Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Clin Lab Anal ; 34(7): e23293, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32157741

RESUMO

BACKGROUND: Atherosclerosis is the most frequent pathological process that causes cardiovascular diseases. OBJECTIVE: The present study aimed to confirm miRNAs associated with atherosclerosis and explore the molecular mechanism of miR-34c and its target high mobility group box protein 1 (HMGB1) in the control of growth of smooth muscle cells in the development of atherosclerosis. METHODS: Real-time PCR was firstly performed to confirm miRNA correlation with atherosclerosis, and computational analysis and luciferase assay were performed to explore the target of miR-34c, Western blot, and real-time PCR were also utilized to reveal the effect of whether high glucose (HG) and miR-34c affect miR-34c, HMGB1 levels, NF-κB p65 and TNF-α levels, and the role of miR-34c on vascular smooth muscle cells (VSMCs) viability induced by HG. Students' unpaired t test was performed to compare data between two groups. RESULTS: MiR-34c level was associated with atherosclerosis with different expression between VSMCs treated with high glucose or normal VSMCs. Then, HMGB1 is a virtual target of miR-34c with predicted binding site resided in HMGB1 3'UTR and further verified by that miR-34c remarkably reduced luciferase activity of wild HMGB1 3'UTR under a concentration-dependent fashion, and miR-34c cannot influence luciferase activity of mutant HMGB1 3'UTR. CONCLUSIONS: The results suggested miR-34c might be a novel therapeutic strategy in the management of atherosclerosis by suppressing the expression of HGMB1 and its downstream effectors.


Assuntos
Proteína HMGB1/genética , MicroRNAs/genética , Músculo Liso Vascular/citologia , Regiões 3' não Traduzidas , Proliferação de Células/genética , Sobrevivência Celular , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Glucose/farmacologia , Proteína HMGB1/metabolismo , Humanos , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , NF-kappa B/genética , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
2.
J Clin Lab Anal ; 32(1)2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28220968

RESUMO

BACKGROUND: Vitamins A, D (Vitamin D2 and vitamin D3) and E, play an important role during pregnancy. METHODS: Sera were collected from 1056 normal pregnant women, who were between 18 and 40 years old, at seven different hospitals in northeastern China. The levels of Vitamin A and E in the sera samples were detected using HPLC (High Performance Liquid Chromatography), and the level of vitamin D was measured by LC-MS (Liquid Chromatography-Mass Spectrometry). Data were analyzed using IBM SPSS Statistics 21. RESULTS: The mean levels of vitamin A, D and E in the 1056 sera samples were 0.39 mg/L (0.38-0.39), 20.44 µg/L (19.86-21.08) and 12.96 mg/L (12.70-13.25), respectively. The levels of vitamin A, D, and E deficiency were 17.05%, 0.19%, and 56.44%, respectively. The levels of vitamin A, D, and E of those between age 21 and 31 among the 1056 pregnant women were similar. The correlation of vitamin E and D was significant at the .01 level (two-tailed), and the correlation of vitamin A and age was significant at the .05 level (2-tailed). CONCLUSION: According to our finding, the levels of vitamin A, D, and E in the sera of pregnant women in northeastern China were affected by where they live and their age. Vitamin D deficiency was very serious, vitamin A deficiency was common, while vitamin E seems to be sufficient.


Assuntos
Gravidez/sangue , Vitamina A/sangue , Vitamina D/sangue , Vitamina E/sangue , Adolescente , Adulto , China/epidemiologia , Feminino , Humanos , Modelos Lineares , Gravidez/estatística & dados numéricos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Adulto Jovem
3.
Int J Med Sci ; 11(11): 1107-15, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25170293

RESUMO

BACKGROUND: Although the protective effects of levocarnitine in patients with ischemic heart disease are related to the attenuation of oxidative stress injury, the exact mechanisms involved have yet to be fully understood. Our aim was to investigate the potential protective effects of levocarnitine pretreatment against oxidative stress in rat H9c2 cardiomyocytes. METHODS: Cardiomyocytes were exposed to H2O2 to create an oxidative stress model. The cells were pretreated with 50, 100, or 200 µM levocarnitine for 1 hour before H2O2 exposure. RESULTS: H2O2 exposure led to significant activation of oxidative stress in the cells, characterized by reduced viability, increased intracellular reactive oxygen species, lipid peroxidation, and reduced intracellular antioxidant activity. Mitochondrial dysfunction was also observed following H2O2 exposure, reflected by the loss of mitochondrial transmembrane potential and intracellular adenosine triphosphate. These pathophysiological processes led to cardiomyocyte apoptosis through activation of the intrinsic apoptotic pathway. More importantly, the levocarnitine pretreatment attenuated the H2O2-induced oxidative injury significantly, preserved mitochondrial function, and partially prevented cardiomyocyte apoptosis during the oxidative stress reaction. Western blotting analyses suggested that levocarnitine pretreatment increased plasma protein levels of Bcl-2, reduced Bax, and attenuated cytochrome C leakage from the mitochondria in the cells. CONCLUSION: Our in vitro study indicated that levocarnitine pretreatment may protect cardiomyocytes from oxidative stress-related damage.


Assuntos
Carnitina/farmacologia , Peróxido de Hidrogênio/farmacologia , Mitocôndrias/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ratos
4.
J Alzheimers Dis ; 73(1): 39-58, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31815697

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder, marked by cortical and hippocampal deposition of amyloid-ß (Aß) plaques and neurofibrillary tangles and cognitive impairment. Studies indicate a prominent link between cerebrovascular abnormalities and the onset and progression of AD, where blood-brain barrier (BBB) dysfunction and metabolic disorders play key risk factors. Pericyte degeneration, endothelial cell damage, astrocyte depolarization, diminished tight junction integrity, and basement membrane disarray trigger BBB damage. Subsequently, the altered expression of low-density lipoprotein receptor-related protein 1 and receptor for advanced glycation end products at the microvascular endothelial cells dysregulate Aß transport across the BBB. White matter lesions and microhemorrhages, dyslipidemia, altered brain insulin signaling, and insulin resistance contribute to tau and Aß pathogenesis, and oxidative stress, mitochondrial damage, inflammation, and hypoperfusion serve as mechanistic links between pathophysiological features of AD and ischemia. Deregulated calcium homeostasis, voltage gated calcium channel functioning, and protein kinase C signaling are also common mechanisms for both AD pathogenesis and cerebrovascular abnormalities. Additionally, APOE polymorphic alleles that characterize impaired cerebrovascular integrity function as primary genetic determinants of AD. Overall, the current review enlightens key vascular risk factors for AD and underscores pathophysiologic relationship between AD and vascular dysfunction.


Assuntos
Doença de Alzheimer/fisiopatologia , Barreira Hematoencefálica/fisiopatologia , Síndrome Metabólica/fisiopatologia , Doenças Vasculares/fisiopatologia , Doença de Alzheimer/genética , Humanos , Síndrome Metabólica/genética , Fatores de Risco , Doenças Vasculares/complicações , Doenças Vasculares/genética
5.
Chem Biol Interact ; 317: 108963, 2020 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-31978391

RESUMO

Epigenetic variations can play remarkable roles in different normal and abnormal situations. Such variations have been shown to have a direct role in the pathogenesis of various diseases either through inhibition of tumor suppressor genes or increasing the expression of oncogenes. Enzymes involving in epigenetic machinery are the main actors in tuning the epigenetic-based controls on gene expressions. Aberrant expression of these enzymes can trigger big chaos in the cellular gene expression networks and finally lead to cancer progression. This situation has been shown in different types of leukemia, where high or low levels of an epigenetic enzyme are partly or highly responsible for the involvement or progression of a disease. DNA hypermethylation, different histone modifications, and aberrant miRNA expressions are three main epigenetic variations, which have been shown to play a role in leukemia progression. Epigenetic based treatments now are considered as novel and effective therapies in order to decrease the abnormal epigenetic modifications in patient cells. Different epigenetic-based approaches have been developed and tested to inhibit or reverse the unusual expression of epigenetic agents in leukemia. Acute myeloid leukemia (AML), the most prevalent acute leukemia in adults, is anaggressive hematological malignancy arising in hematopoietic stem and progenitor cells. With the exception of a few specific AML subtypes, the mainstays of treatment have not significantly changed over the last 20 years, and are still based on standard cytotoxic chemotherapy. In this review, we will discuss the recent development of therapeutics specifically targeting these key epigenetic programs in AML, describe their mechanism of action and present their current clinical development. Finally, we will discuss the opportunities presented by epigenetically targeted therapy in AML and will highlight future challenges ahead for the AML community, to ensure that this novel therapeutics are optimally translated into clinical practice and result in clinical improvement for AML patients.


Assuntos
Antineoplásicos/uso terapêutico , Epigênese Genética , Leucemia/tratamento farmacológico , Leucemia/genética , Metilação de DNA/genética , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Terapia de Alvo Molecular/métodos
6.
Eur J Pharmacol ; 849: 146-153, 2019 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-30721700

RESUMO

Hydrogen sulfide (H2S) is a gaseous molecule and is endogenously produced in the brain by cystathionine beta-synthase, 3-mercaptopyruvate-sulfurtransferase, cysteine aminotransferase and cystathionine γ-lyase. Physiologically, H2S acts as a neuromodulator and regulates synaptic activity of neurons and glia to promote the development of long-term potentiation. A decrease in H2S levels in the brain and plasma has been directly correlated with the degree of severity of Alzheimer disease in patients. A large number of studies have shown a decrease in the H2S levels in experimental models of cognitive dysfunction and exogenous administration of sodium hydrosulfide (NaHS), a H2S donor, has been shown to prevent the development of memory deficits. The beneficial effects of H2S in different models has been ascribed to decrease in neuroinflammation, up-regulation of antioxidant defense, decrease in endoplasmic reticulum (ER) stress, inhibition of phosphatidylinositol 3-kinase (PI3-K)/Akt signaling, inhibition of mitogen activated protein (MAP) kinases, decrease in glutamate and normalization of NMDA receptors, inhibition of matrix metalloproteinases (MMPs), up-regulation of silent information regulator 1 (Sirt 1) and preservation of mitochondrial function. The present review describes the role of H2S in different models of cognitive deficits and human subjects along with possible mechanisms.


Assuntos
Disfunção Cognitiva/metabolismo , Sulfeto de Hidrogênio/metabolismo , Animais , Disfunção Cognitiva/genética , Disfunção Cognitiva/fisiopatologia , Humanos
7.
Life Sci ; 194: 88-97, 2018 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-29241711

RESUMO

Cardiovascular disease (CVD) is a key cause of deaths worldwide, comprising 15-17% of healthcare expenditure in developed countries. Current records estimate an annual global average of 30 million cardiac dysfunction cases, with a predicted escalation by two-three folds for the next 20-30years. Although ß-blockers and angiotensin-converting-enzymes are commonly prescribed to control CVD risk, hepatotoxicity and hematological changes are frequent adverse events associated with these drugs. Search for alternatives identified endogenous cofactor l-carnitine, which is capable of promoting mitochondrial ß-oxidation towards a balanced cardiac energy metabolism. l-Carnitine facilitates transport of long-chain fatty acids into the mitochondrial matrix, triggering cardioprotective effects through reduced oxidative stress, inflammation and necrosis of cardiac myocytes. Additionally, l-carnitine regulates calcium influx, endothelial integrity, intracellular enzyme release and membrane phospholipid content for sustained cellular homeostasis. Carnitine depletion, characterized by reduced expression of "organic cation transporter-2" gene, is a metabolic and autosomal recessive disorder that also frequently associates with CVD. Hence, exogenous carnitine administration through dietary and intravenous routes serves as a suitable protective strategy against ventricular dysfunction, ischemia-reperfusion injury, cardiac arrhythmia and toxic myocardial injury that prominently mark CVD. Additionally, carnitine reduces hypertension, hyperlipidemia, diabetic ketoacidosis, hyperglycemia, insulin-dependent diabetes mellitus, insulin resistance, obesity, etc. that enhance cardiovascular pathology. These favorable effects of l-carnitine have been evident in infants, juvenile, young, adult and aged patients of sudden and chronic heart failure as well. This review describes the mechanism of action, metabolism and pharmacokinetics of l-carnitine. It specifically emphasizes upon the beneficial role of l-carnitine in cardiomyopathy.


Assuntos
Cardiotônicos/uso terapêutico , Carnitina/uso terapêutico , Cardiopatias/tratamento farmacológico , Coração/efeitos dos fármacos , Animais , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patologia , Cardiotônicos/metabolismo , Carnitina/metabolismo , Cardiopatias/metabolismo , Cardiopatias/patologia , Humanos , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Disfunção Ventricular/tratamento farmacológico , Disfunção Ventricular/metabolismo , Disfunção Ventricular/patologia
8.
Exp Ther Med ; 7(5): 1141-1146, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24940400

RESUMO

Glycyrrhizin has a role in immune regulation in the central nervous system, but its impact on sciatic nerve injury had not previously been reported. In this study, a BALB/c mouse model of sciatic nerve injury was used to explore the role of glycyrrhizin in sciatic nerve repair and its underlying mechanism. Glycyrrhizin with intragastric gavage of 10 and 20 mg/kg weight per day (mid- and high-dose, respectively) inhibited p75 neurotrophin receptor (p75NTR) expression at the protein and mRNA levels versus the 5 mg/kg (low-dose) group and control (0.9% NaCl solution) at one, two, four and eight weeks following sciatic nerve injury, and simultaneously improved the action potential amplitude and motor nerve conductive velocity. Combined Marsland, Glees and Erikson's silver stain and Luxol fast blue staining results indicated that high- and mid-dose glycyrrhizin promoted improved sciatic nerve myelination compared with the low-dose or control groups eight weeks after injury. Immunofluorescence staining demonstrated that glycyrrhizin had an inhibitory effect to a certain degree on local hypertrophic scar and inflammatory responses in the mouse model. In conclusion, glycyrrhizin can promote sciatic nerve regeneration and functional repair, in which doses of 10 and 20 mg/kg per day are more effective than lower doses, and such regeneration is associated with the downregulation of p75NTR.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA