Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 542
Filtrar
1.
Cell Physiol Biochem ; 58(2): 172-181, 2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38643508

RESUMO

BACKGROUND/AIMS: Extracellular acidic conditions impair cellular activities; however, some cancer cells drive cellular signaling to adapt to the acidic environment. It remains unclear how ovarian cancer cells sense changes in extracellular pH. This study was aimed at characterizing acid-inducible currents in an ovarian cancer cell line and evaluating the involvement of these currents in cell viability. METHODS: The biophysical and pharmacological properties of membrane currents in OV2944, a mouse ovarian cancer cell line, were studied using the whole-cell configuration of the patch-clamp technique. Viability of this cell type in acidic medium was evaluated using the MTT assay. RESULTS: OV2944 had significant acid-sensitive outwardly rectifying (ASOR) Cl- currents at a pH50 of 5.3. The ASOR current was blocked by pregnenolone sulfate (PS), a steroid ion channel modulator that blocks the ASOR channel as one of its targets. The viability of the cells was reduced after exposure to an acidic medium (pH 5.3) but was slightly restored upon PS administration. CONCLUSION: These results offer first evidence for the presence of ASOR Cl- channel in ovarian cancer cells and indicate its involvement in cell viability under acidic environment.


Assuntos
Sobrevivência Celular , Neoplasias Ovarianas , Pregnenolona , Animais , Feminino , Camundongos , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Linhagem Celular Tumoral , Pregnenolona/farmacologia , Concentração de Íons de Hidrogênio , Sobrevivência Celular/efeitos dos fármacos , Canais de Cloreto/metabolismo , Canais de Cloreto/antagonistas & inibidores , Técnicas de Patch-Clamp , Potenciais da Membrana/efeitos dos fármacos
2.
Anticancer Res ; 42(3): 1313-1325, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35220222

RESUMO

BACKGROUND/AIM: Chloride intracellular channel protein 1 (CLIC1) is known as a promoter of cancer progression, metastasis, and angiogenesis. Thus, CLIC1 could be a future therapeutic target. This study aimed to evaluate the effect of anti-CLIC1 antibodies on tumour cells and vessels of human renal cell carcinoma (RCC) in rabbit cornea and chick embryo chorioallantoic membrane (CAM) models. MATERIALS AND METHODS: Human cc-RCC xenografts on rabbit cornea and CAM surface were performed. Anti-CLIC1 antibodies were applied for 5 consecutive days on both tumor models. We comparatively evaluated treated and untreated tumors by combining ultrasonography with microscopic techniques. RESULTS: RCC implants rapidly recruited blood vessels and had an exponential growth rate on both tumor models. Anti-CLIC1 antibodies suppressed tumor growth by inducing tumor cell necrosis. Tumor vessels regressed rapidly but not completely during anti-CLIC1 antibodies based therapy. CONCLUSION: Anti-CLIC1 antibodies induced tumor necrosis and tumor vasculature regression in human cc-RCC xenografts in both in vivo experimental models.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos Imunológicos/farmacologia , Carcinoma de Células Renais/irrigação sanguínea , Carcinoma de Células Renais/tratamento farmacológico , Canais de Cloreto/antagonistas & inibidores , Neoplasias Renais/irrigação sanguínea , Neoplasias Renais/tratamento farmacológico , Neovascularização Patológica , Animais , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Proliferação de Células/efeitos dos fármacos , Embrião de Galinha , Canais de Cloreto/metabolismo , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Masculino , Terapia de Alvo Molecular , Necrose , Coelhos , Transdução de Sinais , Fatores de Tempo , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Oxid Med Cell Longev ; 2021: 3672112, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34777684

RESUMO

Transmembrane protein 206 (TMEM206), a proton-activated chloride channel, has been implicated in various biochemical processes, including bone metabolism, and has emerged as a novel cancer-related protein in multiple tumor types. However, its role in primary malignant bone tumors, particularly in osteosarcoma (OS), remains unclear. This study is aimed at exploring the effects of TMEM206 gene silencing on the proliferation, migration, invasion, and metastasis of human OS cells in vitro and in vivo using an shRNA-knockdown strategy. We found that TMEM206 is frequently overexpressed and that high levels of TMEM206 correlated with clinical stage and pulmonary metastasis in patients with OS. We provided evidence that TMEM206-silenced OS cancer cells exhibit decreased proliferation, migration, and invasion in vitro. Mechanistically, we identified ß-catenin, a key member of Wnt/ß-catenin signaling, as a downstream effector of TMEM206. TMEM206 silencing inhibits the Wnt/ß-catenin signaling pathway in expression rescue experiments, confirming that TMEM206 silencing attenuates OS cell tumorigenic behavior, at least in part, via the ß-catenin mediated downregulation of Wnt/ß-catenin signaling. More importantly, TMEM206 knockdown-related phenotype changes were replicated in a xenograft nude mouse model where pulmonary metastases of OS cells were suppressed. Together, our results demonstrate that silencing TMEM206 negatively modulates the Wnt/ß-catenin signaling pathway via ß-catenin to suppress proliferation, migration, invasion, and metastasis in OS carcinogenesis, suggesting TMEM206 as a potential oncogenic biomarker and a potential target for OS treatment.


Assuntos
Neoplasias Ósseas/prevenção & controle , Canais de Cloreto/antagonistas & inibidores , Inativação Gênica , Osteossarcoma/prevenção & controle , Via de Sinalização Wnt , Animais , Apoptose , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Proliferação de Células , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Osteossarcoma/genética , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Biomed Pharmacother ; 141: 111860, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34246954

RESUMO

EGFR tyrosine kinase inhibitors (TKIs) are mainly used to treat non-small cell lung cancer; however, adverse effects such as severe diarrhea represent a major obstacle towards the continuation of EGFR-TKIs therapy. Chloride channels, which control the fluid flow in the intestinal lumen, are proposed as an important target to remediate EGFR-TKIs-induced diarrhea, but the mechanism remains unclear. The aim of this study was to clarify the mechanism underlying EGFR-TKIs-induced diarrhea with a particular focus on the role of intestinal chloride channels. Here, we show that osimertinib-treated rats exhibit diarrhea and an increase in fecal water content without showing any severe histopathological changes. This diarrhea was attenuated by intraperitoneal treatment with the calcium-activated chloride channel (CaCC) inhibitor CaCCinh-A01. These findings were confirmed in afatinib-treated rats with diarrhea. Moreover, treatment with the Japanese traditional herbal medicine, hangeshashinto (HST), decreased fecal water content and improved fecal appearance in rats treated with EGFR-TKIs. HST inhibited the ionomycin-induced CaCC activation in HEK293 cells in patch-clamp current experiments and its active ingredients were identified. In conclusion, secretory diarrhea induced by treatment with EGFR-TKIs might be partially mediated by the activation of CaCC. Therefore, blocking the CaCC could be a potential new treatment for EGFR-TKI-induced diarrhea.


Assuntos
Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/metabolismo , Diarreia/induzido quimicamente , Receptores ErbB/antagonistas & inibidores , Inibidores de Proteínas Quinases/toxicidade , Acrilamidas/toxicidade , Afatinib/toxicidade , Compostos de Anilina/toxicidade , Animais , Diarreia/patologia , Fezes/química , Células HEK293 , Humanos , Masculino , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Tiofenos/farmacologia , Água/química
5.
Placenta ; 110: 46-55, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34120018

RESUMO

INTRODUCTION: Placental oxidative stress features in pregnancy pathologies but in clinical trials antioxidant supplementation has not improved outcomes. N-acetylcysteine (NAC) stimulates glutathione production and is proposed as a therapeutic agent in pregnancy. However, key elements of N-acetylcysteine biology, including its cellular uptake mechanism, remains unclear. This study explores how the cystine/glutamate transporter xCT may mediate N-acetylcysteine uptake and how N-acetylcysteine alters placental redox status. METHODS: The involvement of xCT in NAC uptake by the human placenta was studied in perfused placenta and Xenopus oocytes. The effect of short-term N-acetylcysteine exposure on the placental villous proteome was determined using LC-MS. The effect of N-acetylcysteine on Maxi-chloride channel activity was investigated in perfused placenta, villous fragments and cell culture. RESULTS: Maternoplacental N-acetylcysteine administration stimulated intracellular glutamate efflux suggesting a role of the exchange transporter xCT, which was localised to the microvillous membrane of the placental syncytiotrophoblast. Placental exposure to a bolus of N-acetylcysteine inhibited subsequent activation of the redox sensitive Maxi-chloride channel independently of glutathione synthesis. Stable isotope quantitative proteomics of placental villi treated with N-acetylcysteine demonstrated changes in pathways associated with oxidative stress, apoptosis and the acute phase response. DISCUSSION: This study suggests that xCT mediates N-acetylcysteine uptake into the placenta and that N-acetylcysteine treatment of placental tissue alters the placental proteome while regulating the redox sensitive Maxi-chloride channel. Interestingly N-acetylcysteine had antioxidant effects independent of the glutathione pathway. Effective placental antioxidant therapy in pregnancy may require maintaining the balance between normalising redox status without inhibiting physiological redox signalling.


Assuntos
Acetilcisteína/farmacologia , Sistema y+ de Transporte de Aminoácidos/genética , Canais de Cloreto/antagonistas & inibidores , Placenta , Acetilcisteína/metabolismo , Sistema y+ de Transporte de Aminoácidos/metabolismo , Animais , Canais de Cloreto/metabolismo , Vilosidades Coriônicas/efeitos dos fármacos , Vilosidades Coriônicas/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Ácido Glutâmico/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Células HEK293 , Humanos , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Placenta/efeitos dos fármacos , Placenta/metabolismo , Gravidez , Proteoma/efeitos dos fármacos , Proteoma/metabolismo , Xenopus laevis
6.
Biochem Biophys Res Commun ; 555: 95-101, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-33813282

RESUMO

Oral squamous cell carcinoma (OSCC) is a highly aggressive carcinoma with a high incidence of recurrence and distant metastasis. However, the mechanism of epithelial to mesenchymal transition (EMT) during tumor progression and metastasis in OSCC has not yet been fully elucidated. It is well known that the Cl- channel controls cell volume and activates several signaling pathways for cell differentiation. The aim of the present study was to investigate the role of the Cl- channel on EMT in the OSC 20 cell line, which is an OSCC line. OSC-20 cells were cultured with low serum medium containing a Cl- channel blocker NPPB. Morphological changes, gene expression, immunoreactivity, cell volume, and signaling pathway of the NPPB-treated OSC-20 cells were evaluated. The NPPB-treated OSC-20 cells showed typical morphology of mesenchymal cells. The expression levels of the epithelial marker E-cadherin in the NPPB-treated OSC-20 cells were lower than those of the untreated and TGF-ß1-treated OSC-20 cells. On the other hand, mesenchymal markers such as vimentin, ZEB1, and Snail, in the NPPB-treated OSC-20 cells were higher than those in the untreated and TGF-ß1-treated OSC-20 cells. Furthermore, a large number of vimentin-positive cells also appeared in the NPPB-treated OSC-20 cells. Additionally, the cell volume of these cells was significantly increased compared to that of the untreated and TGF-ß1-treated cells. Interestingly, NPPB did not activate the TGF-ß/smad signaling pathway, but activated the Wnt/ß-catenin signaling pathway. These results suggest that Cl- channel dysfunction promoted EMT via activation of the Wnt/ß-catenin signaling pathway in OSCC.


Assuntos
Carcinoma de Células Escamosas/patologia , Canais de Cloreto/metabolismo , Transição Epitelial-Mesenquimal , Neoplasias Bucais/patologia , Antracenos/farmacologia , Antígenos CD/metabolismo , Caderinas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Tamanho Celular/efeitos dos fármacos , Canais de Cloreto/antagonistas & inibidores , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/fisiologia , Humanos , Neoplasias Bucais/metabolismo , Nitrobenzoatos/farmacologia , Tripsina/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos
7.
Int J Mol Med ; 47(4)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33604681

RESUMO

Cataracts have a high incidence and prevalence rate worldwide, and they are the leading cause of blindness. Lens epithelial cell (LEC) apoptosis is often analysed in cataract research since it is the pathological basis of cataracts, except for congenital cataract. Chloride channels are present in ocular tissues, such as in trabecular cells, LECs and other cells. They serve an important role in apoptosis and participate in endoplasmic reticulum (ER) stress and oxidative stress. However, their role in the apoptosis of LECs has not been discussed. The present study examined the effects of the chloride channel blocker 5­nitro­2­(3­phenylpropylamino) benzoic acid (NPPB) in human LECs (HLECs) to elucidate the role of NPPB in HLECs and investigate the role and mechanism of chloride channels in cataract formation. HLECs were exposed to NPPB. Cell survival rate was evaluated using Cell Counting Kit­8 assays. Oxidative stress was detected as reactive oxygen species (ROS) in cells by using a ROS assay kit. Apoptosis was examined by assessing mitochondrial membrane potential and using a JC­1 assay kit, and western blot analysis was performed to measure the expression levels of mitochondrial­dependent apoptosis pathway­associated proteins. ER stress was evaluated by determining the intracellular calcium ion fluorescence intensity, and western blot analysis was performed to measure ER stress­associated protein expression. The results revealed that NPPB treatment decreased the viability of HLECs and increased apoptosis. Additionally, NPPB increased intracellular ROS levels, as well as the number of JC­1 monomers and the protein expression levels of B­cell lymphoma­2 (Bcl­2)­associated X and cleaved caspase­3, and decreased Bcl­2 protein expression. NPPB increased intracellular calcium ions, the protein expression levels of activating transcription factor 6, JNK, C/EBP homologous protein and caspase­12, and the phosphorylation of protein kinase R­like endoplasmic reticulum kinase. N­acetylcysteine and 4­phenylbutyric acid inhibited NPPB­induced oxidative stress, ER stress and apoptosis. Therefore, NPPB treatment decreased cell viability and promoted apoptosis of HLECs via the promotion of oxidative and ER stress.


Assuntos
Apoptose/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Cristalino/efeitos dos fármacos , Nitrobenzoatos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Caspase 3/metabolismo , Catarata/tratamento farmacológico , Catarata/patologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Canais de Cloreto/antagonistas & inibidores , Retículo Endoplasmático/metabolismo , Humanos , Cristalino/citologia , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
8.
Cell Biochem Biophys ; 79(2): 239-252, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33432550

RESUMO

Chloride intracellular channel 1 (CLIC1) is a sensor of oxidative stress in endothelial cells (EC). However, the mechanism by which CLIC1 mediate the regulation of endothelial dysfunction has not been established. In this study, overexpressed CLIC1 impaired the ability of the vascular cells to resist oxidative damage and promoted cellular senescence. Besides, suppressed CLIC1 protected against cellular senescence and dysfunction in Human Umbilical Vein Endothelial Cells (HUVECs) through the Nrf2/HO-1 pathway. We also found that ROS-activated CLIC1-induced oxidative stress in HUVECs. Nrf2 nuclear translocation was inhibited by CLIC1 overexpression, but was enhanced by IAA94 (CLICs inhibitor) treatment or knockdown of CLIC1. The Nrf2/HO-1 pathway plays a critical role in the anti-oxidative effect of suppressing CLIC1. And inhibition of CLIC1 decreases oxidative stress injury by downregulating the levels of ROS, MDA, and the expression of EC effectors (ICAM1 and VCAM1) protein expression and promotes the activity of superoxide dismutase (SOD). The AMPK-mediated signaling pathway activates Nrf2 through Nrf2 phosphorylation and nuclear translocation, which is also regulated by CLIC1. Moreover, the activation of CLIC1 contributes to H2O2-induced mitochondrial dysfunction and activation of mitochondrial fission. Therefore, elucidation of the mechanisms by which CLIC1 is involved in these pivotal pathways may uncover its therapeutic potential in alleviating ECs oxidative stress and age-related cardiovascular disease development.


Assuntos
Senescência Celular , Canais de Cloreto/metabolismo , Transdução de Sinais , Acetilcisteína/farmacologia , Senescência Celular/efeitos dos fármacos , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/genética , Heme Oxigenase-1/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Peróxido de Hidrogênio/farmacologia , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Dinâmica Mitocondrial/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo
9.
Eur J Pharmacol ; 895: 173881, 2021 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-33476655

RESUMO

OBJECTIVES: Volume-regulated anion channels (VRACs), expressed in various cells, play an important role in cell volume regulation. Despite being physiologically defined almost half a century ago, only the molecular candidates of VRAC, TMEM16A, LRRC8A, and bestrophin-1 (BEST1), are known. Here, we aimed to explore the functional significance of VRAC in, HST-1, an oral squamous cell carcinoma (OSCC) cell line. METHODS: Cell proliferation assays, RT-PCR, Western blot, and flow cytometry were used to estimate changes in gene expression and cell proliferation. Ion channel activity was recorded using the patch-clamp technique. Specific genes were knocked-down by siRNA assays. RESULTS: VRAC, identified as a hypotonicity-induced current, was highly functional and associated with the proliferation of HST-1 cells but not of HaCaT (a normal keratinocyte) cells. The pharmacological profile of VRAC in HST-1 was similar to that reported previously. DCPIB, a specific VRAC inhibitor, completely inhibited VRAC and proliferation of HST-1 cells, eventually leading to apoptosis. VRAC in HST-1 was attenuated by the knockdown of TMEM16A and LRRC8A, while knockdown of BEST1 affected cell proliferation. In situ proximity ligation assay showed that TMEM16A and LRRC8A co-localized under isotonic conditions (300 mOsM) but were separated under hypotonic conditions (250 mOsM) on the plasma membrane. CONCLUSIONS: We have found that VRAC acts to regulate the proliferation of human metastatic OSCC cells and the composition of VRAC may involve in the interactions between TMEM16A and LRRC8A in HST-1 cells.


Assuntos
Anoctamina-1/metabolismo , Proliferação de Células , Canais de Cloreto/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Neoplasias da Língua/metabolismo , Anoctamina-1/antagonistas & inibidores , Anoctamina-1/genética , Antineoplásicos/farmacologia , Apoptose , Bestrofinas/genética , Bestrofinas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/genética , Ciclopentanos/farmacologia , Regulação Neoplásica da Expressão Gênica , Humanos , Indanos/farmacologia , Ativação do Canal Iônico , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Ligação Proteica , Transdução de Sinais , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/secundário , Neoplasias da Língua/tratamento farmacológico , Neoplasias da Língua/genética , Neoplasias da Língua/patologia
10.
Biochem Biophys Res Commun ; 533(4): 1240-1246, 2020 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-33069359

RESUMO

PURPOSE: In the present study, we attempted to explore the role of chloride channel 3 (CLC-3) in colorectal cancer (CRC) and its related mechanism. METHODS: First, the expression level of CLC-3 in CRC tumor tissues and cell lines were measured by RT-qPCR, immunohistochemistry or western blot analysis. CLC-3 expression knockdown in CRC cells was achieved by siRNA transfection. The effect of CLC-3 silence on cell viability, cell cycle, invasion and migration of CRC was estimated by CCK8, flow cytometry based cell cycle assay, and transwell assay, respectively. In order to investigate whether Wnt/ß-catenin signaling was perturbed by CLC-3 knockdown, CLC-3 knockdown cells were treated with pathway activator LiCl, followed by the measurement of the expressions of pathway related genes, cell viability, cell cycle, metastasis ability. RESULTS: The expression of CLC-3 was gradually increased from normal adjacent tissues to CRC tumor tissues, and the increase in tumor tissues was related to TNM stages. CLC-3 was overexpressed in four CRC cell lines (HCT116, SW480, LoVo and SW620), compared with NCM460 cells. CLC-3 knockdown significantly reduced cell proliferation, invasion and migration ability, reflected by declined cell viability, arrested G0/G1 cell cycle, decreased invasion and migration ability. In contrast, the declined cell proliferation, invasion and migration of LoVo and SW620 cells induced by CLC-3 knockdown were reversed by the addition of Wnt/ß-catenin activator LiCl. CONCLUSION: CLC-3 contributed to the CRC development and metastasis through Wnt/ß-catenin signaling pathway. CLC-3 could be proposed as the candidate target for CRC treatment.


Assuntos
Canais de Cloreto/fisiologia , Neoplasias Colorretais/metabolismo , Via de Sinalização Wnt , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Técnicas de Silenciamento de Genes , Humanos , Metástase Neoplásica , beta Catenina/metabolismo
11.
Chem Commun (Camb) ; 56(91): 14137-14153, 2020 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-33057487

RESUMO

The use of synthetic ion transporters for alteration of the concentration of ions across cell membranes has drawn attention from scientists over the last two decades. This ion transport property has been sensibly used to reduce the viability of cancer cells mainly due to the disruption of their ion homeostasis, leading to the perturbation of their abnormal pH gradient. The use of the proanionophore strategy has been recently adopted to increase cellular deliverability and reduce unwanted cytotoxicity towards normal cells. Meanwhile, various anionophores exhibiting non-toxic behavior in epithelial cells have shown a great propensity to be used as a putative treatment for channelopathies like cystic fibrosis. Anionophores with Cl- ion transport mediated antibacterial activities have also been successfully used against clinically relevant bacterial strains, many of which are tolerant towards commercial antibiotics. Recent developments in the chloride ion transport mediated biological activities of anionophores have been mainly discussed in this article. It also highlights other aspects, such as design criteria, targeted delivery options, and others, which can be useful for the further development of selective anionophores for specific biological applications.


Assuntos
Antibacterianos/farmacologia , Produtos Biológicos/farmacologia , Canais de Cloreto/antagonistas & inibidores , Antibacterianos/química , Produtos Biológicos/química , Canais de Cloreto/metabolismo , Humanos , Estrutura Molecular
12.
Basic Clin Pharmacol Toxicol ; 127(4): 329-337, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32628359

RESUMO

SCO-101 (Endovion) was discontinued 20 years ago as a new drug under development against sickle cell anaemia. Data from the phase 1 studies remained unpublished. New data indicate that SCO-101 might be efficacious as add-on therapy in cancer. Thus, we report the results from the four phase 1 trials performed between 2001 and 2002. Adult volunteers received SCO-101 or placebo in four independent trials. Adverse events were recorded, and SCO-101 was determined for pharmacokinetic analysis. Ninety-two volunteers completed the trials. The most remarkable adverse effect was a transient and dose-dependent increase in unconjugated bilirubin. Plasma SCO-101 elimination was approximately log linear, with apparent oral clearances of between 315 and 2103 mL/h for single doses, and between 121 and 2433 mL/h at steady state following oral administration. There was a marked decrease in clearance with increasing dose, and for repeated dose versus single dose. Tmax was greater, and Cmax and AUC∞ were lower in the fed state compared to the fasted state. Exposure was equivalent in males and females and for African Americans and Caucasians. In conclusion, SCO-101 appears to be a safe drug with a predictable PK profile. Its efficacy as add-on to standard anticancer drugs has yet to be defined.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Canais de Cloreto/antagonistas & inibidores , Administração Oral , Adulto , Antineoplásicos/sangue , Área Sob a Curva , Estudos de Coortes , Método Duplo-Cego , Feminino , Voluntários Saudáveis , Humanos , Masculino , Farmacocinética
13.
Biosci Biotechnol Biochem ; 84(10): 2096-2103, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32657644

RESUMO

Macrophage foam cell formation and inflammation are a pathological hallmark of atherosclerosis. ClC-2 has been implicated in various pathological processes, including inflammation and lipid metabolic disorder. However, the functional role of ClC-2 in macrophage foam cell formation and inflammation is unclear. Here, we found that ClC-2 was dominantly expressed in macrophages of atherosclerotic plaque and increased in atherogenesis. Knockdown of ClC-2 inhibited ox-LDL -induced lipid uptake and deposition in macrophages. The increase in CD36 expression and the decrease in ABCA1 expression induced by ox-LDL were alleviated by ClC-2 downregulation. Further, ClC-2 lacking limited the ox-LDL-induced secretion of inflammatory cytokines and chemokine, and suppressed Nlrp3 inflammasome activation. Restoration of Nlrp3 expression reversed the effect of ClC-2 downregulation on macrophage lipid accumulation and inflammation. Collectively, our study demonstrates that ClC-2 knockdown ameliorates ox-LDL-induced macrophage foam cell formation and inflammation by inhibiting Nlrp3 inflammasome activation.


Assuntos
Canais de Cloreto/antagonistas & inibidores , Inflamassomos/metabolismo , Macrófagos/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Canais de Cloro CLC-2 , Canais de Cloreto/deficiência , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Colesterol/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Placa Aterosclerótica/metabolismo , Células RAW 264.7
14.
J Neurochem ; 155(3): 274-284, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32248535

RESUMO

Excitatory α7 neuronal nicotinic receptors (nAChR) are widely expressed in the central and peripheral nervous and immune systems and are important for learning, memory, and immune response regulation. Specific α7 nAChR ligands, including positive allosteric modulators are promising to treat cognitive disorders, inflammatory processes, and pain. One of them, PNU-120596, highly increased the neuron response to α7 agonists and retarded desensitization, showing selectivity for α7 as compared to heteromeric nAChRs, but was not examined at the inhibitory ligand-gated channels. We studied PNU-120596 action on anion-conducting channels using voltage-clamp techniques: it slightly potentiated the response of human glycine receptors expressed in PC12 cells, of rat GABAA receptors in cerebellar Purkinje cells and mouse GABAA Rs heterologously expressed in Xenopus oocytes. On the contrary, PNU-120596 exerted an inhibitory effect on the receptors mediating anion currents in Lymnaea stagnalis neurons: two nAChR subtypes, GABA and glutamate receptors. Acceleration of the current decay, contrary to slowing down desensitization in mammalian α7 nAChR, was observed in L. stagnalis neurons predominantly expressing one of the two nAChR subtypes. Thus, PNU-120596 effect on these anion-selective nAChRs was just opposite to the action on the mammalian cation-selective α7 nAChRs. A comparison of PNU-120596 molecule docked to the models of transmembrane domains of the human α7 AChR and two subunits of L. stagnalis nAChR demonstrated some differences in contacts with the amino acid residues important for PNU-120596 action on the α7 nAChR. Thus, our results show that PNU-120596 action depends on a particular subtype of these Cys-loop receptors.


Assuntos
Canais de Cloreto/metabolismo , Isoxazóis/farmacologia , Canais Iônicos de Abertura Ativada por Ligante/metabolismo , Compostos de Fenilureia/farmacologia , Receptor Nicotínico de Acetilcolina alfa7/agonistas , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Regulação Alostérica/efeitos dos fármacos , Regulação Alostérica/fisiologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/genética , Feminino , Humanos , Canais Iônicos de Abertura Ativada por Ligante/antagonistas & inibidores , Canais Iônicos de Abertura Ativada por Ligante/genética , Lymnaea , Células PC12 , Ratos , Ratos Wistar , Xenopus laevis , Receptor Nicotínico de Acetilcolina alfa7/genética
15.
Am J Physiol Renal Physiol ; 318(4): F870-F877, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31984792

RESUMO

Adenosine plays an important role in various aspects of kidney physiology, but the specific targets and mechanisms of actions are not completely understood. The collecting duct has the highest expression of adenosine receptors, particularly adenosine A1 receptors (A1Rs). Interstitial adenosine levels are greatly increased up to a micromolar range in response to dietary salt loading. We have previously shown that the basolateral membrane of principal cells has primarily K+ conductance mediated by Kir4.1/5.1 channels to mediate K+ recycling and to set up a favorable driving force for Na+/K+ exchange (47). Intercalated cells express the Cl- ClC-K2/b channel mediating transcellular Cl- reabsorption. Using patch-clamp electrophysiology in freshly isolated mouse collecting ducts, we found that acute application of adenosine reversely inhibits ClC-K2/b open probability from 0.31 ± 0.04 to 0.17 ± 0.06 and to 0.10 ± 0.05 for 1 and 10 µM, respectively. In contrast, adenosine (10 µM) had no measureable effect on Kir4.1/5.1 channel activity in principal cells. The inhibitory effect of adenosine on ClC-K2/b was abolished in the presence of the A1R blocker 8-cyclopentyl-1,3-dipropylxanthine (10 µM). Consistently, application of the A1R agonist N6-cyclohexyladenosine (1 µM) recapitulated the inhibitory action of adenosine on ClC-K2/b open probability. The effects of adenosine signaling in the collecting duct were independent from its purinergic counterpartner, ATP, having no measurable actions on ClC-K2/b and Kir4.1/5.1. Overall, we demonstrated that adenosine selectively inhibits ClC-K2/b activity in intercalated cells by targeting A1Rs. We propose that inhibition of transcellular Cl- reabsorption in the collecting duct by adenosine would aid in augmenting NaCl excretion during high salt intake.


Assuntos
Agonistas do Receptor A1 de Adenosina/farmacologia , Adenosina/farmacologia , Proteínas de Transporte de Ânions/antagonistas & inibidores , Canais de Cloreto/antagonistas & inibidores , Cloretos/metabolismo , Túbulos Renais Coletores/efeitos dos fármacos , Receptor A1 de Adenosina/efeitos dos fármacos , Reabsorção Renal/efeitos dos fármacos , Animais , Proteínas de Transporte de Ânions/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Canais de Cloreto/metabolismo , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Receptor A1 de Adenosina/metabolismo
16.
Carcinogenesis ; 41(6): 841-849, 2020 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-31560739

RESUMO

Chloride intracellular channel protein 4 (CLIC4) has been implicated in different types of cancers, but the role of CLIC4 in the development of gastric cancer (GC) remains unknown. We analyzed the expression of CLIC4 in 102 pairs of gastric adenocarcinomas by western blot and real-time PCR. Our data revealed that the expression of CLIC4 is reduced in GC tumor tissues compared with adjacent normal tissues. The expression levels of CLIC4 correlate inversely with the clinical stage of GC. CLIC4 expression is lowest in MKN45 cells, which have the highest tumorigenic potential and express the highest levels of cancer stem cell markers CD44 and OCT4, compared with N87 and AGS cells. Exogenous overexpression of CLIC4 downregulated the expression of CD44 and OCT4, and inhibited migration, invasion and epithelial-mesenchymal transition (EMT). Moreover, anchorage-independent growth of GC cells was decreased and the cells became more sensitive to 5-fluorouracil and etoposide treatment when CLIC4 was overexpressed. The ability of N87 cells to form tumors in nude mice was enhanced when CLIC4 was silenced. We, for the first time, demonstrate that CLIC4 suppresses tumor growth by inhibiting cancer cell stemness and EMT.


Assuntos
Biomarcadores Tumorais/metabolismo , Canais de Cloreto/antagonistas & inibidores , Transição Epitelial-Mesenquimal , Células-Tronco Neoplásicas/patologia , Neoplasias Gástricas/patologia , Animais , Antimetabólitos Antineoplásicos/farmacologia , Apoptose , Biomarcadores Tumorais/genética , Movimento Celular , Proliferação de Células , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Feminino , Fluoruracila/farmacologia , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Prognóstico , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Naunyn Schmiedebergs Arch Pharmacol ; 393(3): 501-510, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31659404

RESUMO

5-Nitro-2-(3-phenylpropylamino)-benzoic acid (NPPB) is a non-specific chloride channel blocker. Peritoneal adhesion is an inevitable complication of abdominal surgery and remains an important clinical problem, leading to chronic pain, intestinal obstruction, and female infertility. The aim of this study is to observe the effects of NPPB on peritoneal adhesions and uncover the underlying mechanism. The formation of postoperative peritoneal adhesions was induced by mechanical injury to the peritoneum of rats. MTT assay and wound-healing assay were used to evaluate proliferation and migration of primary cultured adhesion fibroblasts (AFB) respectively. Whole-cell chloride currents were measured using a fully automated patch-clamp workstation. Cell volume changes were monitored by light microscopy and video imaging. Our results demonstrated that NPPB could significantly prevent the formation of peritoneal adhesion in rats and inhibit the proliferation of AFB in a concentration-dependent manner. NPPB also reduced the migration of AFB cells with an IC50 of 53.09 µM. A 47% hypotonic solution successfully activated the ICl,vol in AFB cells. The current could be blocked by extracellular treatment with NPPB. Moreover, 100 µM NPPB almost completely eliminated the capacity of regulatory volume decrease (RVD) in these cells. These data indicate that NPPB could prevent the formation of postoperative peritoneal adhesions. The possible mechanism may be through the inhibition of the proliferation and migration of AFB cells by modulating ICl,vol and cell volume. These results suggest a potential clinical use of NPPB for preventing the formation of peritoneal adhesions.


Assuntos
Movimento Celular/efeitos dos fármacos , Canais de Cloreto/antagonistas & inibidores , Nitrobenzoatos/uso terapêutico , Peritônio/efeitos dos fármacos , Complicações Pós-Operatórias/tratamento farmacológico , Aderências Teciduais/tratamento farmacológico , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Animais , Movimento Celular/fisiologia , Células Cultivadas , Canais de Cloreto/fisiologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Nitrobenzoatos/farmacologia , Peritônio/fisiopatologia , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/fisiopatologia , Ratos , Ratos Sprague-Dawley , Aderências Teciduais/etiologia , Aderências Teciduais/fisiopatologia
18.
Fitoterapia ; 139: 104367, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31629045

RESUMO

Ca2+-activated Cl- channels (CaCCs) wildly exist in many tissues which play an important role in ion transport and excitation conduction, especially fluid secretion and smooth muscle contraction in epithelial tissues. TMEM16A as a classic CaCC expresses in the intestine, and has become a potential target of intestinal physiological and pathological researches and therapeutic drug screening. In this study, we identified trans-δ-viniferin (TVN), a resveratrol dimmer, could inhibit TMEM16A activity in TMEM16A expressed FRT cells with IC50 of 19.7 µM, it also prevented Ca2+-activated Cl- current in HT-29 cells with IC50 of 4.65 µM and in colonic mucosa. In the mechanism studies, TVN showed no significant inhibition on CFTR and basal Na+/K+-ATPase in both intestinal epithelial cells and colonic tissues, except for inhibition of calcium concentration and Ca2+-activated K+ channel to some degree. In anti-diarrheal studies, TVN could effectively prevent diarrhea caused by rotavirus infection and reduce the pellet number in IBS-D mice. These physiological effects are at least partially attributed to the inhibitory effect of TVN on CaCC-mediated intestinal fluid secretion and the reduction of smooth muscle contraction force by inhibiting TMEM16A. Collectively, the present study identified a new pharmacological target of TVN which provided the theoretical basis for the application of TVN in the treatment of rotavirus-infected diarrhea and IBS-D.


Assuntos
Benzofuranos/farmacologia , Canais de Cloreto/antagonistas & inibidores , Diarreia/tratamento farmacológico , Células Epiteliais/efeitos dos fármacos , Resorcinóis/farmacologia , Estilbenos/farmacologia , Animais , Cálcio/análise , Diarreia/virologia , Motilidade Gastrointestinal/efeitos dos fármacos , Células HT29 , Humanos , Doenças Inflamatórias Intestinais/tratamento farmacológico , Mucosa Intestinal/citologia , Camundongos , Camundongos Endogâmicos C57BL , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Ratos , Rotavirus
19.
J Pharmacol Sci ; 140(3): 273-283, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31444000

RESUMO

Secretory diarrhea is one of the most common causes of death world-wide especially in children under 5 years old. Isoliquiritigenin (ISLQ), a plant-derived chalcone, has previously been shown to exert anti-secretory action in vitro and in vivo by inhibiting CFTR Cl- channels. However, its CFTR inhibition potency is considerably low (IC50 > 10 µM) with unknown mechanism of action. This study aimed to identify novel chalcone derivatives with improved potency and explore their mechanism of action. Screening of 27 chalcone derivatives identified CHAL-025 as the most potent chalcone analog that reversibly inhibited CFTR-mediated Cl- secretion in T84 cells with an IC50 of ∼1.5 µM. As analyzed by electrophysiological and biochemical analyses, the mechanism of CFTR inhibition by CHAL-025 is through AMP-activated protein kinase (AMPK), a negative regulator of CFTR activity. Furthermore, Western blot analyses and molecular dynamics (MD) results suggest that CHAL-025 activates AMPK by binding at the allosteric site of an upstream kinase calcium/calmodulin-dependent protein kinase kinase ß (CaMKKß). Interestingly, CHAL-025 inhibited both cholera toxin (CT) and bile acid-induced Cl- secretion in T84 cells and prevented CT-induced intestinal fluid secretion in mice. Therefore, CHAL-025 represents a promising anti-diarrheal agent that inhibits CFTR Cl- channel activity via CaMKKß-AMPK pathways.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Chalconas/farmacologia , Canais de Cloreto/antagonistas & inibidores , Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Diarreia/tratamento farmacológico , Animais , Transporte Biológico/efeitos dos fármacos , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Linhagem Celular , Cloretos/metabolismo , Diarreia/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Secreções Intestinais/efeitos dos fármacos , Secreções Intestinais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR
20.
Exp Mol Med ; 51(7): 1-11, 2019 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-31316050

RESUMO

Chloride intracellular channel 1 (CLIC1) is a promising therapeutic target in cancer due to its intrinsic characteristics; it is overexpressed in specific tumor types and its localization changes from cytosolic to surface membrane depending on activities and cell cycle progression. Ca2+ and reactive oxygen species (ROS) are critical signaling molecules that modulate diverse cellular functions, including cell death. In this study, we investigated the function of CLIC1 in Ca2+ and ROS signaling in A549 human lung cancer cells. Depletion of CLIC1 via shRNAs in A549 cells increased DNA double-strand breaks both under control conditions and under treatment with the putative anticancer agent chelerythrine, accompanied by a concomitant increase in the p-JNK level. CLIC1 knockdown greatly increased basal ROS levels, an effect prevented by BAPTA-AM, an intracellular calcium chelator. Intracellular Ca2+ measurements clearly showed that CLIC1 knockdown significantly increased chelerythrine-induced Ca2+ signaling as well as the basal Ca2+ level in A549 cells compared to these levels in control cells. Suppression of extracellular Ca2+ restored the basal Ca2+ level in CLIC1-knockdown A549 cells relative to that in control cells, implying that CLIC1 regulates [Ca2+]i through Ca2+ entry across the plasma membrane. Consistent with this finding, the L-type Ca2+ channel (LTCC) blocker nifedipine reduced the basal Ca2+ level in CLIC1 knockdown cells to that in control cells. Taken together, our results demonstrate that CLIC1 knockdown induces an increase in the intracellular Ca2+ level via LTCC, which then triggers excessive ROS production and consequent JNK activation. Thus, CLIC1 is a key regulator of Ca2+ signaling in the control of cancer cell survival.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Canais de Cloreto/antagonistas & inibidores , Neoplasias Pulmonares/tratamento farmacológico , Espécies Reativas de Oxigênio/antagonistas & inibidores , Células A549 , Morte Celular , Membrana Celular/metabolismo , Canais de Cloreto/metabolismo , Quebras de DNA de Cadeia Dupla , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/metabolismo , RNA Interferente Pequeno/genética , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA