Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Avian Med Surg ; 36(1): 92-97, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35526170

Assuntos
Animais
2.
J Cell Mol Med ; 26(12): 3364-3377, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35488446

RESUMO

Transcriptome profiling of tubulointerstitial tissue in glomerulonephritis may reveal a potential tubulointerstitial injury-related biomarker. We profiled manually microdissected tubulointerstitial tissue from biopsy cores of 65 glomerulonephritis cases, including 43 patients with IgA nephropathy, 3 with diabetes mellitus nephropathy, 3 with focal segmental glomerulosclerosis, 3 with lupus nephritis, 4 with membranous nephropathy and 9 with minimal change disease, and additional 22 nephrectomy controls by RNA sequencing. A potential biomarker was selected based on the false discovery rate, and experiments were performed in TNF-α-stimulated primary cultured human tubular epithelial cells (hTECs). We identified 3037 genes with low expression and 2852 genes with high expression in the disease samples compared to the controls. Dual-specificity phosphatase 1 (DUSP1) exhibited universal low expression in various diseases (log2 fold change, -3.87), with the lowest false discovery rate (7.03E-132). In further experimental validation study, DUSP1 overexpression ameliorated inflammatory markers related to MAP kinase pathways in hTECs, while pharmacologic inhibition of DUSP1 increased these markers. The combination of DUSP1 overexpression with low-concentration corticosteroid treatment resulted in more potent suppression of inflammation than high-concentration corticosteroid treatment alone. The profiled transcriptomes provide insights into the pathophysiology of tubulointerstitial injury in kidney diseases and may reveal a potential therapeutic biomarker.


Assuntos
Glomerulonefrite por IGA , Glomerulonefrite , Biomarcadores , Biópsia , Glomerulonefrite/tratamento farmacológico , Glomerulonefrite/genética , Glomerulonefrite por IGA/tratamento farmacológico , Glomerulonefrite por IGA/genética , Glomerulonefrite por IGA/metabolismo , Humanos , Monoéster Fosfórico Hidrolases , RNA-Seq
3.
Vet Pathol ; 59(4): 707-711, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35038930

RESUMO

Documented natural infections with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in exotic and companion animals following human exposures are uncommon. Those documented in animals are typically mild and self-limiting, and infected animals have only infrequently died or been euthanized. Through a coordinated One Health initiative, necropsies were conducted on 5 animals from different premises that were exposed to humans with laboratory-confirmed SARS-CoV-2 infection. The combination of epidemiologic evidence of exposure and confirmatory real-time reverse transcriptase-polymerase chain reaction testing confirmed infection in 3 cats and a tiger. A dog was a suspect case based on epidemiologic evidence of exposure but tested negative for SARS-CoV-2. Four animals had respiratory clinical signs that developed 2 to 12 days after exposure. The dog had bronchointerstitial pneumonia and the tiger had bronchopneumonia; both had syncytial-like cells with no detection of SARS-CoV-2. Individual findings in the 3 cats included metastatic mammary carcinoma, congenital renal disease, and myocardial disease. Based on the necropsy findings and a standardized algorithm, SARS-CoV-2 infection was not considered the cause of death in any of the cases. Continued surveillance and necropsy examination of animals with fatal outcomes will further our understanding of natural SARS-CoV-2 infection in animals and the potential role of the virus in development of lesions.


Assuntos
COVID-19 , Doenças do Cão , Saúde Única , Animais , COVID-19/veterinária , Doenças do Cão/diagnóstico , Cães , Animais de Estimação , SARS-CoV-2
4.
Biomed Pharmacother ; 144: 112324, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34678732

RESUMO

DA-1241 is a novel small molecule G protein-coupled receptor 119 (GPR119) agonist in early clinical development for type 2 diabetic patients. This study aimed to elucidate the pharmacological characteristics of DA-1241 for its hypoglycemic action. DA-1241 potently and selectively activated GPR119 with enhanced maximum efficacy. DA-1241 increased intracellular cAMP in HIT-T15 insulinoma cells (EC50, 14.7 nM) and increased insulin secretion (EC50, 22.3 nM) in association with enhanced human insulin promoter activity. Accordingly, postprandial plasma insulin levels were increased in mice after single oral administration of DA-1241. Postprandial glucose excursion was significantly reduced by single oral administration of DA-1241 in wild-type mice but not in GPR119 knockout mice. GLP-1 secretion was increased by DA-1241 treatment in mice. Thus, upon combined sitagliptin and DA-1241 treatment in high-fat diet/streptozotocin (HFD/STZ)-induced diabetic mice, plasma active GLP-1 levels were synergistically increased. Accordingly, blood glucose and triglyceride levels were significantly lowered both by DA-1241 and sitagliptin alone and in combination. Immunohistochemical analysis revealed that ß-cell mass with reduced PDX1 levels in the islets from HFD/STZ diabetic mice was significantly preserved by DA-1241, whereas increased glucagon and BiP levels were significantly suppressed. In HIT-T15 insulinoma cells subjected to ER stress, decreased cell viability was significantly rescued by treatment with DA-1241. Additionally, increased apoptosis was largely attenuated by DA-1241 by inhibiting BiP and CHOP expression through suppression of p38 MAPK. In conclusion, these studies provide evidence that DA-1241 can be a promising antidiabetic drug by potentially preserving pancreatic functions through suppressing ER stress and increasing PDX1 expression.


Assuntos
Glicemia/efeitos dos fármacos , Diabetes Mellitus Experimental/tratamento farmacológico , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Proteínas de Homeodomínio/metabolismo , Hipoglicemiantes/farmacologia , Oxidiazóis/farmacologia , Pâncreas/efeitos dos fármacos , Piperidinas/farmacologia , Pirimidinas/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Transativadores/metabolismo , Animais , Apoptose/efeitos dos fármacos , Glicemia/metabolismo , Linhagem Celular Tumoral , Cricetinae , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/patologia , Dieta Hiperlipídica , Proteínas de Homeodomínio/genética , Insulina/sangue , Masculino , Camundongos Endogâmicos ICR , Camundongos Knockout , Oxidiazóis/uso terapêutico , Pâncreas/metabolismo , Pâncreas/patologia , Piperidinas/uso terapêutico , Pirimidinas/uso terapêutico , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Estreptozocina , Transativadores/genética , Triglicerídeos/sangue , Regulação para Cima
5.
Anticancer Res ; 39(10): 5483-5494, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31570442

RESUMO

BACKGROUND/AIM: Canine mammary gland tumors (CMGTs) are the most common tumors in female dogs. Rivoceranib (also known as apatinib) is a novel anti-angiogenic tyrosine kinase inhibitor that selectively binds to vascular endothelial growth factor receptor-2 (VEGFR2). The aim of this study was to disclose the antitumor effects of rivoceranib on CMGT cell lines. MATERIALS AND METHODS: The direct effects of rivoceranib on CMGT cells in vitro were analyzed by cell proliferation and migration assays. Cell-cycle distribution and apoptotic ratio were analyzed by flow cytometry. Expression levels of phosphorylated VEGFR2 were evaluated by western blot analysis. RESULTS: Rivoceranib treatment significantly reduced the proliferation and migration of CMGT cells in a dose-dependent manner. Flow cytometry results revealed significant increases in G0/G1 phase arrest and apoptosis proportional to the drug concentration used. Rivoceranib reduced the level of phosphorylated VEGFR2. CONCLUSION: We confirm that rivoceranib exerts antitumor effects on CMGT cells by inhibiting biological functions.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Mamárias Animais/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cães , Feminino , Fase G1/efeitos dos fármacos , Neoplasias Mamárias Animais/metabolismo , Fosforilação/efeitos dos fármacos , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Eur J Pharmacol ; 830: 95-104, 2018 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-29727638

RESUMO

Due to very limited preclinical reports, pharmacodynamic interactions between dipeptidyl peptidase 4 (DPP4) inhibitors and peroxisome proliferator-activated receptor γ (PPARγ) agonists are not conclusive yet. This study aimed to evaluate the pharmacological responses from adding evogliptin, a DPP4 inhibitor, to pioglitazone, a PPARγ agonist, in diabetic db/db mice after a 2-week treatment. This combination led to further decrease in both fasting and fed blood glucose levels compared to evogliptin alone (P < 0.05), but combination effects were more dramatic in fasting glucose levels (P < 0.05 vs. each treatment alone). Of note, plasma glucagon and high-molecular-weight (HMW) form of adiponectin were also further altered by the combination (P < 0.05 vs. each treatment alone). In line with these results, hepatic gluconeogenic gene expression was normalized by this combination. However, although evogliptin or pioglitazone directly suppressed glucose output in HepG2 hepatocytes, their combination did not further reduce hepatic glucose output. By contrast, glucose utilization of HepG2 cells was synergistically enhanced by this combination regardless of insulin presence (P < 0.05 vs. each treatment alone). These results suggest that the combination of evogliptin and pioglitazone is more efficacious in fasting glucose control through systemic alterations such as decreasing glucagon and increasing adiponectin, and through enhancing glucose utilization. To our knowledge, this is the first report regarding the significant combination effects of DPP4 inhibitors plus PPARγ agonists on plasma HMW adiponectin and hepatic glucose utilization. Our findings provide insight that the evogliptin and pioglitazone combination therapy may be more beneficial in type 2 diabetic patients characterized by exaggerated glucagon dysregulation.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Inibidores da Dipeptidil Peptidase IV/farmacologia , Fígado/efeitos dos fármacos , Piperazinas/farmacologia , Tiazolidinedionas/farmacologia , Animais , Glicemia/análise , Diabetes Mellitus Experimental/sangue , Sinergismo Farmacológico , Jejum/sangue , Glucagon/sangue , Glucose/metabolismo , Células Hep G2 , Humanos , Fígado/metabolismo , Masculino , Camundongos , Pioglitazona
7.
Oncotarget ; 6(10): 8132-43, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25924237

RESUMO

Voltage-gated potassium (Kv) channels are known to be involved in cancer development and cancer cell proliferation. KV9.3, an electronically silent subunit, forms heterotetramers with KV2.1 in excitable cells and modulates its electrophysiological properties. However, the role of KV9.3 alone in non-excitable cancer cells has not been studied. Here, we evaluated the effect of silencing KV9.3 on cancer cell proliferation in HCT15 colon carcinoma cells and A549 lung adenocarcinoma cells. We confirmed the expression of KV9.3 mRNA in HCT15 and A549 cells and showed that silencing KV9.3 using small interfering RNA caused G0/G1 cell cycle arrest and alterations in cell cycle regulatory proteins in both HCT15 and A549 cells without affecting apoptosis. Also, stable knockdown of KV9.3 expression using short-hairpin RNA inhibited tumor growth in SCID mouse xenograft model. Using a bioinformatics approach, we identified Sp1 binding sites in the promoter region of the gene encoding KV9.3. We further found that Sp1 bound to this region and showed that the Sp1 inhibitor, mithramycin A, induced a concentration-dependent decrease in KV9.3 expression. Taken together, these data suggest that knockdown of KV9.3 inhibits proliferation in colon carcinoma and lung adenocarcinoma cell lines and may be regulated by Sp1.


Assuntos
Adenocarcinoma/metabolismo , Neoplasias do Colo/metabolismo , Neoplasias Pulmonares/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Animais , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Expressão Gênica , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA