Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 486
Filtrar
1.
Biol Open ; 13(5)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38809145

RESUMEN

Bone is increasingly recognized as a target for diabetic complications. In order to evaluate the direct effects of high glucose on bone, we investigated the global transcriptional changes induced by hyperglycemia in osteoblasts in vitro. Rat bone marrow-derived mesenchymal stromal cells were differentiated into osteoblasts for 10 days, and prior to analysis, they were exposed to hyperglycemia (25 mM) for the short-term (1 or 3 days) or long-term (10 days). Genes and pathways regulated by hyperglycemia were identified using mRNA sequencing and verified with qPCR. Genes upregulated by 1-day hyperglycemia were, for example, related to extracellular matrix organization, collagen synthesis and bone formation. This stimulatory effect was attenuated by 3 days. Long-term exposure impaired osteoblast viability, and downregulated, for example, extracellular matrix organization and lysosomal pathways, and increased intracellular oxidative stress. Interestingly, transcriptional changes by different exposure times were mostly unique and only 89 common genes responding to glucose were identified. In conclusion, short-term hyperglycemia had a stimulatory effect on osteoblasts and bone formation, whereas long-term hyperglycemia had a negative effect on intracellular redox balance, osteoblast viability and function.


Asunto(s)
Regulación de la Expresión Génica , Glucosa , Osteoblastos , Osteoblastos/metabolismo , Osteoblastos/efectos de los fármacos , Animales , Glucosa/metabolismo , Ratas , Regulación de la Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Hiperglucemia/metabolismo , Hiperglucemia/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Transcriptoma , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Supervivencia Celular/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Células Cultivadas , Estrés Oxidativo/efectos de los fármacos
2.
Biol Open ; 13(5)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38742438

RESUMEN

Bone is increasingly recognized as a target for diabetic complications. In order to evaluate the direct effects of high glucose on bone, we investigated the global transcriptional changes induced by hyperglycemia in osteoblasts in vitro. Rat bone marrow-derived mesenchymal stromal cells were differentiated into osteoblasts for 10 days, and prior to analysis, they were exposed to hyperglycemia (25 mM) for the short-term (1 or 3 days) or long-term (10 days). Genes and pathways regulated by hyperglycemia were identified using mRNA sequencing and verified with qPCR. Genes upregulated by 1-day hyperglycemia were, for example, related to extracellular matrix organization, collagen synthesis and bone formation. This stimulatory effect was attenuated by 3 days. Long-term exposure impaired osteoblast viability, and downregulated, for example, extracellular matrix organization and lysosomal pathways, and increased intracellular oxidative stress. Interestingly, transcriptional changes by different exposure times were mostly unique and only 89 common genes responding to glucose were identified. In conclusion, short-term hyperglycemia had a stimulatory effect on osteoblasts and bone formation, whereas long-term hyperglycemia had a negative effect on intracellular redox balance, osteoblast viability and function.


Asunto(s)
Regulación de la Expresión Génica , Glucosa , Osteoblastos , Osteoblastos/metabolismo , Osteoblastos/efectos de los fármacos , Animales , Glucosa/metabolismo , Ratas , Regulación de la Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Hiperglucemia/metabolismo , Hiperglucemia/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Transcriptoma , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Supervivencia Celular/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Células Cultivadas , Estrés Oxidativo/efectos de los fármacos
3.
J Cell Mol Med ; 28(9): e18336, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38686489

RESUMEN

Diabetic kidney disease (DKD), a primary microvascular complication arising from diabetes, may result in end-stage renal disease. Epigenetic regulation of endothelial mesenchymal transition (EndMT) has been recently reported to exert function in metabolic memory and DKD. Here, we investigated the mechanism which Sirt7 modulated EndMT in human glomerular endothelial cells (HGECs) in the occurrence of metabolic memory in DKD. Lower levels of SDC1 and Sirt7 were noted in the glomeruli of both DKD patients and diabetes-induced renal injury rats, as well as in human glomerular endothelial cells (HGECs) with high blood sugar. Endothelial-to-mesenchymal transition (EndMT) was sustained despite the normalization of glycaemic control. We also found that Sirt7 overexpression associated with glucose normalization promoted the SDC1 expression and reversed EndMT in HGECs. Furthermore, the sh-Sirt7-mediated EndMT could be reversed by SDC1 overexpression. The ChIP assay revealed enrichment of Sirt7 and H3K18ac in the SDC1 promoter region. Furthermore, hypermethylated in cancer 1 (HIC1) was found to be associated with Sirt7. Overexpression of HIC1 with normoglycaemia reversed high glucose-mediated EndMT in HGECs. The knockdown of HIC1-mediated EndMT was reversed by SDC1 upregulation. In addition, the enrichment of HIC1 and Sirt7 was observed in the same promoter region of SDC1. The overexpressed Sirt7 reversed EndMT and improved renal function in insulin-treated diabetic models. This study demonstrated that the hyperglycaemia-mediated interaction between Sirt7 and HIC1 exerts a role in the metabolic memory in DKD by inactivating SDC1 transcription and mediating EndMT despite glucose normalization in HGECs.


Asunto(s)
Nefropatías Diabéticas , Células Endoteliales , Hiperglucemia , Factores de Transcripción de Tipo Kruppel , Sirtuinas , Sindecano-1 , Sindecano-1/metabolismo , Sindecano-1/genética , Humanos , Animales , Hiperglucemia/metabolismo , Hiperglucemia/genética , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Ratas , Masculino , Células Endoteliales/metabolismo , Sirtuinas/metabolismo , Sirtuinas/genética , Transición Epitelial-Mesenquimal/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/complicaciones , Ratas Sprague-Dawley , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Epigénesis Genética , Regulación de la Expresión Génica , Regiones Promotoras Genéticas , Transición Endotelial-Mesenquimatosa
4.
Sci Rep ; 14(1): 9100, 2024 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-38643275

RESUMEN

Diabetes constitutes a major public health problem, with dramatic consequences for patients. Both genetic and environmental factors were shown to contribute to the different forms of the disease. The monogenic forms, found both in humans and in animal models, specially help to decipher the role of key genes in the physiopathology of the disease. Here, we describe the phenotype of early diabetes in a colony of NOD mice, with spontaneous invalidation of Akt2, that we called HYP. The HYP mice were characterised by a strong and chronic hyperglycaemia, beginning around the age of one month, especially in male mice. The phenotype was not the consequence of the acceleration of the autoimmune response, inherent to the NOD background. Interestingly, in HYP mice, we observed hyperinsulinemia before hyperglycaemia occurred. We did not find any difference in the pancreas' architecture of the NOD and HYP mice (islets' size and staining for insulin and glucagon) but we detected a lower insulin content in the pancreas of HYP mice compared to NOD mice. These results give new insights about the role played by Akt2 in glucose homeostasis and argue for the ß cell failure being the primary event in the course of diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1 , Hiperglucemia , Islotes Pancreáticos , Animales , Humanos , Masculino , Ratones , Diabetes Mellitus Tipo 1/genética , Hiperglucemia/genética , Insulina , Islotes Pancreáticos/patología , Ratones Endogámicos NOD , Páncreas/patología , Proteínas Proto-Oncogénicas c-akt/genética
5.
J Cell Physiol ; 239(5): e31211, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38304971

RESUMEN

Cataract, a leading cause of blindness, is characterised by lens opacification. Type 2 diabetes is associated with a two- to fivefold higher prevalence of cataracts. The risk of cataract formation increases with the duration of diabetes and the severity of hyperglycaemia. Hydroxyapatite deposition is present in cataractous lenses that could be the consequence of osteogenic differentiation and calcification of lens epithelial cells (LECs). We hypothesised that hyperglycaemia might promote the osteogenic differentiation of human LECs (HuLECs). Osteogenic medium (OM) containing excess phosphate and calcium with normal (1 g/L) or high (4.5 g/L) glucose was used to induce HuLEC calcification. High glucose accelerated and intensified OM-induced calcification of HuLECs, which was accompanied by hyperglycaemia-induced upregulation of the osteogenic markers Runx2, Sox9, alkaline phosphatase and osteocalcin, as well as nuclear translocation of Runx2. High glucose-induced calcification was abolished in Runx2-deficient HuLECs. Additionally, high glucose stabilised the regulatory alpha subunits of hypoxia-inducible factor 1 (HIF-1), triggered nuclear translocation of HIF-1α and increased the expression of HIF-1 target genes. Gene silencing of HIF-1α or HIF-2α attenuated hyperglycaemia-induced calcification of HuLECs, while hypoxia mimetics (desferrioxamine, CoCl2) enhanced calcification of HuLECs under normal glucose conditions. Overall, this study suggests that high glucose promotes HuLEC calcification via Runx2 and the activation of the HIF-1 signalling pathway. These findings may provide new insights into the pathogenesis of diabetic cataracts, shedding light on potential factors for intervention to treat this sight-threatening condition.


Asunto(s)
Diferenciación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal , Células Epiteliales , Glucosa , Subunidad alfa del Factor 1 Inducible por Hipoxia , Cristalino , Osteogénesis , Humanos , Cristalino/metabolismo , Cristalino/patología , Diferenciación Celular/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Glucosa/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Osteocalcina/metabolismo , Osteocalcina/genética , Catarata/patología , Catarata/metabolismo , Catarata/genética , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Calcinosis/metabolismo , Calcinosis/patología , Calcinosis/genética , Hiperglucemia/metabolismo , Hiperglucemia/genética , Hiperglucemia/patología , Transducción de Señal , Fosfatasa Alcalina/metabolismo , Fosfatasa Alcalina/genética , Células Cultivadas
6.
Acta Diabetol ; 61(4): 515-524, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38244081

RESUMEN

AIMS: Diabetic osteoporosis (DOP) is the most common secondary form of osteoporosis. Diabetes mellitus affects bone metabolism; however, the underlying pathophysiological mechanisms remain unclear. Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) expression is upregulated in conditions characterized by vascular injury, such as atherosclerosis, hypertension, and diabetes. Additionally, Notch, HIF-1α, and VEGF are involved in angiogenesis and bone formation. Therefore, we aimed to investigate the expression of Notch, HIF-1α, and VEGF in the LOX-1 silencing state. METHODS: Rat bone H-type vascular endothelial cells (THVECs) were isolated and cultured in vitro. Cell identification was performed using immunofluorescent co-expression of CD31 and Emcn. Lentiviral silencing vector (LV-LOX-1) targeting LOX-1 was constructed using genetic recombination technology and transfected into the cells. The experimental groups included the following: NC group, HG group, LV-LOX-1 group, LV-CON group, HG + LV-LOX-1 group, HG + LV-CON group, HG + LV-LOX-1 + FLI-06 group, HG + LV-CON + FLI-06 group, HG + LV-LOX-1 + LW6 group, and HG + LV-CON + LW6 group. The levels of LOX-1, Notch, Hif-1α, and VEGF were detected using PCR and WB techniques to investigate whether the expression of LOX-1 under high glucose conditions has a regulatory effect on downstream molecules at the gene and protein levels, as well as the specific molecular mechanisms involved. RESULTS: High glucose (HG) conditions led to a significant increase in LOX-1 expression, leading to inhibition of angiogenesis, whereas silencing LOX-1 can reverse this phenomenon. Further analysis reveals that changes in LOX-1 will promote changes in Notch/HIF-1α and VEGF. Moreover, Notch mediates the activation of HIF-1α and VEGF. CONCLUSIONS: The activation of LOX-1 and the inhibition of Notch/HIF-1α/VEGF in THVECs are the main causes of DOP. These findings contribute to our understanding of the pathogenesis of DOP and offer a novel approach for preventing and treating osteoporosis.


Asunto(s)
Diabetes Mellitus , Hiperglucemia , Osteoporosis , Animales , Ratas , Células Endoteliales/metabolismo , Glucosa , Hiperglucemia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Receptores Depuradores de Clase E/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
Int J Epidemiol ; 53(1)2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38205890

RESUMEN

BACKGROUND: Diabetes (regardless of type) and obesity are associated with a range of musculoskeletal disorders. The causal mechanisms driving these associations are unknown for many upper limb pathologies. We used genetic techniques to test the causal link between glycemia, obesity and musculoskeletal conditions. METHODS: In the UK Biobank's unrelated European cohort (N = 379 708) we performed mendelian randomisation (MR) analyses to test for a causal effect of long-term high glycaemia and adiposity on four musculoskeletal pathologies: frozen shoulder, Dupuytren's disease, carpal tunnel syndrome and trigger finger. We also performed single-gene MR using rare variants in the GCK gene. RESULTS: Using MR, we found evidence that long-term high glycaemia has a causal role in the aetiology of upper limb conditions. A 10-mmol/mol increase in genetically predicted haemoglobin A1C (HbA1c) was associated with frozen shoulder: odds ratio (OR) = 1.50 [95% confidence interval (CI), 1.20-1.88], Dupuytren's disease: OR = 1.17 (95% CI, 1.01-1.35), trigger finger: OR = 1.30 (95% CI, 1.09-1.55) and carpal tunnel syndrome: OR = 1.20 (95% CI, 1.09-1.33). Carriers of GCK mutations have increased odds of frozen shoulder: OR = 7.16 (95% CI, 2.93-17.51) and carpal tunnel syndrome: OR = 2.86 (95% CI, 1.50-5.44) but not Dupuytren's disease or trigger finger. We found evidence that an increase in genetically predicted body mass index (BMI) of 5 kg/m2 was associated with carpal tunnel syndrome: OR = 1.13 (95% CI, 1.10-1.16) and associated negatively with Dupuytren's disease: OR = 0.94 (95% CI, 0.90-0.98), but no evidence of association with frozen shoulder or trigger finger. Trigger finger (OR 1.96 (95% CI, 1.42-2.69) P = 3.6e-05) and carpal tunnel syndrome [OR 1.63 (95% CI, 1.36-1.95) P = 8.5e-08] are associated with genetically predicted unfavourable adiposity increase of one standard deviation of body fat. CONCLUSIONS: Our study consistently demonstrates a causal role of long-term high glycaemia in the aetiology of upper limb musculoskeletal conditions. Clinicians treating diabetes patients should be aware of these complications in clinic, specifically those managing the care of GCK mutation carriers. Upper limb musculoskeletal conditions should be considered diabetes complications.


Asunto(s)
Bursitis , Síndrome del Túnel Carpiano , Diabetes Mellitus , Contractura de Dupuytren , Hiperglucemia , Enfermedades Musculoesqueléticas , Trastorno del Dedo en Gatillo , Humanos , Contractura de Dupuytren/epidemiología , Contractura de Dupuytren/genética , Contractura de Dupuytren/complicaciones , Síndrome del Túnel Carpiano/epidemiología , Síndrome del Túnel Carpiano/genética , Síndrome del Túnel Carpiano/complicaciones , Trastorno del Dedo en Gatillo/complicaciones , Hiperglucemia/complicaciones , Hiperglucemia/epidemiología , Hiperglucemia/genética , Extremidad Superior , Enfermedades Musculoesqueléticas/complicaciones , Factores de Riesgo , Bursitis/complicaciones , Obesidad/complicaciones , Obesidad/epidemiología , Obesidad/genética
8.
Metabolism ; 152: 155766, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38145825

