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.
J Cachexia Sarcopenia Muscle ; 14(4): 1737-1752, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37212018

RESUMO

BACKGROUND: It has been observed that Slo1 knockout mice have reduced motor function, and people with certain Slo1 mutations have movement problems, but there is no answer whether the movement disorder is caused by the loss of Slo1 in the nervous system, or skeletal muscle, or both. Here, to ascertain in which tissues Slo1 functions to regulate motor function and offer deeper insight in treating related movement disorder, we generated skeletal muscle-specific Slo1 knockout mice, studied the functional changes in Slo1-deficient skeletal muscle and explored the underlying mechanism. METHODS: We used skeletal muscle-specific Slo1 knockout mice (Myf5-Cre; Slo1flox/flox mice, called CKO) as in vivo models to examine the role of Slo1 in muscle growth and muscle regeneration. The forelimb grip strength test was used to assess skeletal muscle function and treadmill exhaustion test was used to test whole-body endurance. Mouse primary myoblasts derived from CKO (myoblast/CKO) mice were used to extend the findings to in vitro effects on myoblast differentiation and fusion. Quantitative real-time PCR, western blot and immunofluorescence approaches were used to analyse Slo1 expression during myoblast differentiation and muscle regeneration. To investigate the involvement of genes in the regulation of muscle dysfunction induced by Slo1 deletion, RNA-seq analysis was performed in primary myoblasts. Immunoprecipitation and mass spectrometry were used to identify the protein interacting with Slo1. A dual-luciferase reporter assay was used to identify whether Slo1 deletion affects NFAT activity. RESULTS: We found that the body weight and size of CKO mice were not significantly different from those of Slo1flox/flox mice (called WT). Deficiency of Slo1 in muscles leads to reduced endurance (~30% reduction, P < 0.05) and strength (~30% reduction, P < 0.001). Although there was no difference in the general morphology of the muscles, electron microscopy revealed a considerable reduction in the content of mitochondria in the soleus muscle (~40% reduction, P < 0.01). We found that Slo1 was expressed mainly on the cell membrane and showed higher expression in slow-twitch fibres. Slo1 protein expression is progressively reduced during muscle postnatal development and regeneration after injury, and the expression is strongly reduced during myoblast differentiation. Slo1 deletion impaired myoblast differentiation and slow-twitch fibre formation. Mechanistically, RNA-seq analysis showed that Slo1 influences the expression of genes related to myogenic differentiation and slow-twitch fibre formation. Slo1 interacts with FAK to influence myogenic differentiation, and Slo1 deletion diminishes NFAT activity. CONCLUSIONS: Our data reveal that Slo1 deficiency impaired skeletal muscle regeneration and slow-twitch fibre formation.


Assuntos
Transtornos dos Movimentos , Músculo Esquelético , Animais , Camundongos , Diferenciação Celular/genética , Camundongos Knockout , Transtornos dos Movimentos/metabolismo , Músculo Esquelético/metabolismo , Mioblastos/metabolismo
2.
Commun Biol ; 5(1): 1151, 2022 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-36310238

RESUMO

Paraquat (PQ) is an efficient herbicide but leads to high mortality with no antidote in mammals. PQ produces reactive oxygen species (ROS), leading to epithelial-mesenchymal transition (EMT) for pulmonary fibrosis in type II alveolar (AT II) cells. Intriguingly, strategies reducing ROS exhibit limited therapeutic effects, indicating other targets existing for PQ toxicity. Herein we report that PQ is also an agonist for STIM1 that increases intracellular calcium levels. Particularly, PQ promotes STIM1 puncta formation and association with TRPC1 or ORAI for extracellular calcium entry and thus intracellular calcium influx. Further studies reveal the importance of P584&Y586 residues in STIM1 for PQ association that facilitates STIM1 binding to TRPC1. Consequently, the STIM1-TRPC1 route facilitates PQ-induced EMT for pulmonary fibrosis as well as cell death. Our results demonstrate that PQ is an agonist of STIM1 that induces extracellular calcium entry, increases intracellular calcium levels, and thus promotes EMT in AT II cells.


Assuntos
Paraquat , Fibrose Pulmonar , Animais , Paraquat/farmacologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Cálcio , Espécies Reativas de Oxigênio , Transição Epitelial-Mesenquimal , Mamíferos
3.
Toxicol Res (Camb) ; 10(4): 733-741, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34484664

RESUMO

Paraquat (PQ) and diquat (DQ), two highly efficient herbicides sharing similar chemical backbone, both induce reactive oxygen species and are highly toxic to humans and livestock, however, PQ but not DQ poisoning result in pulmonary fibrosis, the leading cause of high mortality rate in patients suffering PQ toxicity. Understanding the unique mechanism of PQ different from DQ therefore would provide potential strategies to reduce PQ-induced pulmonary fibrosis. Here, we identified that PQ but not DQ continuously upregulates TGF-ß expression in alveolar type II (AT II) cells. Importantly, such high expression of TGF-ß increases cytosolic calcium levels and further promotes the activation of calcineurin-NFAT axis. TGF-ß mainly activates NFATc1 and NFATc2, but not NFATc3 or NFATc4. Administration of the inhibitors targeting cytosolic calcium or calcineurin largely reverses PQ-induced epithelial-mesenchymal transition (EMT), whereas DQ has little effects on activation of NFAT and EMT. Ultimately, PQ poisoned patients exhibit significantly reduced blood calcium levels compared to DQ poisoning, possibly via the large usage of calcium by AT II cells. All in all, we found a vicious cycle that the upregulated TGF-ß in PQ-induced EMT further aggravates EMT via promotion of the calcium-calcineurin axis, which could be potential drug targets for treating PQ-induced pulmonary fibrosis.

4.
Drug Metab Dispos ; 48(11): 1129-1136, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32878767

RESUMO

Cytochrome P450 2J2 (CYP2J2) enzyme attracts more attention because it not only metabolizes clinical drugs but also mediates the biotransformation of important endogenous substances and the regulation of physiologic function. Although CYP2J2 is very important, few animal models are available to study its function in vivo In particular, a CYP2J gene knockout (KO) rat model for drug metabolism and pharmacokinetics is not available. In this report, the CRISPR/Cas9 technology was used to delete rat CYP2J3/10, the orthologous genes of CYP2J2 in humans. The CYP2J3/10 KO rats were viable and fertile and showed no off-target effect. Compared with wild-type (WT) rats, the mRNA and protein expression of CYP2J3/10 in liver, small intestine, and heart of KO rats were completely absent. At the same time, CYP2J4 mRNA expression and protein expression were significantly decreased in these tissues. Further in vitro and in vivo metabolic studies of astemizole, a typical substrate of CYP2J, indicated that CYP2J was functionally inactive in KO rats. The heart function indexes of WT and KO rats were also measured and compared. The myocardial enzymes, including creatine kinase-muscle brain type (CK-MB), creatine kinase (CK), and CK-MB/CK ratio, of KO rats increased by nearly 140%, 80%, and 60%, respectively. In conclusion, this study successfully developed a new CYP2J3/10 KO rat model, which is a useful tool to study the function of CYP2J in drug metabolism and cardiovascular disease. SIGNIFICANCE STATEMENT: Human CYP2J2 is involved not only in clinical drug metabolism but also in the biotransformation of important endogenous substances. Therefore, it is very important to construct new animal models to study its function in vivo. This study successfully developed a new CYP2J knockout rat model by using CRISPR/Cas9 technology. This rat model provides a useful tool to study the role of CYP2J in drug metabolism and diseases.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Animais , Astemizol/farmacocinética , Biotransformação , Sistemas CRISPR-Cas/genética , Citocromo P-450 CYP2J2 , Sistema Enzimático do Citocromo P-450/genética , Avaliação Pré-Clínica de Medicamentos/métodos , Estudos de Viabilidade , Feminino , Técnicas de Silenciamento de Genes , Masculino , Modelos Animais , Ratos , Ratos Transgênicos
5.
Acta Pharm Sin B ; 10(5): 850-860, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32528832

RESUMO

Organic anion transporting polypeptide 1B1 and 1B3 (OATP1B1/3) as important uptake transporters play a fundamental role in the transportation of exogenous drugs and endogenous substances into cells. Rat OATP1B2, encoded by the Slco1b2 gene, is homologous to human OATP1B1/3. Although OATP1B1/3 is very important, few animal models can be used to study its properties. In this report, we successfully constructed the Slco1b2 knockout (KO) rat model via using the CRISPR/Cas9 technology for the first time. The novel rat model showed the absence of OATP1B2 protein expression, with no off-target effects as well as compensatory regulation of other transporters. Further pharmacokinetic study of pitavastatin, a typical substrate of OATP1B2, confirmed the OATP1B2 function was absent. Since bilirubin and bile acids are the substrates of OATP1B2, the contents of total bilirubin, direct bilirubin, indirect bilirubin, and total bile acids in serum are significantly higher in Slco1b2 KO rats than the data of wild-type rats. These results are consistent with the symptoms caused by the absence of OATP1B1/3 in Rotor syndrome. Therefore, this rat model is not only a powerful tool for the study of OATP1B2-mediated drug transportation, but also a good disease model to study hyperbilirubinemia-related diseases.

6.
Front Oncol ; 10: 293, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32211326

RESUMO

Cancers have been considered as one of the most severe health problems in the world. Efforts to elucidate the cancer progression reveal the importance of bone metastasis for tumor malignancy, one of the leading causes for high mortality rate. Multiple cancers develop bone metastasis, from which breast cancers exhibit the highest rate and have been well-recognized. Numerous cells and environmental factors have been believed to synergistically facilitate bone metastasis in breast cancers, from which breast cancer cells, osteoclasts, osteoblasts, and their produced cytokines have been well-recognized to form a vicious cycle that aggravates tumor malignancy. Except the cytokines or chemokines, calcium ions are another element largely released from bones during bone metastasis that leads to hypercalcemia, however, have not been well-characterized yet in modulation of bone metastasis. Calcium ions act as a type of unique second messenger that exhibits omnipotent functions in numerous cells, including tumor cells, osteoclasts, and osteoblasts. Calcium ions cannot be produced in the cells and are dynamically fluxed among extracellular calcium pools, intracellular calcium storages and cytosolic calcium signals, namely calcium homeostasis, raising a possibility that calcium ions released from bone during bone metastasis would further enhance bone metastasis and aggravate tumor progression via the vicious cycle due to abnormal calcium homeostasis in breast cancer cells, osteoclasts and osteoblasts. TRPs, VGCCs, SOCE, and P2Xs are four major calcium channels/routes mediating extracellular calcium entry and affect calcium homeostasis. Here we will summarize the overall functions of these four calcium channels in breast cancer cells, osteoclasts and osteoblasts, providing evidence of calcium homeostasis as a vicious cycle in modulation of bone metastasis in breast cancers.

7.
Biochem Pharmacol ; 169: 113612, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31437461

RESUMO

Paclitaxel, a broad-spectrum antitumor drug, is widely used as a cytotoxic drug, while sorafenib as a multi-kinase inhibitor is a classic targeted drug. A number of clinical trials have combined paclitaxel and sorafenib for cancer treatment, with the expectation of better therapeutic effects. However, the toxicity and side effects in the treatment are significantly increased. In this report, the organic anion transport polypeptide 1b2 (Oatp1b2) overexpression cell model and the Oatp1b2 knockout (KO) rat model were used to investigate the drug-drug interactions (DDI) between paclitaxel and sorafenib. In Oatp1b2-overexpressed cells, sorafenib inhibited the uptake of paclitaxel in a concentration-dependent manner. In wild-type (WT) rats, sorafenib increased the systemic exposure and slowed the elimination of paclitaxel, resulting in DDI. In Oatp1b2 KO rats, however, the DDI disappeared. Interestingly, paclitaxel did not alter the pharmacokinetic profiles of sorafenib. Further studies found that sorafenib was not the substrate of Oatp1b2 in rats. In general, the combination of paclitaxel and sorafenib caused Oatp1b2-mediated DDI in vitro and in vivo, because sorafenib inhibited Oatp1b2 activity and affected the pharmacokinetic properties of paclitaxel. This study may provide useful information for understanding the role of OATP1B in paclitaxel-sorafenib interaction.


Assuntos
Paclitaxel/farmacocinética , Membro 1B3 da Família de Transportadores de Ânion Orgânico Carreador de Soluto/fisiologia , Sorafenibe/farmacologia , Animais , Interações Medicamentosas , Células HEK293 , Humanos , Masculino , Ratos , Sorafenibe/farmacocinética
8.
Biochem Pharmacol ; 164: 368-376, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31063713

RESUMO

Obesity increases the incidences of metabolic syndrome, including type 2 diabete, fatty liver, dyslipidemia, hyperglycemia, heart disease, hypertension and cancer. In particular, pharmacokinetics and pharmacodynamics of many drugs have changed in obese patients. However, little is known about the hepatic drug-metabolizing enzymes and transporters that are influenced by diet-induced obese. In this report, we established obesity and fatty liver models in male rats by high-fat diet. The expression profiles of drug-metabolizing enzymes and transporters were studied by quantitative real-timePCR and Western blotting analysis. The function of these enzymes and transporters were assessed by their substrates and cocktail methods. The expression and activity of phase I enzymes (CYP1A2, CYP2B1, CYP2C11, CYP3A1, CYP4A1 and FMO1) and phase II enzymes (UGT1A1, UGT1A3, UGT1A6, UGT1A9, UGT2B7, NAT1 and GSTT1) were decreased in the liver of obese rats. In addition, the mRNA levels of hepatic transporter Slco1a2, Slco1b2, Slc22a5, Abcc2, Abcc3, Abcb1a and Abcg2 decreased significantly in obese animals, while Abcb1b increased significantly. Furthermore, the decreased expression of hepatic phase I and II enzymes and transporter may be due to changes of Hnf4α, LXRα and FXR. In conclusion, the diet-induced obese altered the expression and function of hepatic drug-metabolizing enzymes and transporters in male rats, thereby impacting drug metabolism and pharmacokinetics.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Dieta Hiperlipídica/efeitos adversos , Fígado/enzimologia , Obesidade/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Animais , Masculino , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/patologia , Obesidade/etiologia , Obesidade/patologia , Ratos , Ratos Sprague-Dawley
9.
Fitoterapia ; 136: 104161, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31048010

RESUMO

Ailanthone (AIL) has many biological activities including antimalarial, antiviral and anticancer. Our previous study also found that AIL targets p23 against castration-resistant prostate cancer. In this report, the preclinical safety of AIL was evaluated by acute toxicity, subacute toxicity and toxicokinetics in mice. In the acute toxicity study, the LD50 of AIL was 27.3 mg/kg, and severe pathological damages were mainly found in the liver and gastrointestinal tract. In the subacute toxicity study, mice were orally administered at doses of 2.5, 5 and 10 mg/kg for 28 days. The results showed the body weight of male mice in the 10 mg/kg dose group decreased, but that of female mice increased. Biochemical and histopathological analysis showed that AIL could cause steatohepatitis, splenomegaly, gastrointestinal mucosal damage and reproductive system abnormalities. In addition, AIL presented the reversible hematotoxicity. To determine the relationship between AIL toxicity and dose/exposure in vivo, toxicokinetics of AIL were carried out after a single oral dose of 15 mg/kg. The stomach was identified as the main target organ, followed by the intestine and kidney. On the basis of this study, the dose of 2.5 mg/kg had no adverse effect on mice. To sum up, this study is the first time to evaluate the systemic toxicity of AIL, which is useful for the further development of AIL.


Assuntos
Antineoplásicos Fitogênicos/toxicidade , Fígado/efeitos dos fármacos , Neoplasias de Próstata Resistentes à Castração , Quassinas/toxicidade , Animais , Antineoplásicos Fitogênicos/farmacologia , Feminino , Masculino , Camundongos , Quassinas/farmacologia , Testes de Toxicidade Aguda , Testes de Toxicidade Subaguda , Toxicocinética
10.
Drug Metab Dispos ; 47(2): 71-79, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30478157

RESUMO

Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein-9 nuclease (Cas9) technology is widely used as a tool for gene editing in rat genome site-specific engineering. Multidrug resistance 1 [MDR1 (also known as P-glycoprotein)] is a key efflux transporter that plays an important role not only in the transport of endogenous and exogenous substances, but also in tumor MDR. In this report, a novel MDR1 (Mdr1a/b) double-knockout (KO) rat model was generated by the CRISPR/Cas9 system without any off-target effect detected. Western blot results showed that MDR1 was completely absent in the liver, small intestine, brain, and kidney of KO rats. Further pharmacokinetic studies of digoxin, a typical substrate of MDR1, confirmed the deficiency of MDR1 in vivo. To determine the possible compensatory mechanism of Mdr1a/b (-/-) rats, the mRNA levels of the CYP3A subfamily and transporter-related genes were compared in the brain, liver, kidney, and small intestine of KO and wild-type rats. In general, a new Mdr1a/b (-/-) rat model has been successfully generated and characterized. This rat model is a useful tool for studying the function of MDR1 in drug absorption, tumor MDR, and drug target validation.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Digoxina/farmacocinética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Administração Oral , Animais , Encéfalo/metabolismo , Sistemas CRISPR-Cas/genética , Citocromo P-450 CYP3A/análise , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Digoxina/administração & dosagem , Feminino , Técnicas de Inativação de Genes/métodos , Intestino Delgado/metabolismo , Rim/metabolismo , Fígado/metabolismo , Masculino , Modelos Animais , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...