microenvironment. Microvesicle (MV) shedding from the cell membrane to the microenvironment plays an important role in communication between cells. The aim of the present study was to determine whether MCF-7 adriamycin-resistant cells (MCF-7/ADM) shed MVs that alter the characteristics of human microvessel endothelial cells (HMECs). MVs from tumor cells transferred a Ca(2+)-permeable channel TrpC5 to HMECs, inducing the expression of P-glycoprotein (P-gp) by activation of the transcription factor NFATc3 (nuclear factor of activated T cells isoform c3). Expression of the mdr1 gene was blocked by the TrpC5-blocking antibody T5E3, and the production of P-gp in HMECs was reduced by blockade of TrpC5. Thus, we postulate that endothelial cells acquire the resistant protein upon exposure to TrpC5-containg MVs in the microenvironment, and express P-gp in the TrpC5-NFATc3 signal pathway.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Fatores de Transcrição NFATC/metabolismo , Canais de Cátion TRPC/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Antineoplásicos/farmacologia , Neoplasias da Mama/irrigação sanguínea , Comunicação Celular , Micropartículas Derivadas de Células/metabolismo , Células Cultivadas , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Células Endoteliais/metabolismo , Feminino , Expressão Gênica , Xenoenxertos , Humanos , Células MCF-7 , Camundongos , Camundongos Nus , Transdução de Sinais , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...