RESUMEN

BACKGROUND: Excessive hepatic glucose production is a hallmark that contributes to hyperglycemia in type 2 diabetes (T2D). The regulatory network governing this process remains incompletely understood. Here, we demonstrate that TOX3, a high-mobility group family member, acts as a major transcriptional driver for hepatic glucose production. METHODS: Tox3-overexpressed and knockout mice were constructed to explore its metabolic functions. Transcriptomic and chromatin-immunoprecipitation sequencing (ChIP-seq) were used to identify downstream targets of TOX3. Both FoxO1 silencing and inhibitor approaches were used to assess the contribution of FoxO1. TOX3 expression levels were examined in the livers of mice and human subjects. Finally, Tox3 was genetically manipulated in diet-induced obese mice to evaluate its therapeutic potential. RESULTS: Hepatic Tox3 overexpression activates the gluconeogenic program, resulting in hyperglycemia and insulin resistance in mice. Hepatocyte-specific Tox3 knockout suppresses gluconeogenesis and improves insulin sensitivity. Mechanistically, integrated hepatic transcriptomic and ChIP-seq analyses identify FoxO1 as a direct target of TOX3. TOX3 stimulates FoxO1 transcription by directly binding to and activating its promoter, whereas FoxO1 silencing abrogates TOX3-induced dysglycemia in mice. In human subjects, hepatic TOX3 expression shows a significant positive correlation with blood glucose levels under normoglycemic conditions, yet is repressed by high glucose during T2D. Importantly, hepatic Tox3 deficiency markedly protects against and ameliorates the hyperglycemia and glucose intolerance in diet-induced diabetic mice. CONCLUSIONS: Our findings establish TOX3 as a driver for excessive gluconeogenesis through activating hepatic FoxO1 transcription. TOX3 could serve as a promising target for preventing and treating hyperglycemia in T2D.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Hiperglucemia , Resistencia a la Insulina , Animales , Humanos , Ratones , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Gluconeogénesis/genética , Glucosa/metabolismo , Hiperglucemia/genética , Hiperglucemia/metabolismo , Hígado/metabolismo , Ratones Endogámicos C57BL
9.
Cell Death Dis ; 14(12): 825, 2023 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-38092733

RESUMEN

Chronic hyperglycaemia is a devastating factor that causes diabetes-induced damage to the retina and kidney. However, the precise mechanism by which hyperglycaemia drives apoptotic cell death is incompletely known. Herein, we found that FOXD1, a FOX family transcription factor specifically expressed in the retina and kidney, regulated the transcription of BCL-2, a master regulator of cell survival. Intriguingly, the protein level of FOXD1, which responded negatively to hyperglycaemic conditions, was controlled by the TRIM21-mediated K48-linked polyubiquitination and subsequent proteasomal degradation. The TRIM21-FOXD1-BCL-2 signalling axis was notably active during diabetes-induced damage to murine retinal and renal tissues. Furthermore, we found that tartary buckwheat flavonoids effectively reversed the downregulation of FOXD1 protein expression and thus restored BCL-2 expression and facilitated the survival of retinal and renal tissues. In summary, we identified a transcription factor responsible for BCL-2 expression, a signalling axis (TRM21-FOXD1-BCL-2) underlying hyperglycaemia-triggered apoptosis, and a potential treatment for deleterious diabetic complications.


Asunto(s)
Diabetes Mellitus , Hiperglucemia , Animales , Ratones , Apoptosis/genética , Diabetes Mellitus/genética , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Hiperglucemia/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética
10.
J Clin Invest ; 134(3)2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-38032734

RESUMEN

Pancreatic ß cells are specialized for coupling glucose metabolism to insulin peptide production and secretion. Acute glucose exposure robustly and coordinately increases translation of proinsulin and proteins required for secretion of mature insulin peptide. By contrast, chronically elevated glucose levels that occur during diabetes impair ß cell insulin secretion and have been shown experimentally to suppress insulin translation. Whether translation of other genes critical for insulin secretion is similarly downregulated by chronic high glucose is unknown. Here, we used high-throughput ribosome profiling and nascent proteomics in MIN6 insulinoma cells to elucidate the genome-wide impact of sustained high glucose on ß cell mRNA translation. Before induction of ER stress or suppression of global translation, sustained high glucose suppressed glucose-stimulated insulin secretion and downregulated translation of not only insulin, but also mRNAs related to insulin secretory granule formation, exocytosis, and metabolism-coupled insulin secretion. Translation of these mRNAs was also downregulated in primary rat and human islets following ex vivo incubation with sustained high glucose and in an in vivo model of chronic mild hyperglycemia. Furthermore, translational downregulation decreased cellular abundance of these proteins. Our study uncovered a translational regulatory circuit during ß cell glucose toxicity that impairs expression of proteins with critical roles in ß cell function.


Asunto(s)
Hiperglucemia , Células Secretoras de Insulina , Islotes Pancreáticos , Neoplasias Pancreáticas , Ratas , Humanos , Animales , Secreción de Insulina , ARN Mensajero/metabolismo , Insulina/metabolismo , Hiperglucemia/genética , Hiperglucemia/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Péptidos/metabolismo , Neoplasias Pancreáticas/metabolismo , Islotes Pancreáticos/metabolismo
11.
Mol Med Rep ; 28(5)2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37711034

RESUMEN

Exosomal microRNAs (miRNAs/miRs) are potential biomarkers for the diagnosis and treatment of cardiovascular disease, and hyperglycemia serves an important role in the development of atherosclerosis. The present study aimed to investigate the expression profile of serum­derived exosomal miRNAs in coronary heart disease (CHD) with hyperglycemia, and to identify effective biomarkers for predicting coronary artery lesions. Serum samples were collected from eight patients with CHD and hyperglycemia and eight patients with CHD and normoglycemia, exosomes were isolated and differentially expressed miRNAs (DEMIs) were filtered using a human miRNA microarray. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed using standard enrichment computational methods for the target genes of DEMIs. Receiver operating characteristic (ROC) curve analysis was applied to evaluate the values of the selected DEMIs in predicting the severity of coronary stenosis. A total of 10 DEMIs, including four upregulated miRNAs (hsa­let­7b­5p, hsa­miR­4313, hsa­miR­4665­3p and hsa­miR­940) and six downregulated miRNAs (hsa­miR­4459, hsa­miR­4687­3p, hsa­miR­6087, hsa­miR­6089, hsa­miR­6740­5p and hsa­miR­6800­5p), were screened in patients with CHD and hyperglycemia. GO analysis showed that the 'cellular process', 'single­organism process' and 'biological regulation' were significantly enriched. KEGG pathway analysis revealed that the 'mTOR signaling pathway', 'FoxO signaling pathway' and 'neurotrophin signaling pathway' were significantly enriched. Among these DEMIs, only hsa­let­7b­5p expression was positively correlated with both hemoglobin A1C levels and Synergy between Percutaneous Coronary Intervention with Taxus and Cardiac Surgery score. ROC curves showed that hsa­let­7b­5p could serve as an effective biomarker for differentiating the severity of coronary stenosis. In conclusion, the present study demonstrated that serum­derived exosomal hsa­let­7b­5p is upregulated in patients with CHD and hyperglycemia, and may serve as a noninvasive biomarker for the severity of coronary stenosis.


Asunto(s)
Aterosclerosis , Estenosis Coronaria , Hiperglucemia , MicroARNs , Humanos , Biomarcadores , Estenosis Coronaria/diagnóstico , Estenosis Coronaria/genética , Hiperglucemia/complicaciones , Hiperglucemia/genética , MicroARNs/genética
12.
Mol Med ; 29(1): 92, 2023 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-37415117

RESUMEN

BACKGROUND: Diabetic nephropathy (DN) is a major complication of diabetes mellitus. Clinical reports indicate that smoking is a significant risk factor for chronic kidney disease, and the tobacco epidemic exacerbates kidney damage in patients with DN. However, the underlying molecular mechanisms remain unclear. METHOD: In the present study, we used a diabetic mouse model to investigate the molecular mechanisms for nicotine-exacerbated DN. Twelve-week-old female mice were injected with streptozotocin (STZ) to establish a hyperglycemic diabetic model. After four months, the control and hyperglycemic diabetic mice were further divided into four groups (control, nicotine, diabetic mellitus, nicotine + diabetic mellitus) by intraperitoneal injection of nicotine or PBS. After two months, urine and blood were collected for kidney injury assay, and renal tissues were harvested for further molecular assays using RNA-seq analysis, real-time PCR, Western blot, and immunohistochemistry. In vitro studies, we used siRNA to suppress Grem1 expression in human podocytes. Then we treated them with nicotine and high glucose to compare podocyte injury. RESULT: Nicotine administration alone did not cause apparent kidney injury, but it significantly increased hyperglycemia-induced albuminuria, BUN, plasma creatinine, and the kidney tissue mRNA expression of KIM-1 and NGAL. Results from RNA-seq analysis, real-time PCR, Western blot, and immunohistochemistry analysis revealed that, compared to hyperglycemia or nicotine alone, the combination of nicotine treatment and hyperglycemia significantly increased the expression of Grem1 and worsened DN. In vitro experiments, suppression of Grem1 expression attenuated nicotine-exacerbated podocyte injury. CONCLUSION: Grem1 plays a vital role in nicotine-exacerbated DN. Grem1 may be a potential therapeutic target for chronic smokers with DN.


Asunto(s)
Diabetes Mellitus Experimental , Nefropatías Diabéticas , Hiperglucemia , Humanos , Ratones , Femenino , Animales , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/inducido químicamente , Regulación hacia Arriba , Nicotina/efectos adversos , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inducido químicamente , Hiperglucemia/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo
13.
Int J Mol Sci ; 24(14)2023 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-37511575

RESUMEN

Diabetes mellitus is a burdensome disease that affects various cellular functions through altered glucose metabolism. Several reports have linked diabetes to cancer development; however, the exact molecular mechanism of how diabetes-related traits contribute to cancer progression is not fully understood. The current study aimed to explore the molecular mechanism underlying the potential effect of hyperglycemia combined with hyperinsulinemia on the progression of breast cancer cells. To this end, gene dysregulation induced by the exposure of MCF7 breast cancer cells to hyperglycemia (HG), or a combination of hyperglycemia and hyperinsulinemia (HGI), was analyzed using a microarray gene expression assay. Hyperglycemia combined with hyperinsulinemia induced differential expression of 45 genes (greater than or equal to two-fold), which were not shared by other treatments. On the other hand, in silico analysis performed using a publicly available dataset (GEO: GSE150586) revealed differential upregulation of 15 genes in the breast tumor tissues of diabetic patients with breast cancer when compared with breast cancer patients with no diabetes. SLC26A11, ALDH1A3, MED20, PABPC4 and SCP2 were among the top upregulated genes in both microarray data and the in silico analysis. In conclusion, hyperglycemia combined with hyperinsulinemia caused a likely unique signature that contributes to acquiring more carcinogenic traits. Indeed, these findings might potentially add emphasis on how monitoring diabetes-related metabolic alteration as an adjunct to diabetes therapy is important in improving breast cancer outcomes. However, further detailed studies are required to decipher the role of the highlighted genes, in this study, in the pathogenesis of breast cancer in patients with a different glycemic index.


Asunto(s)
Neoplasias de la Mama , Diabetes Mellitus Tipo 2 , Diabetes Mellitus , Hiperglucemia , Hiperinsulinismo , Humanos , Femenino , Neoplasias de la Mama/genética , Hiperglucemia/complicaciones , Hiperglucemia/genética , Hiperglucemia/metabolismo , Hiperinsulinismo/complicaciones , Hiperinsulinismo/genética , Hiperinsulinismo/metabolismo , Índice Glucémico , Diabetes Mellitus Tipo 2/patología
14.
J Mol Med (Berl) ; 101(8): 1015-1028, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37462767

RESUMEN

Multiple molecular pathways including the receptor for advanced glycation end-products-diaphanous related formin 1 (RAGE-Diaph1) signaling are known to play a role in diabetic peripheral neuropathy (DPN). Evidence suggests that neuropathological alterations in type 1 diabetic spinal cord may occur at the same time as or following peripheral nerve abnormalities. We demonstrated that DPN was associated with perturbations of RAGE-Diaph1 signaling pathway in peripheral nerve accompanied by widespread spinal cord molecular changes. More than 500 differentially expressed genes (DEGs) belonging to multiple functional pathways were identified in diabetic spinal cord and of those the most enriched was RAGE-Diaph1 related PI3K-Akt pathway. Only seven of spinal cord DEGs overlapped with DEGs from type 1 diabetic sciatic nerve and only a single gene cathepsin E (CTSE) was common for both type 1 and type 2 diabetic mice. In silico analysis suggests that molecular changes in spinal cord may act synergistically with RAGE-Diaph1 signaling axis in the peripheral nerve. KEY MESSAGES: Molecular perturbations in spinal cord may be involved in the progression of diabetic peripheral neuropathy. Diabetic peripheral neuropathy was associated with perturbations of RAGE-Diaph1 signaling pathway in peripheral nerve accompanied by widespread spinal cord molecular changes. In silico analysis revealed that PI3K-Akt signaling axis related to RAGE-Diaph1 was the most enriched biological pathway in diabetic spinal cord. Cathepsin E may be the target molecular hub for intervention against diabetic peripheral neuropathy.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Neuropatías Diabéticas , Hiperglucemia , Animales , Ratones , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Neuropatías Diabéticas/genética , Neuropatías Diabéticas/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/complicaciones , Catepsina E , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Nervio Ciático/patología , Hiperglucemia/genética , Hiperglucemia/patología
15.
J Agric Food Chem ; 71(24): 9324-9336, 2023 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-37294881

RESUMEN

This work aimed to investigate the hypoglycemic effects and underlying mechanism of whole grain proso millet (Panicum miliaceum L.; WPM) on type 2 diabetes mellitus (T2DM). The results showed that WPM supplementation significantly reduced fasting blood glucose (FBG) and serum lipid levels in T2DM mice induced by a high-fat diet (HFD) combined with streptozotocin (STZ), with improved glucose tolerance, liver and kidney injury, and insulin resistance. In addition, WPM significantly inhibited the expression of gluconeogenesis-related genes G6pase, Pepck, Foxo1, and Pgc-1α. Further study by miRNA high-throughput sequencing revealed that WPM supplementation mainly altered the liver miRNA expression profile of T2DM mice by increasing the expression of miR-144-3p_R-1 and miR-423-5p, reducing the expression of miR-22-5p_R-1 and miR-30a-3p. GO and KEGG analyses showed that the target genes of these miRNAs were mainly enriched in the PI3K/AKT signaling pathway. WPM supplementation significantly increased the level of PI3K, p-AKT, and GSK3ß in the liver of T2DM mice. Taken together, WPM exerts antidiabetic effects by improving the miRNA profile and activating the PI3K/AKT signaling pathway to inhibit gluconeogenesis. This study implies that PM can act as a dietary supplement to attenuate T2DM.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Hiperglucemia , MicroARNs , Panicum , Ratones , Animales , Panicum/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Granos Enteros , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/genética , Hiperglucemia/metabolismo , Hígado/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Hipoglucemiantes/metabolismo
16.
Exp Eye Res ; 233: 109519, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37277067

RESUMEN

Heparanase (HPA) is believed that might mediate histone 3 lysine 9 acetylation (H3K9ac) to regulate vascular endothelial growth factor (VEGF) gene expressions in the hyperglycemia and hypoxia human retinal endothelial cells (HRECs). Cultured human retinal endothelial cells (HRECs) in hyperglycemia, hypoxia, siRNA, and normal medium, respectively. Distributions of H3K9ac and HPA in HRECs were analyzed by immunofluorescence. Western blot and real-time PCR were respectively used to evaluate the expression of HPA, H3K9ac, and VEGF. The differences in occupancies of H3K9ac and RNA polymerase II at VEGF gene promoter among three groups were studied by Chromatin immunoprecipitation (ChIP) combined with real-time PCR. Co-immunoprecipitation (Co-IP) was used to measure the status of HPA and H3K9ac. Re-ChIP was used to verify whether HPA and H3K9ac associate to the transcription of VEGF gene. HPA was consistent with that of H3K9ac in the hyperglycemia and hypoxia groups. And the fluorescent lights of H3K9ac and HPA in siRNA groups were similar to the control group, fainter than that of hyperglycemia, hypoxia, and non-silencing groups. Western blot results showed that the expressions of HPA, H3K9ac, and VEGF in hyperglycemia and hypoxia HRECs were statistically higher than that of the control. HPA, H3K9ac, and VEGF expressions in siRNA groups were statistically lower than hyperglycemia and hypoxia HRECs. The same trends also were found in real-time PCR. ChIP exhibited the occupancies of H3K9ac and RNA Pol II at VEGF gene promoter in hyperglycemia and hypoxia groups were significantly more increased than in the control group. Co-IP revealed that HPA combined with H3K9ac in hyperglycemia and hypoxia groups; while it was not discovered in the control group. Re-ChIP showed that HPA combined with H3K9ac at VEGF gene promoter in the hyperglycemia and hypoxia HRECs nuclear. In our study HPA can influence expressions of H3K9ac and VEGF in the hyperglycemia and hypoxia HRECs. HPA can probably combine with H3K9ac and regulate the transcription of the VEGF gene in the hyperglycemia and hypoxia HRECs.


Asunto(s)
Células Endoteliales , Hiperglucemia , Humanos , Células Endoteliales/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Histonas/genética , Acetilación , Hiperglucemia/genética , Hiperglucemia/metabolismo , Células Cultivadas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transcripción Genética , Hipoxia/genética , Hipoxia/metabolismo
17.
Ophthalmic Res ; 66(1): 913-920, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37062273

RESUMEN

INTRODUCTION: Vascular endothelial cell injury and angiogenesis induced by hyperglycemia are the main pathological basis of vascular complications in diabetes mellitus. Our study aimed to investigate the role and mechanism of miR-210-3p in high glucose (HG)-induced angiogenesis. METHODS: Human umbilical vein endothelial cells (HUVECs) were treated with HG to mimic the pathological process of hyperglycemia. HUVECs were divided into the control group, HG group, HG+inhibitor-NC group, and HG+miR-210-3p inhibitor group. Proliferation and migration were tested by wound healing assay, tube formation, and Transwell assay. Quantitation real-time PCR and Western blots were performed to determine the expression of miR-210-3p and relative proteins, respectively. RESULTS: The level of miR-210-3p significantly increased in HUVECs treated by HG. The knockdown of miR-210-3p attenuated the tube formation, proliferation, and migration of cultured HUVECs in vitro to inhibit angiogenesis by increasing the expression of fibroblast growth factor receptor-like 1 (FGFRL1) and then attenuating the phosphorylation of signal transducer and activator of transcription 3 (STAT3), extracellular regulated protein kinases, and protein kinase B (Akt). CONCLUSION: Our study revealed that miR-210-3p might be a promising target for treating diabetic-associated vascular injury.


Asunto(s)
Diabetes Mellitus , Hiperglucemia , MicroARNs , Humanos , Regulación hacia Abajo , MicroARNs/genética , Angiogénesis , Células Endoteliales de la Vena Umbilical Humana , Diabetes Mellitus/metabolismo , Hiperglucemia/genética , Hiperglucemia/metabolismo , Glucosa/toxicidad , Proliferación Celular , Receptor Tipo 5 de Factor de Crecimiento de Fibroblastos/metabolismo
18.
Stem Cell Rev Rep ; 19(5): 1554-1575, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37060532

RESUMEN

Mesenchymal stem cells (MSCs) is promising in promoting wound healing mainly due to their paracrine function. Nonetheless, the transplanted MSCs presented poor survival with cell dysfunction and paracrine problem in diabetic environment, thus limiting their therapeutic efficacy and clinical application. JAM-A, an adhesion molecule, has been reported to play multi-functional roles in diverse cells. We therefore investigated the potential effect of JAM-A on MSCs under diabetic environment and explored the underlying mechanism. Indeed, high-glucose condition inhibited MSCs viability and JAM-A expression. However, JAM-A abnormality was rescued by lentivirus transfection and JAM-A overexpression promoted MSCs proliferation, migration and adhesion under hyperglycemia. Moreover, JAM-A overexpression attenuated high-glucose-induced ROS production and MSCs apoptosis. The bio-effects of JAM-A on MSCs under hyperglycemia were confirmed by RNA-seq with enrichment analyses. Moreover, Luminex chip results showed JAM-A overexpression dramatically upregulated PDGF-BB and VEGF in the supernatant of MSCs, which was verified by RT-qPCR and western blotting. The supernatant was further found to facilitate HUVECs proliferation, migration and angiogenesis under hyperglycemia. In vivo experiments revealed JAM-A overexpression significantly enhanced MSCs survival, promoted wound angiogenesis, and thus accelerated diabetic wound closure, partially by enhancing PDGF-BB and VEGF expression. This study firstly demonstrated that JAM-A expression of MSCs was inhibited upon high-glucose stimulation. JAM-A overexpression alleviated high-glucose-induced MSCs dysfunction, enhanced their anti-oxidative capability, protected MSCs from hyperglycemia-induced apoptosis and improved their survival, thus strengthening MSCs paracrine function to promote angiogenesis and significantly accelerating diabetic wound healing, which offers a promising strategy to maximize MSCs-based therapy in diabetic wound.


Asunto(s)
Diabetes Mellitus , Hiperglucemia , Células Madre Mesenquimatosas , Neovascularización Fisiológica , Cicatrización de Heridas , Heridas y Lesiones , Humanos , Becaplermina/genética , Becaplermina/metabolismo , Supervivencia Celular/genética , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Glucosa/farmacología , Hiperglucemia/genética , Hiperglucemia/metabolismo , Células Madre Mesenquimatosas/metabolismo , Neovascularización Fisiológica/genética , Comunicación Paracrina/genética , Cordón Umbilical/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas/genética , Heridas y Lesiones/genética , Heridas y Lesiones/metabolismo
19.
J Transl Med ; 21(1): 156, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36855062

RESUMEN

BACKGROUND: Although the relationship between type 2 diabetes (T2D) and the increased risk of colorectal carcinogenesis is widely defined in clinical studies, the therapeutic methods and molecular mechanism of T2D-induced colon cancer and how does hyperglycemia affect the progression is still unknown. Here, we studied the function of lactoferrin (LF) in suppressing the progression of colon cancer in T2D mice, and uncovered the related molecular mechanisms in DNA 5mC and RNA m6A levels. METHODS: We examined the effects of LF (50% iron saturation) on the migration and invasion of colon tumor cells under high concentration of glucose. Then, transcriptomics and DNA methylation profilings of colon tumor cells was co-analyzed to screen out the special gene (NT5DC3), and the expression level of NT5DC3 in 75 clinical blood samples was detected by q-PCR and western blot, to investigate whether NT5DC3 was a biomarker to distinguish T2D patients and T2D-induced colon cancer patients from healthy volunteers. Futhermore, in T2D mouse with xenografted colon tumor models, the inhibitory effects of LF and NT5DC3 protein on colon tumors were investigated. In addition, epigenetic alterations were measured to examine the 5mC/m6A modification sites of NT5DC3 regulated by LF. Utilizing siRNA fragments of eight m6A-related genes, the special gene (WTAP) regulating m6A of NT5DC was proved, and the effect of LF on WTAP/NT5DC3/HKDC1 axis was finally evaluated. RESULTS: A special gene NT5DC3 was screened out through co-analysis of transcriptomics and DNA methylation profiling, and HKDC1 might be a downstream sensor of NT5DC3. Mechanistically, LF-dependent cellular DNA 5mC and RNA m6A profiling remodeling transcriptionally regulate NT5DC3 expression. WTAP plays a key role in regulating NT5DC3 m6A modification and subsequently controls NT5DC3 downstream target HKDC1 expression. Moreover, co-treatment of lactoferrin and NT5DC3 protein restrains the growth of colon tumors by altering the aberrant epigenetic markers. Strikingly, clinical blood samples analysis demonstrates NT5DC3 protein expression is required to direct the distinction of T2D or T2D-induced colon cancer with healthy humans. CONCLUSIONS: Together, this study reveals that lactoferrin acts as a major factor to repress the progression of colon cancer under hyperglycemia, thus, significantly expanding the landscape of natural dietary mediated tumor suppression.


Asunto(s)
Neoplasias del Colon , Diabetes Mellitus Tipo 2 , Hiperglucemia , Humanos , Animales , Ratones , Lactoferrina/genética , Neoplasias del Colon/genética , Hiperglucemia/complicaciones , Hiperglucemia/genética , Modelos Animales de Enfermedad , Factores de Empalme de ARN , Proteínas de Ciclo Celular , Hexoquinasa
20.
Int J Mol Sci ; 24(6)2023 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-36982499

RESUMEN

Hyperglycemia plays a key role in the development of microvascular complications, endothelial dysfunction (ED), and inflammation. It has been demonstrated that cathepsin S (CTSS) is activated in hyperglycemia and is involved in inducing the release of inflammatory cytokines. We hypothesized that blocking CTSS might alleviate the inflammatory responses and reduce the microvascular complications and angiogenesis in hyperglycemic conditions. In this study, we treated human umbilical vein endothelial cells (HUVECs) with high glucose (HG; 30 mM) to induce hyperglycemia and measured the expression of inflammatory cytokines. When treated with glucose, hyperosmolarity could be linked to cathepsin S expression; however, many have mentioned the high expression of CTSS. Thus, we made an effort to concentrate on the immunomodulatory role of the CTSS knockdown in high glucose conditions. We validated that the HG treatment upregulated the expression of inflammatory cytokines and CTSS in HUVEC. Further, siRNA treatment significantly downregulated CTSS expression along with inflammatory marker levels by inhibiting the nuclear factor-kappa B (NF-κB) mediated signaling pathway. In addition, CTSS silencing led to the decreased expression of vascular endothelial markers and downregulated angiogenic activity in HUVECs, which was confirmed by a tube formation experiment. Concurrently, siRNA treatment reduced the activation of complement proteins C3a and C5a in HUVECs under hyperglycemic conditions. These findings show that CTSS silencing significantly reduces hyperglycemia-induced vascular inflammation. Hence, CTSS may be a novel target for preventing diabetes-induced microvascular complications.


Asunto(s)
Complicaciones de la Diabetes , Hiperglucemia , Humanos , FN-kappa B/metabolismo , Transducción de Señal , Hiperglucemia/complicaciones , Hiperglucemia/genética , Hiperglucemia/metabolismo , Glucosa/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Inflamación/genética , Inflamación/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Complicaciones de la Diabetes/metabolismo , Citocinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA