Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Life Sci ; 345: 122608, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38574885

RESUMEN

BACKGROUND AND AIMS: The protein phosphatase 1 regulatory inhibitor subunit 1A (PPP1R1A) has been linked with insulin secretion and diabetes mellitus. Yet, its full significance in pancreatic ß-cell function remains unclear. This study aims to elucidate the role of the PPP1R1A gene in ß-cell biology using human pancreatic islets and rat INS-1 (832/13) cells. RESULTS: Disruption of Ppp1r1a in INS-1 cells was associated with reduced insulin secretion and impaired glucose uptake; however, cell viability, ROS, apoptosis or proliferation were intact. A significant downregulation of crucial ß-cell function genes such as Ins1, Ins2, Pcsk1, Cpe, Pdx1, Mafa, Isl1, Glut2, Snap25, Vamp2, Syt5, Cacna1a, Cacna1d and Cacnb3, was observed upon Ppp1r1a disruption. Furthermore, silencing Pdx1 in INS-1 cells altered PPP1R1A expression, indicating that PPP1R1A is a target gene for PDX1. Treatment with rosiglitazone increased Ppp1r1a expression, while metformin and insulin showed no effect. RNA-seq analysis of human islets revealed high PPP1R1A expression, with α-cells showing the highest levels compared to other endocrine cells. Muscle tissues exhibited greater PPP1R1A expression than pancreatic islets, liver, or adipose tissues. Co-expression analysis revealed significant correlations between PPP1R1A and genes associated with insulin biosynthesis, exocytosis machinery, and intracellular calcium transport. Overexpression of PPP1R1A in human islets augmented insulin secretion and upregulated protein expression of Insulin, MAFA, PDX1, and GLUT1, while silencing of PPP1R1A reduced Insulin, MAFA, and GLUT1 protein levels. CONCLUSION: This study provides valuable insights into the role of PPP1R1A in regulating ß-cell function and glucose homeostasis. PPP1R1A presents a promising opportunity for future therapeutic interventions.


Asunto(s)
Células Secretoras de Insulina , Islotes Pancreáticos , Humanos , Ratas , Animales , Islotes Pancreáticos/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Secreción de Insulina/genética , Línea Celular , Glucosa/metabolismo , Canales de Calcio/metabolismo
2.
Cells ; 13(8)2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38667300

RESUMEN

Interleukin-6 (IL6) is a pleiotropic cytokine implicated in metabolic disorders and inflammation, yet its precise influence on insulin secretion and glucose metabolism remains uncertain. This study examined IL6 expression in pancreatic islets from individuals with/without diabetes, alongside a series of functional experiments, including siRNA silencing; IL6 treatment; and assessments of glucose uptake, cell viability, apoptosis, and expression of key ß-cell genes, which were conducted in both INS-1 cells and human islets to elucidate the effect of IL6 on insulin secretion. Serum levels of IL6 from Emirati patients with type 2 diabetes (T2D) were measured, and the effect of antidiabetic drugs on IL6 levels was studied. The results revealed that IL6 mRNA expression was higher in islets from diabetic and older donors compared to healthy or young donors. IL6 expression correlated negatively with PDX1, MAFB, and NEUROD1 and positively with SOX4, HES1, and FOXA1. Silencing IL6 in INS-1 cells reduced insulin secretion and glucose uptake independently of apoptosis or oxidative stress. Reduced expression of IL6 was associated with the downregulation of Ins, Pdx1, Neurod1, and Glut2 in INS-1 cells. In contrast, IL6 treatment enhanced insulin secretion in INS-1 cells and human islets and upregulated insulin expression. Serum IL6 levels were elevated in patients with T2D and associated with higher glucose, HbA1c, and triglycerides, regardless of glucose-lowering medications. This study provides a new understanding of the role of IL6 in ß-cell function and the pathophysiology of T2D. Our data highlight differences in the response to IL6 between INS-1 cells and human islets, suggesting the presence of species-specific variations across different experimental models. Further research is warranted to unravel the precise mechanisms underlying the observed effects of IL-6 on insulin secretion.


Asunto(s)
Diabetes Mellitus Tipo 2 , Secreción de Insulina , Interleucina-6 , Islotes Pancreáticos , Humanos , Interleucina-6/metabolismo , Interleucina-6/sangre , Secreción de Insulina/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/sangre , Masculino , Persona de Mediana Edad , Femenino , Adulto , Glucosa/metabolismo , Insulina/metabolismo , Insulina/sangre , Ratas , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Animales , Línea Celular , Anciano , Apoptosis/efectos de los fármacos
3.
Life Sci ; 339: 122421, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38232799

RESUMEN

AIMS: In this study, we investigated the role of the FTO gene in pancreatic ß-cell biology and its association with type 2 diabetes (T2D). To address this issue, human pancreatic islets and rat INS-1 (832/13) cells were used to perform gene silencing, overexpression, and functional analysis of FTO expression; levels of FTO were also measured in serum samples obtained from diabetic and obese individuals. RESULTS: The findings revealed that FTO expression was reduced in islets from hyperglycemic/diabetic donors compared to normal donors. This reduction correlated with decreased INS and GLUT1 expression and increased PDX1, GCK, and SNAP25 expression. Silencing of Fto in INS-1 cells impaired insulin release and mitochondrial ATP production and increased apoptosis in pro-apoptotic cytokine-treated cells. However, glucose uptake and reactive oxygen species production rates remained unaffected. Downregulation of key ß-cell genes was observed following Fto-silencing, while Glut2 and Gck were unaffected. RNA-seq analysis identified several dysregulated genes involved in metal ion binding, calcium ion binding, and protein serine/threonine kinase activity. Furthermore, our findings showed that Pdx1 or Mafa-silencing did not influence FTO protein expression. Overexpression of FTO in human islets promoted insulin secretion and upregulated INS, PDX1, MAFA, and GLUT1 expression. Serum FTO levels did not significantly differ between individuals with diabetes or obesity and their healthy counterparts. CONCLUSION: These findings suggest that FTO plays a crucial role in ß-cell survival, metabolism, and function and point to a potential therapeutic utility of FTO in T2D patients.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Islotes Pancreáticos , Humanos , Ratas , Animales , Secreción de Insulina/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Islotes Pancreáticos/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Obesidad/genética , Obesidad/metabolismo , Glucosa/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo
4.
Heliyon ; 9(9): e19234, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37662743

RESUMEN

Rosmarinic acid (RA), a natural ester phenolic compound, is known to have antioxidant and anti-inflammatory properties. RA has also been reported to exhibit a hypoglycemic effect; however, the mechanisms underlying this effect have yet to be investigated. Therefore, the present study focused on the anti-diabetic effects and mechanism of RA in INS-1 cells using in vitro model. Streptozotocin (STZ) at a concentration of 3 mM was applied to INS-1 cells for 4 h to create a diabetic model. The cells were pretreated for 24 h with various concentrations (1 and 2.5 µM) of RA. The Cell viability, glucose-stimulated insulin secretion (GSIS), glucose uptake, lipid peroxidation, reactive oxygen species (ROS), apoptosis, and protein expression of Bcl-2, NF-κB, 1L-1ß, and PARP were assessed. Results showed that STZ-treated INS-1 cells exhibited reduced cell viability, insulin release, insulin content, glucose uptake, and elevated MDA and ROS levels. Cells pretreated with RA maintained the function and morphology of ß-cells against STZ-induced damage. Moreover, RA sustained high protein expression levels of Bcl-2 and low expression levels of NF-κB, IL-1ß, and PARP. In conclusion, RA preserved ß-cells function against STZ-induced damage by altering NF-κB and Bcl-2 pathways.

5.
Mol Cell Endocrinol ; 574: 111987, 2023 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-37311518

RESUMEN

The role of "Family with sequence similarity 105, member A" (FAM105A) in pancreatic ß-cell function in relation to type 2 diabetes mellitus (T2D) is not fully understood. To address this issue, various molecular and functional experiments were conducted on primary human islets and INS-1 cells. RNA-seq expression analysis showed that FAM105A is highly expressed in human islets and its expression is reduced in diabetic islets compared to healthy islets. FAM105A expression correlated negatively with HbA1c levels and body mass index (BMI). Co-expression analysis showed a significant correlation between FAM105A with PDX1, GCK, GLUT1 and INSR, but not the INS gene. Silencing of Fam105a impaired insulin release, content, glucose uptake, and mitochondria ATP content but did not affect cell viability, reactive oxygen species (ROS) or apoptosis levels. Silencing of Fam105a was associated with reduced Pdx1 and Glut2 expression at mRNA and protein levels. RNA-seq analysis of dysregulated genes in Fam105a-silenced cells showed an overall downregulation of gene expression in ß-cells and insulin secretion pathway. Disrupting Pdx1 did not affect Fam105a expression in INS-1 cells. Overall, the results suggest that FAM105A plays an important role in pancreatic ß-cells biology and may be involved in the development of T2D.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Islotes Pancreáticos , Humanos , Secreción de Insulina , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Supervivencia Celular/genética , Glucosa/metabolismo , Islotes Pancreáticos/metabolismo
7.
Int J Mol Sci ; 24(5)2023 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-36902422

RESUMEN

Inflammasomes have been implicated in the pathogenesis of type 2 diabetes (T2D). However, their expression and functional importance in pancreatic ß-cells remain largely unknown. Mitogen-activated protein kinase 8 interacting protein-1 (MAPK8IP1) is a scaffold protein that regulates JNK signaling and is involved in various cellular processes. The precise role of MAPK8IP1 in inflammasome activation in ß-cells has not been defined. To address this gap in knowledge, we performed a set of bioinformatics, molecular, and functional experiments in human islets and INS-1 (832/13) cells. Using RNA-seq expression data, we mapped the expression pattern of proinflammatory and inflammasome-related genes (IRGs) in human pancreatic islets. Expression of MAPK8IP1 in human islets was found to correlate positively with key IRGs, including the NOD-like receptor (NLR) family pyrin domain containing 3 (NLRP3), Gasdermin D (GSDMD) and Apoptosis-associated speck-like protein containing a CARD (ASC), but correlate inversely with Nuclear factor kappa ß1 (NF-κß1), Caspase-1 (CASP-1), Interleukin-18 (IL-18), Interleukin-1ß (IL-1ß) and Interleukin 6 (IL-6). Ablation of Mapk8ip1 by siRNA in INS-1 cells down-regulated the basal expression levels of Nlrp3, NLR family CARD domain containing 4 (Nlrc4), NLR family CARD domain containing 1 (Nlrp1), Casp1, Gsdmd, Il-1ß, Il-18, Il-6, Asc, and Nf-κß1 at the mRNA and/or protein level and decreased palmitic acid (PA)-induced inflammasome activation. Furthermore, Mapk8ip1-silened cells substantially reduced reactive oxygen species (ROS) generation and apoptosis in palmitic acid-stressed INS-1 cells. Nonetheless, silencing of Mapk8ip1 failed to preserve ß-cell function against inflammasome response. Taken together, these findings suggest that MAPK8IP1 is involved in regulating ß-cells by multiple pathways.


Asunto(s)
Diabetes Mellitus Tipo 2 , Inflamasomas , Células Secretoras de Insulina , Humanos , Caspasa 1/metabolismo , Inflamasomas/metabolismo , Interleucina-18 , Interleucina-1beta/metabolismo , Interleucina-6 , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas NLR , Ácido Palmítico , Proteínas Adaptadoras Transductoras de Señales/genética , Células Secretoras de Insulina/metabolismo
8.
Exp Biol Med (Maywood) ; 248(4): 339-349, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36740767

RESUMEN

Mounting evidence points to a link between growth differentiation factor-15 (GDF15) expression and the onset and progression of diabetes mellitus. However, the exact role of GDF15 in pancreatic ß-cell function is unclear. To examine the role of GDF15 in ß-cell function, bioinformatics analysis and functional experiments involving GDF15 silencing and overexpression were performed in INS-1 cells and human islets. Public microarray and RNA-seq expression data showed that islets obtained from diabetic donors express high levels of GDF15 compared to islets obtained from normal donors. Moreover, analysis of RNA-seq expression data revealed that GDF15 expression correlates positively with that of insulin (INS), KCNJ11, GLUT1, MAFA, INSR and negatively with that of Glucokinase (GCK) and Alpha-Ketoglutarate Dependent Dioxygenase (FTO). No T2D-associated genetic variants in the GDF15 were found to pass genome-wide significance in the TIGER portal. Expression silencing of Gdf15 in INS-1 cells reduced insulin release, glucose uptake levels, increased reactive oxygen species (ROS) production and apoptosis levels. While Gdf15-silenced cells downregulated mRNA expression of Ins, Pdx1, Mafa, and Glut2 genes, its overexpression human islets was associated with increased insulin secretion and upregulated expression of MAFA and GLUT1 but not INS or GCK. Silencing of Pdx1 or Mafa in INS-1 cells did not affect the expression of GDF15. These findings suggest that GDF15 plays a significant role in pancreatic ß-cell function.


Asunto(s)
Células Secretoras de Insulina , Islotes Pancreáticos , Humanos , Secreción de Insulina , Transportador de Glucosa de Tipo 1/metabolismo , Islotes Pancreáticos/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Glucosa/metabolismo , Factor 15 de Diferenciación de Crecimiento/genética , Factor 15 de Diferenciación de Crecimiento/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo
9.
Metabolites ; 13(2)2023 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-36837926

RESUMEN

Mitogen-activated protein kinase 8 interacting protein-1 (MAPK8IP1) gene has been recognized as a susceptibility gene for diabetes. However, its action in the physiology of pancreatic ß-cells is not fully understood. Herein, bioinformatics and genetic analyses on the publicly available database were performed to map the expression of the MAPK8IP1 gene in human pancreatic islets and to explore whether this gene contains any genetic variants associated with type 2 diabetes (T2D). Moreover, a series of functional experiments were executed in a rat insulinoma cell line (INS-1 832/13) to investigate the role of the Mapk8ip1 gene in ß-cell function. Metabolic engineering using RNA-sequencing (RNA-seq) data confirmed higher expression levels of MAPK8IP1 in human islets compared to other metabolic tissues. Additionally, comparable expression of MAPK8IP1 expression was detected in sorted human endocrine cells. However, ß-cells exhibited higher expression of MAPK8IP1 than ductal and PSC cells. Notably, MAPK8IP1 expression was reduced in diabetic islets, and the expression was positively correlated with insulin and the ß-cell transcription factor PDX1 and MAFA. Using the TIGER portal, we found that one genetic variant, "rs7115753," in the proximity of MAPK8IP1, passes the genome-wide significance for the association with T2D. Expression silencing of Mapk8ip1 by small interfering RNA (siRNA) in INS-1 cells reduced insulin secretion, glucose uptake rate, and reactive oxygen species (ROS) production. In contrast, insulin content, cell viability, and apoptosis without cytokines were unaffected. However, silencing of Mapk8ip1 reduced cytokines-induced apoptosis and downregulated the expression of several pancreatic ß-cell functional markers including, Ins1, Ins2, Pdx1, MafA, Glut2, Gck, Insr, Vamp2, Syt5, and Cacna1a at mRNA and/or protein levels. Finally, we reported that siRNA silencing of Pdx1 resulted in the downregulation of MAPK8IP1 expression in INS-1 cells. In conclusion, our findings confirmed that MAPK8IP1 is an important component of pancreatic ß-cell physiology and insulin secretion.

11.
Biology (Basel) ; 11(7)2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-36101450

RESUMEN

Various studies have suggested a link between vitamin A (VA), all-trans-retinol, and type 2 diabetes (T2D). However, the functional role/expression of vitamin A receptors (Rarα, ß, and γ) in pancreatic ß-cells is not clear yet. Accordingly, we performed a series of bioinformatics, molecular and functional experiments in human islet and INS-1 cells to evaluate the role of Rarß on insulin secretion and pancreatic ß-cell function. Microarray and RNA-sequencing (RAN-seq) expression analysis showed that RARα, ß, and γ are expressed in human pancreatic islets. RNA-seq expression of RARß in diabetic/hyperglycemic human islets (HbA1c ≥ 6.3%) revealed a significant reduction (p = 0.004) compared to nondiabetic/normoglycemic cells (HbA1c < 6%). The expression of RARß with INS and PDX1 showed inverse association, while positive correlations were observed with INSR and HbA1c levels. Exploration of the T2D knowledge portal (T2DKP) revealed that several genetic variants in RARß are associated with BMI. The most associated variant is rs6804842 (p = 1.2 × 10−25). Silencing of Rarß in INS-1 cells impaired insulin secretion without affecting cell viability or apoptosis. Interestingly, reactive oxygen species (ROS) production levels were elevated and glucose uptake was reduced in Rarß-silenced cells. mRNA expression of Ins1, Pdx1, NeuroD1, Mafa, Snap25, Vamp2, and Gck were significantly (p < 0.05) downregulated in Rarß-silenced cells. For protein levels, Pro/Insulin, PDX1, GLUT2, GCK, pAKT/AKT, and INSR expression were downregulated considerably (p < 0.05). The expression of NEUROD and VAMP2 were not affected. In conclusion, our results indicate that Rarß is an important molecule for ß-cell function. Hence, our data further support the potential role of VA receptors in the development of T2D.

12.
Biology (Basel) ; 11(3)2022 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-35336762

RESUMEN

EXOC6 and EXOC6B (EXOC6/6B) components of the exocyst complex are involved in the secretory granule docking. Recently, EXOC6/6B were anticipated as a molecular link between dysfunctional pancreatic islets and ciliated lung epithelium, making diabetic patients more prone to severe SARS-CoV-2 complications. However, the exact role of EXOC6/6B in pancreatic ß-cell function and risk of T2D is not fully understood. Herein, microarray and RNA-sequencing (RNA-seq) expression data demonstrated the expression of EXOC6/6B in human pancreatic islets. Expression of EXOC6/6B was not affected by diabetes status. Exploration of the using the translational human pancreatic islet genotype tissue-expression resource portal (TIGER) revealed three genetic variants (rs947591, rs2488071 and rs2488073) in the EXOC6 gene that were associated (p < 2.5 × 10−20) with the risk of T2D. Exoc6/6b silencing in rat pancreatic ß-cells (INS1-832/13) impaired insulin secretion, insulin content, exocytosis machinery and glucose uptake without cytotoxic effect. A significant decrease in the expression Ins1, Ins1, Pdx1, Glut2 and Vamp2 was observed in Exoc6/6b-silenced cells at the mRNA and protein levels. However, NeuroD1, Gck and InsR were not influenced compared to the negative control. In conclusion, our data propose that EXOC6/6B are crucial regulators for insulin secretion and exocytosis machinery in ß-cells. This study identified several genetic variants in EXOC6 associated with the risk of T2D. Therefore, EXOC6/6B could provide a new potential target for therapy development or early biomarkers for T2D.

13.
Front Endocrinol (Lausanne) ; 13: 844472, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35282461

RESUMEN

Metabolic syndrome (MetS) is a serious health condition that is becoming extremely threatening in Saudi Arabia. The link between vitamin D receptor (VDR) gene polymorphisms and maternal MetS has been observed in several ethnic groups, but is yet to be clarified in the Arabian population. This study aims to investigate the relationship between the FokI VDR genotype and the risk of MetS and its components in pregnant Saudi women. A cross-sectional study was conducted using 368 pregnant Saudi women on first trimester screened for MetS (44 with MetS and 324 without MetS). Measurements included anthropometrics, glycemic and lipid profile and 25(OH)D. TaqMan genotyping assay was used to determine Fokl VDR genotype of participants. Vitamin D deficiency (25(OH)D <50nmol/l) was seen in 85% of the participants. An estimated 12% of participants had MetS. In the MetS group, the FokI VDR genotyping frequencies for FF, Ff, and ff genotypes were 50%, 36.4% and 13.6%, respectively. In controls, the frequencies were 62.7%, 31.4% and 5.9%, respectively. No significant association between the individual MetS components and FokI VDR genotypes were observed. Nevertheless, carriers of the ff allele had a significant risk for full maternal MetS [Odds Ratio 4.2 (95% Confidence Interval 1.4-12.2; adjusted p=0.009). The study suggests that the ff FokI VDR genotype is a genetic marker of maternal MetS in pregnant Arabian women. Prospective studies that include neonatal outcomes may confirm present findings.


Asunto(s)
Síndrome Metabólico , Receptores de Calcitriol/genética , Árabes/genética , Estudios Transversales , Femenino , Humanos , Recién Nacido , Síndrome Metabólico/epidemiología , Síndrome Metabólico/genética , Polimorfismo Genético , Embarazo , Estudios Prospectivos
14.
Medicina (Kaunas) ; 58(3)2022 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-35334523

RESUMEN

Background and Objectives: In this study, we aimed to investigate the link between common -2548G>A (rs7799039) promoter variant of the human leptin gene (LEP) with leptin and serum glucose leptin levels in obese Saudi patients. Materials and Methods: A total of 206 Saudi adults (80 obese normotensive nondiabetics, 76 obese hypertensive with Type 2 Diabetes and 50 normotensive nondiabetic controls) were genotyped for -2548G>A LEP polymorphism using the polymerase chain reaction-restriction fragment-length polymorphism technique. Results: Participants with minor AA genotype had significantly higher blood glucose levels (6.8 ± 0.55 mmol/L vs. 5.8 ± 0.30 mmol/L; p < 0.04) and HOMA-IR (4.1 ± 0.84 vs. 2.6 ± 0.67; p = 0.03) against those carrying major GG genotype. Participants with heterozygous GA genotype had significantly higher serum leptin levels against those carrying major GG genotype (40.0 ± 2.6 ng/mL vs. 29.6 ± 2.6 ng/mL; p = 0.04). Further investigation showed that individuals with AA, GA, GA + AA genotypes are at greater risk of developing hyperglycemia compared to those with GG genotype [OR 3.7(1.6−8.4), p = 0.001; 3.2 (1.2−8.6), p = 0.03; 3.5 (1.6−7.7), p = 0.001, respectively]. Additionally, the -2548AA allele was shown to be a risk factor for hyperglycemia [OR 1.9 (1.2−3.0), p = 0.006]. Our data revealed no relationship between this variant of the LEP gene with systolic and diastolic BP, signifying that this genetic variant is not a significant marker of obesity and hypertension in the Saudi population. Conclusions: AA and GA genotypes and LEP gene -2548AA alleles may signify potent risk factors predisposing healthy individuals to develop T2DM regardless of blood-pressure profile.


Asunto(s)
Diabetes Mellitus Tipo 2 , Leptina/genética , Adulto , Presión Sanguínea , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/genética , Frecuencia de los Genes , Glucosa , Humanos , Obesidad/complicaciones , Obesidad/genética , Polimorfismo de Nucleótido Simple , Arabia Saudita/epidemiología
15.
Exp Clin Endocrinol Diabetes ; 130(1): 22-29, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33036033

RESUMEN

Let7b-5p is a member of the Let-7 miRNA family and one of the top expressed miRNAs in human islets that implicated in glucose homeostasis. The levels of Let7b-5p in type 2 diabetes (T2DM) patients or its role in ß-cell function is still unclear. In the current study, we measured the serum levels of let7b-5p in Emirati patients with T2DM (with/without complications) and control subjects. Overexpression or silencing of let7b-5p in INS-1 (832/13) cells was performed to investigate the impact on insulin secretion, content, cell viability, apoptosis, and key functional genes. We found that serum levels of let7b-5p are significantly (p<0.05) higher in T2DM-patients or T2DM with complications compared to control subjects. Overexpression of let7b-5p increased insulin content and decreased glucose-stimulated insulin secretion, whereas silencing of let7b-5p reduced insulin content and secretion. Modulation of the expression levels of let7b-5p did not influence cell viability nor apoptosis. Analysis of mRNA and protein expression of hallmark genes in let7b-5p transfected cells revealed a marked dysregulation of Insulin, Pancreatic And Duodenal Homeobox 1 (PDX1), glucokinase (GCK), glucose transporter 2 (GLUT2), and INSR. In conclusion, an appropriate level of let7b-5p is essential to maintain ß-cell function and may be regarded as a biomarker for T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Regulación de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , MicroARNs/metabolismo , Humanos , Emiratos Árabes Unidos , Regulación hacia Arriba
16.
Pediatr Res ; 91(3): 539-544, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-33824452

RESUMEN

BACKGROUND: This study aimed to assess the telomere length and plasma telomere repeat-binding factor 2 (TRF2) levels in addition to other inflammatory markers in children with sickle cell disease (SCD). METHODS: We enrolled 106 children (90 SCD and 26 controls) aged 1-15 years from the Hematology unit of King Fahad Medical City (KFMC), Saudi Arabia. Genomic DNA extracted from blood and leukocyte TL was determined using quantitative reverse transcription PCR, whereas TRF2, C-reactive protein, interleukin-6, and DNA oxidative damage were determined by using respective commercially available assays. RESULTS: Leukocyte TL was inversely correlated with age in the SCD patients (r = -0.24, P = 0.02) and the controls (r = -0.68, P < 0.0001). In addition, SCD patients had significantly shorter TL (7.74 ± 0.81 kb) (P = 0.003) than controls (8.28 ± 0.73 kb). In contrast, no significant difference in TL among the SCD genotypes (HbSS and HbSß0) has been observed. A modest, positive correlation was seen between TL and reticulocyte % (r = 0.21; P = 0.06). There were no significant differences in the TL and TRF2 concentrations between subjects with HbSS and HbSß0 genotypes. CONCLUSIONS: Short leukocyte TL was significantly associated with SCD. An inverse association was observed between TL and hemoglobin. Hydroxyurea treatment revealed no impact on TL. IMPACT: This study explored the TL and plasma TRF2 in Saudi children with SCD. This is the first documentation that SCD children have shorter TL than their healthy counterparts, and no association between TL and TRF2 has been observed. Hydroxyurea treatment showed no impact on TL in children with SCD. This study is the first of its kind in children with SCD. It will pave the way for another study with a larger sample size in a diverse population to scrutinize these findings better.


Asunto(s)
Anemia de Células Falciformes , Hidroxiurea , Anemia de Células Falciformes/tratamiento farmacológico , Anemia de Células Falciformes/genética , Biomarcadores , Niño , Humanos , Hidroxiurea/uso terapéutico , Leucocitos , Proteínas de Unión a Telómeros
17.
Sci Rep ; 11(1): 20692, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34667273

RESUMEN

Copine 3 (CPNE3) is a calcium-dependent phospholipid-binding protein that has been found to play an essential role in cancer progression and stages. However, its role in pancreatic ß-cell function has not been investigated. Therefore, we performed a serial of bioinformatics and functional experiments to explore the potential role of Cpne3 on insulin secretion and ß-cell function in human islets and INS-1 (832/13) cells. RNA sequencing and microarray data revealed that CPNE3 is highly expressed in human islets compared to other CPNE genes. In addition, expression of CPNE3 was inversely correlated with HbA1c and reduced in human islets from hyperglycemic donors. Silencing of Cpne3 in INS-1 cells impaired glucose-stimulated insulin secretion (GSIS), insulin content and glucose uptake efficiency without affecting cell viability or inducing apoptosis. Moreover, mRNA and protein expression of the key regulators in glucose sensing and insulin secretion (Insulin, GLUT2, NeuroD1, and INSR) were downregulated in Cpne3-silenced cells. Taken together, data from the present study provides a new understanding of the role of CPNE3 in maintaining normal ß-cell function, which might contribute to developing a novel target for future management of type 2 diabetes therapy.


Asunto(s)
Proteínas Portadoras/metabolismo , Glucosa/metabolismo , Secreción de Insulina/fisiología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Humanos
18.
Curr Issues Mol Biol ; 43(3): 1325-1334, 2021 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-34698098

RESUMEN

Post-menopausal osteoporosis (PMO) is a multifactorial bone disorder in elderly women. Various vitamin D receptor (VDR) gene variants have been studied and associated with osteoporosis in other populations, but not in a homogenous Arab ethnic group. Herein, the current study explores the association between VDR polymorphisms and susceptibility to osteoporosis in Saudi postmenopausal women. In total, 600 Saudi postmenopausal women (N = 300 osteoporosis; N = 300 control) were genotyped for VDR gene variants (rs7975232, rs1544410, rs731236) using TaqMan® SNP genotyping assays. Bone mineral density (BMD) for the lumbar spine and femur was assessed using dual-energy X-ray absorptiometry (DEXA). The heterozygous frequency distributions AC of rs7975232, CT of rs1544410, and AG of rs731236 were significantly higher in the osteoporosis group than controls (p < 0.05). Heterozygous AC of rs7975232 (1.6; 95% CI 1.1-2.3; p < 0.023), CT of rs1544410 (1.6; 95% CI 1.1-2.4; p < 0.022), and AG of rs731236 (1.6; 95% CI 1.1-2.4; p < 0.024) were significantly associated with increased risk of osteoporosis, independent of age and BMI. In conclusion, VDR gene variants rs7975232, rs1544410, rs731236 had a significant effect on BMD and were associated with osteoporosis risk in Saudi postmenopausal women.


Asunto(s)
Variación Genética , Osteoporosis/etiología , Posmenopausia , Receptores de Calcitriol/genética , Anciano , Alelos , Densidad Ósea , Estudios de Casos y Controles , Femenino , Genotipo , Humanos , Persona de Mediana Edad , Osteoporosis/diagnóstico , Osteoporosis/metabolismo , Polimorfismo de Nucleótido Simple , Receptores de Calcitriol/metabolismo
19.
Mol Cell Endocrinol ; 538: 111462, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34547407

RESUMEN

Iron overload promotes the generation of reactive oxygen species (ROS). Pancreatic ß-cells can counter oxidative stress through multiple anti-oxidant responses. Herein, RNA-sequencing was used to describe the expression profile of iron regulatory genes in human islets with or without diabetes. Functional experiments including siRNA silencing, qPCR, western blotting, cell viability, ELISA and RNA-sequencing were performed as means of identifying the genetic signature of the protective response following iron overload-induced stress in human islets and INS-1. FTH1 and FTL genes were highly expressed in human islets and INS-1 cells, while hepcidin (HAMP) was low. FXN, DMT1 and FTHL1 genes were differentially expressed in diabetic islets compared to control. Silencing of Hamp in INS-1 cells impaired insulin secretion and influenced the expression of ß-cell key genes. RNA-sequencing analysis in iron overloaded INS-1 cells identified Id1 and Id3 as the top down-regulated genes, while Hmox1 was the top upregulated. Expression of ID1, ID3 and HMOX1 was validated at the protein level in INS-1 cells and human islets. Differentially expressed genes (DEGs) were enriched for TGF-ß, regulating stem cells, ferroptosis, and HIF-1 signaling. Hmox1-silenced cells treated with FAC elevated the expression of Id1 and Id3 expression than untreated cells. Our findings suggest that HMOX1, ID1 and ID3 define the response mechanism against iron-overload-induced stress in ß-cells.


Asunto(s)
Hemo-Oxigenasa 1/genética , Hiperglucemia/genética , Proteína 1 Inhibidora de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/genética , Sobrecarga de Hierro/genética , Proteínas de Neoplasias/genética , Animales , Apoferritinas/genética , Apoferritinas/metabolismo , Cadáver , Estudios de Casos y Controles , Células Cultivadas , Ferritinas/genética , Ferritinas/metabolismo , Técnicas de Silenciamiento del Gen , Hemo-Oxigenasa 1/metabolismo , Hepcidinas/genética , Hepcidinas/metabolismo , Humanos , Hiperglucemia/metabolismo , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Proteínas Inhibidoras de la Diferenciación/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Sobrecarga de Hierro/metabolismo , Proteínas de Unión a Hierro/genética , Proteínas de Unión a Hierro/metabolismo , Proteínas de Neoplasias/metabolismo , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Ratas , Regulación hacia Arriba , Frataxina
20.
Diabetes Metab Syndr Obes ; 14: 3389-3397, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34345175

RESUMEN

INTRODUCTION: Previous studies have suggested the involvement of chronic low-grade inflammation in the pathogenesis of diabetic neuropathy (DNP). However, none of these studies have examined the levels of monocyte chemoattractant protein-1 (MCP-1) in type 2 diabetes mellitus (T2DM) patients with confirmed diagnosis of neuropathy. Therefore, the present study aims to investigate the levels of MCP-1 along with IL-6, IL-8 and TGF-ß in patients with T2DM and confirmed neuropathy and identify correlations, if any, between MCP-1 and other parameters. METHODS: A single center cross-sectional clinical study was conducted at University Hospital Sharjah (UHS) and University of Sharjah. One hundred and two patients with T2DM were recruited from diabetes clinics at UHS and were stratified into different groups based on diagnosis of DNP and other parameters. Several analyses were conducted to evaluate and compare the levels of MCP-1, IL-6, IL-8, and TGF-ß across these groups of patients and identify correlations, if any, between MCP-1 and other variables. RESULTS: A significant increase was found in the levels of MCP-1 in T2DM patients with DNP compared to the patients without DNP (p=0.002, p-adj=0.007). Further analysis has shown that levels of IL-8 (p=0.008) and TGF-ß (p=0.06) were increased and decreased, respectively, in patients with DNP compared to patients without DNP. Moreover, strong correlations were found between MCP-1, IL-8 and TGF-ß levels. CONCLUSION: The key finding of the present study is the significant elevation in levels of MCP-1 in T2DM patients with DNP compared to the patients without DNP and IL-8 and TGF-ß were strong predictors of MCP-1 increased levels.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...