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

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Front Pharmacol ; 14: 1139673, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36992833

RESUMO

Oral GSH supplementation along with antidiabetic treatment was shown to restore the body stores of GSH significantly and reduce oxidative DNA damage (8-OHdG) in Indian Type 2 diabetic (T2D) patients over 6 months in our recent clinical study. Post hoc analysis of the data also suggested that elder patients benefit from improved HbA1c and fasting insulin. We modeled longitudinal changes in diabetic individuals using a linear mixed-effects (LME) framework and obtained i) the distribution of individual trajectories with and without GSH supplementation and ii) the overall rates of changes in the different study arms. Serial changes in elder and younger diabetic individuals were also modeled independently to examine differences in their progression. The average linear trajectories obtained from the model explain how biochemical parameters in T2D patients progress over 6 months on GSH supplementation. Model estimates show improvements in erythrocytic GSH of 108 µM per month and a reduction in 8-OHdG at a rate of 18.5 ng/µg DNA per month in T2D patients. GSH replenishes faster in younger people than in the elder. 8-OHdG reduced more rapidly in the elder (24 ng/µg DNA per month) than in younger (12 ng/µg DNA per month) individuals. Interestingly, elder individuals show a substantial reduction in HbA1c (0.1% per month) and increased fasting insulin (0.6 µU/mL per month). Changes in GSH correlate strongly with changes in HbA1c, 8-OHdG, and fasting insulin in the elder cohort. The model estimates strongly suggest it improves the rate of replenishment in erythrocytic GSH stores and reduces oxidative DNA damage. Elder and younger T2D patients respond differently to GSH supplementation: It improves the rate of reduction in HbA1c and increases fasting insulin in elder patients. These model forecasts have clinical implications that aid in personalizing treatment targets for using oral GSH as adjuvant therapy in diabetes.

2.
Nat Prod Res ; 37(18): 3145-3151, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36373743

RESUMO

Adipogenesis involves commitment of stem cells and their differentiation into mature adipocytes. It is tightly regulated by hormones, nutrients and adipokines. Many natural compounds are being tested for their anti-adipogenic activity which can be attributed to apoptosis induction in adipocytes, blocking adipocyte differentiation, or inhibiting intracellular triglyceride synthesis and accumulation. In this study, we have determined molecular mechanism of two phytocompounds: andrographolide (AN) and pterostilbene (PT) during differentiation of the human MSCs into adipocyte. Interestingly, AN upregulates miR27a, whereas, PT upregulated SIRT1 which inhibits the expression of PPARγ. Thus, our results clearly demonstrate that both AN and PT inhibited adipogenesis by blocking a surge of reactive oxygen species (ROS) during differentiation and inhibiting expression of crucial transcription factors like SREBP1c and PPARγ.

3.
J Biomol Struct Dyn ; : 1-15, 2023 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-37753733

RESUMO

In diabetes, persistent hyperglycemia generates excess reactive oxygen species (ROS), leading to oxidative stress (OS). In response to OS, transcription factors (TFs) Nrf2 and FoxO1 get activated, which induce the expression of antioxidant enzymes catalase (CAT) and superoxide dismutase (SOD). It is well documented that the antioxidant response in diabetic individuals is very low. Since Nrf2 and FoxO1 are the major TFs activating these genes, we were interested in determining if single nucleotide polymorphisms (SNPs) in genes for these TFs have any association with lowered antioxidant enzyme activity in diabetic individuals. The activity of CAT and SOD and total antioxidant capacity (TAC) were quantified from the serum samples of diabetic (n = 98) and non-diabetic (n = 90) individuals. Genomic DNA was isolated, and Nrf2 and FoxO1 were amplified and sequenced by Illumina NextSeq500. Data were screened for SNPs in amplified regions. An independent samples t-test to find an association between CAT, SOD, and TAC and allele frequency of SNP with the diabetic condition was carried out. We found decreased CAT and SOD activity and significantly low TAC in diabetic individuals. Thirty-two and thirty-four SNPs and Single-nucleotide variants (SNVs) were observed in Nrf2 and FoxO1, respectively. However, a statistically significant difference in the allele frequency distribution between study groups was observed only in two intronic SNPs, rs17524059:A > C and rs60373589:Indel(A) of Nrf2 and FoxO1, respectively. SNPs, rs17524059 in the Nrf2 and rs60373589 of FoxO1, were not associated with reduced CAT and SOD activity and level of TAC in Indian diabetic individuals.Communicated by Ramaswamy H. Sarma.

4.
Front Endocrinol (Lausanne) ; 14: 1264072, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38053728

RESUMO

Introduction: The development of continuous glucose monitoring (CGM) over the last decade has provided access to many consecutive glucose concentration measurements from patients. A standard method for estimating glycated hemoglobin (HbA1c), already established in the literature, is based on its relationship with the average blood glucose concentration (aBG). We showed that the estimates obtained using the standard method were not sufficiently reliable for an Indian population and suggested two new methods for estimating HbA1c. Methods: Two datasets providing a total of 128 CGM and their corresponding HbA1c levels were received from two centers: Health Centre, Savitribai Phule Pune University, Pune and Joshi Hospital, Pune, from patients already diagnosed with diabetes, non-diabetes, and pre-diabetes. We filtered 112 data-sufficient CGM traces, of which 80 traces were used to construct two models using linear regression. The first model estimates HbA1c directly from the average interstitial fluid glucose concentration (aISF) of the CGM trace and the second model proceeds in two steps: first, aISF is scaled to aBG, and then aBG is converted to HbA1c via the Nathan model. Our models were tested on the remaining 32 data- sufficient traces. We also provided 95% confidence and prediction intervals for HbA1c estimates. Results: The direct model (first model) for estimating HbA1c was HbA1cmmol/mol = 0.319 × aISFmg/dL + 16.73 and the adapted Nathan model (second model) for estimating HbA1c is HbA1cmmol/dL = 0.38 × (1.17 × ISFmg/dL) - 5.60. Discussion: Our results show that the new equations are likely to provide better estimates of HbA1c levels than the standard model at the population level, which is especially suited for clinical epidemiology in Indian populations.


Assuntos
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Humanos , Hemoglobinas Glicadas , Glicemia , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/epidemiologia , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/epidemiologia , Automonitorização da Glicemia/métodos , Índia/epidemiologia
5.
FEMS Microbiol Lett ; 3702023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-37935462

RESUMO

The aim of this study was to check the effect of long-term oral glutathione (GSH) supplementation on alteration in gut microbiome of Indian diabetic individuals. Early morning fresh stool sample of diabetic individuals recruited in a randomized clinical trial wherein they were given 500 mg GSH supplementation orally once a day for a period of 6 months was collected and gut microbiome was analysed using high throughput 16S rRNA metagenomic sequencing. Long-term GSH supplementation as reported in our earlier work showed significant increase in body stores of GSH and stabilized decreased glycated haemoglobin (HbA1c). Analysis of gut microbiome revealed that abundance of phylum Proteobacteria significantly decreased (P < 0.05) in individuals with GSH supplementation after 6 months compared to those without it. Beneficial dominant genera such as Megasphaera, Bacteroides, and Megamonas were found to be significantly enriched (P < 0.05), while pathogenic Escherichia/Shigella was found to be depleted (P < 0.05) after supplementation. Data clearly demonstrate that GSH supplementation along with antidiabetic treatment helps restore the gut microbiome by enriching beneficial bacteria of healthy gut and reducing significantly the load of pathogenic bacteria of diabetic gut.


Assuntos
Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Humanos , RNA Ribossômico 16S/genética , Glutationa , Diabetes Mellitus Tipo 2/tratamento farmacológico , Suplementos Nutricionais
6.
Antioxidants (Basel) ; 11(5)2022 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-35624890

RESUMO

Complications in type 2 diabetes (T2D) arise from hyperglycemia-induced oxidative stress. Here, we examined the effectiveness of supplementation with the endogenous antioxidant glutathione (GSH) during anti-diabetic treatment. A total of 104 non-diabetic and 250 diabetic individuals on anti-diabetic therapy, of either sex and aged between 30 and 78 years, were recruited. A total of 125 diabetic patients were additionally given 500 mg oral GSH supplementation daily for a period of six months. Fasting and PP glucose, insulin, HbA1c, GSH, oxidized glutathione (GSSG), and 8-hydroxy-2-deoxy guanosine (8-OHdG) were measured upon recruitment and after three and six months of supplementation. Statistical significance and effect size were assessed longitudinally across all arms. Blood GSH increased (Cohen's d = 1.01) and 8-OHdG decreased (Cohen's d = −1.07) significantly within three months (p < 0.001) in diabetic individuals. A post hoc sub-group analysis showed that HbA1c (Cohen's d = −0.41; p < 0.05) and fasting insulin levels (Cohen's d = 0.56; p < 0.05) changed significantly in diabetic individuals above 55 years. GSH supplementation caused a significant increase in blood GSH and helped maintain the baseline HbA1c overall. These results suggest GSH supplementation is of considerable benefit to patients above 55 years, not only supporting decreased glycated hemoglobin (HbA1c) and 8-OHdG but also increasing fasting insulin. The clinical implication of our study is that the oral administration of GSH potentially complements anti-diabetic therapy in achieving better glycemic targets, especially in the elderly population.

7.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1865(10): 158777, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32755726

RESUMO

AIMS: PPARγ is a crucial transcription factor involved in development of hepatic steatosis, an early stage of NAFLD. PPARγ is tightly regulated through various positive and negative regulators including miRNAs. In this study, we report for the first time miR-3666 as a negative regulator of PPARγ and its involvement in development of hepatic steatosis. METHODS: Binding of miR-3666 to regulate PPARγ was checked by luciferase assay and was confirmed by mutating PPARγ 3'UTR. Regulation of PPARγ was determined by overexpression of miR-3666 in HepG2 cells. Hepatic steatotic state in HepG2 cells was developed by exposure to excess palmitic acid and expression of PPARγ, miR-3666 and some PPARγ target and non-target genes was checked. Involvement of mir-3666 by regulating PPARγ in hepatic steatosis was also examined in liver of HFD fed mice. RESULTS: On overexpression of miR-3666, PPARγ expression decreased significantly in a dose-dependent manner in HepG2 cells. Binding of miR-3666 to PPARγ was confirmed as the luciferase activity using pMIR-REPORT with PPARγ 3'UTR decreased in PA treated HepG2 cells overexpressing miR-3666 and remained unchanged when PPARγ 3'UTR was mutated. In PA treated HepG2 cells during development of hepatic steatosis PPARγ was significantly up-regulated concomitant with down-regulation of miR-3666. Overexpression of miR-3666 in these cells decreased the extent of hepatic steatosis. Significant up-regulation of PPARγ and down-regulation of miR-3666 was also observed in liver of HFD fed mice indicating that miR-3666 regulates PPARγ in vivo. CONCLUSIONS: miR-3666 negatively regulates PPARγ by binding to its 3'UTR during development of hepatic steatosis.


Assuntos
Fígado Gorduroso/genética , MicroRNAs/genética , Hepatopatia Gordurosa não Alcoólica/genética , PPAR gama/genética , Regiões 3' não Traduzidas/genética , Animais , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Regulação da Expressão Gênica/genética , Células Hep G2 , Humanos , Fígado/metabolismo , Fígado/patologia , Camundongos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Ácido Palmítico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA