Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.663
1.
Cardiovasc Diabetol ; 23(1): 150, 2024 May 03.
Article En | MEDLINE | ID: mdl-38702777

BACKGROUND: Vasculopathy is the most common complication of diabetes. Endothelial cells located in the innermost layer of blood vessels are constantly affected by blood flow or vascular components; thus, their mechanosensitivity plays an important role in mediating vascular regulation. Endothelial damage, one of the main causes of hyperglycemic vascular complications, has been extensively studied. However, the role of mechanosensitive signaling in hyperglycemic endothelial damage remains unclear. METHODS: Vascular endothelial-specific Piezo1 knockout mice were generated to investigate the effects of Piezo1 on Streptozotocin-induced hyperglycemia and vascular endothelial injury. In vitro activation or knockdown of Piezo1 was performed to evaluate the effects on the proliferation, migration, and tubular function of human umbilical vein endothelial cells in high glucose. Reactive oxygen species production, mitochondrial membrane potential alternations, and oxidative stress-related products were used to assess the extent of oxidative stress damage caused by Piezo1 activation. RESULTS: Our study found that in VECreERT2;Piezo1flox/flox mice with Piezo1 conditional knockout in vascular endothelial cells, Piezo1 deficiency alleviated streptozotocin-induced hyperglycemia with reduced apoptosis and abscission of thoracic aortic endothelial cells, and decreased the inflammatory response of aortic tissue caused by high glucose. Moreover, the knockout of Piezo1 showed a thinner thoracic aortic wall, reduced tunica media damage, and increased endothelial nitric oxide synthase expression in transgenic mice, indicating the relief of endothelial damage caused by hyperglycemia. We also showed that Piezo1 activation aggravated oxidative stress injury and resulted in severe dysfunction through the Ca2+-induced CaMKII-Nrf2 axis in human umbilical vein endothelial cells. In Piezo1 conditional knockout mice, Piezo1 deficiency partially restored superoxide dismutase activity and reduced malondialdehyde content in the thoracic aorta. Mechanistically, Piezo1 deficiency decreased CaMKII phosphorylation and restored the expression of Nrf2 and its downstream molecules HO-1 and NQO1. CONCLUSION: In summary, our study revealed that Piezo1 is involved in high glucose-induced oxidative stress injury and aggravated endothelial dysfunction, which have great significance for alleviating endothelial damage caused by hyperglycemia.


Blood Glucose , Diabetes Mellitus, Experimental , Human Umbilical Vein Endothelial Cells , Ion Channels , Mice, Knockout , Nitric Oxide Synthase Type III , Oxidative Stress , Animals , Humans , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Diabetes Mellitus, Experimental/metabolism , Ion Channels/metabolism , Ion Channels/genetics , Blood Glucose/metabolism , Nitric Oxide Synthase Type III/metabolism , Mechanotransduction, Cellular , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/deficiency , Cells, Cultured , Cell Proliferation , Apoptosis , Male , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/physiopathology , Diabetic Angiopathies/pathology , Diabetic Angiopathies/genetics , Diabetic Angiopathies/etiology , Cell Movement , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aorta, Thoracic/physiopathology , Mice , Streptozocin , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Endothelium, Vascular/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics
2.
Atherosclerosis ; 392: 117527, 2024 May.
Article En | MEDLINE | ID: mdl-38583286

BACKGROUND AND AIMS: Diabetic atherosclerotic vascular disease is characterized by extensive vascular calcification. However, an elevated blood glucose level alone does not explain this pathogenesis. We investigated the metabolic markers underlying diabetic atherosclerosis and whether extracellular Hsp90α (eHsp90α) triggers vascular endothelial calcification in this particular metabolic environment. METHODS: A parallel human/animal model metabolomics approach was used. We analyzed 40 serum samples collected from 24 patients with atherosclerosis and from the STZ-induced ApoE-/- mouse model. A multivariate statistical analysis of the data was performed, and mouse aortic tissue was collected for the assessment of plaque formation. In vitro, the effects of eHsp90α on endothelial cell calcification were assessed by serum analysis, Western blotting and immunoelectron microscopy. RESULTS: Diabetic ApoE-/- mice showed more severe plaque lesions and calcification damage. Stearamide, oleamide, l-thyroxine, l-homocitrulline and l-citrulline are biomarkers of diabetic ASVD; l-thyroxine was downregulated in both groups, and the thyroid sensitivity index was correlated with serum Hsp90α concentration. In vitro studies showed that eHsp90α increased Runx2 expression in endothelial cells through the LRP1 receptor. l-thyroxine reduced the increase in Runx2 levels caused by eHsp90α and affected the distribution and expression of LRP1 through hydrogen bonding with glutamine at position 1054 in the extracellular segment of LRP1. CONCLUSIONS: This study provides a mechanistic link between characteristic serum metabolites and diabetic atherosclerosis and thus offers new insight into the role of extracellular Hsp90α in promoting vascular calcification.


Diabetes Mellitus, Experimental , HSP90 Heat-Shock Proteins , Mice, Knockout, ApoE , Plaque, Atherosclerotic , Thyroxine , Vascular Calcification , Humans , Animals , HSP90 Heat-Shock Proteins/metabolism , Vascular Calcification/metabolism , Vascular Calcification/pathology , Male , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/complications , Thyroxine/blood , Female , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Middle Aged , Core Binding Factor Alpha 1 Subunit/metabolism , Mice , Atherosclerosis/metabolism , Atherosclerosis/pathology , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Diabetic Angiopathies/etiology , Metabolomics/methods , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Metabolome/drug effects , Aged , Mice, Inbred C57BL , Aortic Diseases/metabolism , Aortic Diseases/pathology , Aortic Diseases/blood , Biomarkers/blood , Human Umbilical Vein Endothelial Cells/metabolism
3.
Front Endocrinol (Lausanne) ; 15: 1359255, 2024.
Article En | MEDLINE | ID: mdl-38645427

Diabetic vascular complications are prevalent and severe among diabetic patients, profoundly affecting both their quality of life and long-term prospects. These complications can be classified into macrovascular and microvascular complications. Under the impact of risk factors such as elevated blood glucose, blood pressure, and cholesterol lipids, the vascular endothelium undergoes endothelial dysfunction, characterized by increased inflammation and oxidative stress, decreased NO biosynthesis, endothelial-mesenchymal transition, senescence, and even cell death. These processes will ultimately lead to macrovascular and microvascular diseases, with macrovascular diseases mainly characterized by atherosclerosis (AS) and microvascular diseases mainly characterized by thickening of the basement membrane. It further indicates a primary contributor to the elevated morbidity and mortality observed in individuals with diabetes. In this review, we will delve into the intricate mechanisms that drive endothelial dysfunction during diabetes progression and its associated vascular complications. Furthermore, we will outline various pharmacotherapies targeting diabetic endothelial dysfunction in the hope of accelerating effective therapeutic drug discovery for early control of diabetes and its vascular complications.


Diabetic Angiopathies , Endothelium, Vascular , Humans , Endothelium, Vascular/physiopathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/etiology , Diabetic Angiopathies/physiopathology , Diabetic Angiopathies/pathology , Animals , Oxidative Stress/physiology
4.
Front Endocrinol (Lausanne) ; 15: 1367376, 2024.
Article En | MEDLINE | ID: mdl-38660516

Background: The systemic immuno-inflammation index (SII), neutrophil-to-lymphocyte ratio (NLR), and platelet-to-lymphocyte ratio (PLR) are widely used and have been shown to be predictive indicators of various diseases. Diabetic nephropathy (DN), retinopathy (DR), and peripheral neuropathy (DPN) are the most prominent and common microvascular complications, which have seriously negative impacts on patients, families, and society. Exploring the associations with these three indicators and diabetic microvascular complications are the main purpose. Methods: There were 1058 individuals with type 2 diabetes mellitus (T2DM) in this retrospective cross-sectional study. SII, NLR, and PLR were calculated. The diseases were diagnosed by endocrinologists. Logistic regression and subgroup analysis were applied to evaluate the association between SII, NLP, and PLR and diabetic microvascular complications. Results: SII, NLR, and PLR were significantly associated with the risk of DN [odds ratios (ORs): 1.52, 1.71, and 1.60, respectively] and DR [ORs: 1.57, 1.79, and 1.55, respectively] by multivariate logistic regression. When NLR ≥2.66, the OR was significantly higher for the risk of DPN (OR: 1.985, 95% confidence interval: 1.29-3.05). Subgroup analysis showed no significant positive associations across different demographics and comorbidities, including sex, age, hypertension, HbA1c (glycated hemoglobin), and dyslipidemia. Conclusion: This study found a positive relationship between NLR and DN, DR, and DPN. In contrast, SII and PLR were found to be only associated with DN and DR. Therefore, for the diagnosis of diabetic microvascular complications, SII, NLR and PLR are highly valuable.


Blood Platelets , Diabetes Mellitus, Type 2 , Diabetic Angiopathies , Lymphocytes , Neutrophils , Humans , Male , Female , Middle Aged , Neutrophils/pathology , Retrospective Studies , Cross-Sectional Studies , Lymphocytes/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/blood , Diabetic Angiopathies/blood , Diabetic Angiopathies/diagnosis , Diabetic Angiopathies/immunology , Diabetic Angiopathies/pathology , Blood Platelets/pathology , Aged , Inflammation/blood , Inflammation/pathology , Diabetic Neuropathies/blood , Diabetic Neuropathies/pathology , Diabetic Neuropathies/etiology , Diabetic Neuropathies/diagnosis , Diabetic Retinopathy/blood , Diabetic Retinopathy/diagnosis , Diabetic Retinopathy/immunology , Diabetic Nephropathies/blood , Diabetic Nephropathies/pathology , Diabetic Nephropathies/diagnosis , Lymphocyte Count , Platelet Count , Adult
5.
Cardiovasc Diabetol ; 23(1): 140, 2024 Apr 25.
Article En | MEDLINE | ID: mdl-38664681

BACKGROUND: Diabetic vascular remodeling is the most important pathological basis of diabetic cardiovascular complications. The accumulation of advanced glycation end products (AGEs) caused by elevated blood glucose promotes the proliferation and migration of vascular smooth muscle cells (VSMCs), leading to arterial wall thickening and ultimately vascular remodeling. Therefore, the excessive proliferation and migration of VSMCs is considered as an important therapeutic target for vascular remodeling in diabetes mellitus. However, due to the lack of breakthrough in experiments, there is currently no effective treatment for the excessive proliferation and migration of VSMCs in diabetic patients. Bcl-2-associated athanogene 3 (BAG3) protein is a multifunctional protein highly expressed in skeletal muscle and myocardium. Previous research has confirmed that BAG3 can not only regulate cell survival and apoptosis, but also affect cell proliferation and migration. Since the excessive proliferation and migration of VSMCs is an important pathogenesis of vascular remodeling in diabetes, the role of BAG3 in the excessive proliferation and migration of VSMCs and its molecular mechanism deserve further investigation. METHODS: In this study, BAG3 gene was manipulated in smooth muscle to acquire SM22αCre; BAG3FL/FL mice and streptozotocin (STZ) was used to simulate diabetes. Expression of proteins and aortic thickness of mice were detected by immunofluorescence, ultrasound and hematoxylin-eosin (HE) staining. Using human aorta smooth muscle cell line (HASMC), cell viability was measured by CCK-8 and proliferation was measured by colony formation experiment. Migration was detected by transwell, scratch experiments and Phalloidin staining. Western Blot was used to detect protein expression and Co-Immunoprecipitation (Co-IP) was used to detect protein interaction. RESULTS: In diabetic vascular remodeling, AGEs could promote the interaction between BAG3 and signal transducer and activator of transcription 3 (STAT3), leading to the enhanced interaction between STAT3 and Janus kinase 2 (JAK2) and reduced interaction between STAT3 and extracellular signal-regulated kinase 1/2 (ERK1/2), resulting in accumulated p-STAT3(705) and reduced p-STAT3(727). Subsequently, the expression of matrix metallopeptidase 2 (MMP2) is upregulated, thus promoting the migration of VSMCs. CONCLUSIONS: BAG3 upregulates the expression of MMP2 by increasing p-STAT3(705) and decreasing p-STAT3(727) levels, thereby promoting vascular remodeling in diabetes. This provides a new orientation for the prevention and treatment of diabetic vascular remodeling.


Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins , Cell Movement , Cell Proliferation , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , STAT3 Transcription Factor , Signal Transduction , Vascular Remodeling , STAT3 Transcription Factor/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Animals , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Phosphorylation , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Diabetic Angiopathies/physiopathology , Diabetic Angiopathies/etiology , Diabetic Angiopathies/genetics , Male , Cells, Cultured , Mice, Knockout , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Humans , Mice, Inbred C57BL , Glycation End Products, Advanced/metabolism
6.
Front Endocrinol (Lausanne) ; 14: 1112363, 2023.
Article En | MEDLINE | ID: mdl-36824356

Diabetes mellitus (DM) is a metabolic disease characterized by chronic hyperglycaemia, with absolute insulin deficiency or insulin resistance as the main cause, and causes damage to various target organs including the heart, kidney and neurovascular. In terms of the pathological and physiological mechanisms of DM, oxidative stress is one of the main mechanisms leading to DM and is an important link between DM and its complications. Oxidative stress is a pathological phenomenon resulting from an imbalance between the production of free radicals and the scavenging of antioxidant systems. The main site of reactive oxygen species (ROS) production is the mitochondria, which are also the main organelles damaged. In a chronic high glucose environment, impaired electron transport chain within the mitochondria leads to the production of ROS, prompts increased proton leakage and altered mitochondrial membrane potential (MMP), which in turn releases cytochrome c (cyt-c), leading to apoptosis. This subsequently leads to a vicious cycle of impaired clearance by the body's antioxidant system, impaired transcription and protein synthesis of mitochondrial DNA (mtDNA), which is responsible for encoding mitochondrial proteins, and impaired DNA repair systems, contributing to mitochondrial dysfunction. This paper reviews the dysfunction of mitochondria in the environment of high glucose induced oxidative stress in the DM model, and looks forward to providing a new treatment plan for oxidative stress based on mitochondrial dysfunction.


Diabetes Mellitus , Diabetic Angiopathies , Humans , Reactive Oxygen Species/metabolism , Antioxidants/metabolism , Oxidative Stress/physiology , Mitochondria/metabolism , DNA, Mitochondrial/genetics , Diabetes Mellitus/metabolism , Glucose/metabolism , Diabetic Angiopathies/pathology
7.
Sci Rep ; 12(1): 264, 2022 01 07.
Article En | MEDLINE | ID: mdl-34997031

Diabetes can cause microvessel impairment. However, these conjunctival pathological changes are not easily recognized, limiting their potential as independent diagnostic indicators. Therefore, we designed a deep learning model to explore the relationship between conjunctival features and diabetes, and to advance automated identification of diabetes through conjunctival images. Images were collected from patients with type 2 diabetes and healthy volunteers. A hierarchical multi-tasking network model (HMT-Net) was developed using conjunctival images, and the model was systematically evaluated and compared with other algorithms. The sensitivity, specificity, and accuracy of the HMT-Net model to identify diabetes were 78.70%, 69.08%, and 75.15%, respectively. The performance of the HMT-Net model was significantly better than that of ophthalmologists. The model allowed sensitive and rapid discrimination by assessment of conjunctival images and can be potentially useful for identifying diabetes.


Algorithms , Conjunctiva/blood supply , Diabetes Mellitus, Type 2/pathology , Diabetic Angiopathies/pathology , Diagnosis, Computer-Assisted , Diagnostic Techniques, Ophthalmological , Image Interpretation, Computer-Assisted , Microvessels/pathology , Case-Control Studies , Diabetes Mellitus, Type 2/complications , Diabetic Angiopathies/etiology , Humans , Predictive Value of Tests , Prospective Studies , Reproducibility of Results
8.
J Cardiovasc Pharmacol ; 79(1): e1-e10, 2022 01 01.
Article En | MEDLINE | ID: mdl-34654782

ABSTRACT: In the context of diabetes mellitus, various pathological changes cause tissue ischemia and hypoxia, which can lead to the compensatory formation of neovascularization. However, disorders of the internal environment and dysfunctions of various cells contribute to the dysfunction of neovascularization. Although the problems of tissue ischemia and hypoxia have been partially solved, neovascularization also causes many negative effects. In the process of small blood vessel renewal, pericytes are extremely important for maintaining the normal growth and maturation of neovascularization. Previously, our understanding of pericytes was very limited, and the function of pericytes was not yet clear. Recently, multiple new functions of pericytes have been identified, affecting various processes in angiogenesis and relating to various diseases. Therefore, the importance of pericytes has gradually become apparent. This article presents the latest research progress on the role of pericytes in diabetic angiogenesis, characterizes pericytes, summarizes various potential therapeutic targets, and highlights research directions for the future treatment of various diabetes-related diseases.


Diabetic Angiopathies/pathology , Neovascularization, Pathologic , Pericytes/pathology , Angiogenic Proteins/metabolism , Animals , Cell Hypoxia , Diabetic Angiopathies/metabolism , Humans , Pericytes/metabolism , Phenotype , Signal Transduction
9.
Microvasc Res ; 139: 104272, 2022 01.
Article En | MEDLINE | ID: mdl-34699845

Endothelial injury plays a vital role in vascular lesions from diabetes mellitus (DM). Therapeutic targets against endothelial damage may provide critical venues for the treatment of diabetic vascular diseases. Peroxisome proliferator-activated receptor ß (PPARß) is a crucial regulator in DM and its complications. However, the molecular signal mediating the roles of PPARß in DM-induced endothelial dysfunction is not fully understood. The impaired endothelium-dependent relaxation and destruction of the endothelium structures appeared in high glucose incubated rat aortic rings. A high glucose level significantly decreased the expression of PPARß and endothelial nitric oxide synthase (eNOS) at the mRNA and protein levels, and reduced the concentration of nitric oxide (NO), which occurred in parallel with an increase in the expression of inducible nitric oxide synthase (iNOS) and 3-nitrotyrosine. The effect of high glucose was inhibited by GW0742, a PPARß agonist. Both GSK0660 (PPARß antagonist) and NG-nitro-l-arginine-methyl ester (NOS inhibitor) could reverse the protective effects of GW0742. These results suggest that the activation of nitrative stress may, at least in part, mediate the down-regulation of PPARß in high glucose-impaired endothelial function in rat aorta. PPARß-nitrative stress may hold potential in treating vascular complications from DM.


Aorta, Thoracic/drug effects , Diabetic Angiopathies/metabolism , Endothelial Cells/drug effects , Glucose/toxicity , Hyperglycemia/metabolism , Nitrosative Stress/drug effects , PPAR-beta/metabolism , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aorta, Thoracic/physiopathology , Diabetic Angiopathies/genetics , Diabetic Angiopathies/pathology , Diabetic Angiopathies/physiopathology , Down-Regulation , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Hyperglycemia/genetics , Hyperglycemia/pathology , Hyperglycemia/physiopathology , Male , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , PPAR-beta/genetics , Rats, Sprague-Dawley , Signal Transduction , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Vasodilation/drug effects
10.
Front Endocrinol (Lausanne) ; 12: 722861, 2021.
Article En | MEDLINE | ID: mdl-34759889

Objective: We aimed to investigate the alterations of brain functional connectivity (FC) in type 2 diabetes mellitus (T2DM) patients without clinical evidence of cognitive impairment and microvascular complications (woCIMC-T2DM) using resting-state functional MRI (rs-fMRI) and to determine whether its value was correlated with clinical indicators. Methods: A total of 27 T2DM and 26 healthy controls (HCs) were prospectively examined. Cognitive impairment was excluded using the Mini-Mental State Examination (MMSE) and the Montreal Cognitive Assessment (MoCA) scales, and microvascular complications were excluded by fundus photography, microalbuminuria, and other indicators. The correlation maps, derived from rs-fMRI with posterior cingulate cortex (PCC) as the seed, were compared between T2DM patients and HCs. Pearson's correlation analysis was performed to determine the relationship between the FC of PCC and the clinical indicators. Results: Compared with HC, woCIMC-T2DM patients showed significantly decreased FCs with PCC (PCC-FCs) in the anterior cingulate cortex (ACC), right superior frontal gyrus, right medial frontal gyrus, and right angular gyrus. Meanwhile, increased PCC-FCs was observed in the right superior temporal gyrus and calcarine fissure (CAL). The FC of PCC-ACC was negatively correlated with glycosylated hemoglobin (HbA1c) and diabetes duration, and the FC of PCC-CAL was significantly positively correlated with HbA1c and diabetes duration. Conclusion: The FC, especially of the PCC with cognitive and visual brain regions, was altered before clinically measurable cognitive impairment and microvascular complications occurred in T2DM patients. In addition, the FC of the PCC with cognitive and visual brain regions was correlated with HbA1c and diabetes duration. This indicates that clinicians should pay attention not only to blood glucose control but also to brain function changes before the occurrence of adverse complications, which is of great significance for the prevention of cognitive dysfunction and visual impairment.


Diabetes Mellitus, Type 2/psychology , Gyrus Cinguli/pathology , Nerve Net/pathology , Adult , Aged , Case-Control Studies , China , Cognitive Dysfunction/complications , Cognitive Dysfunction/diagnosis , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/pathology , Diabetic Angiopathies/diagnosis , Diabetic Angiopathies/pathology , Diabetic Angiopathies/psychology , Female , Gyrus Cinguli/diagnostic imaging , Humans , Magnetic Resonance Imaging , Male , Microvessels/diagnostic imaging , Microvessels/pathology , Middle Aged , Nerve Net/diagnostic imaging , Neuropsychological Tests
11.
Eur J Pharmacol ; 913: 174630, 2021 Dec 15.
Article En | MEDLINE | ID: mdl-34774495

Endothelial dysfunction contributes to the development of diabetic complications and the production of circulating microparticles (MPs). Our previous study showed that diabetic mice-derived MPs (DM MPs) had increased levels of extracellular regulated protein kinase 1/2 (ERK1/2) and impaired endothelial-dependent relaxation in aortas when compared with control mice-derived MPs. This study was designed to investigate whether PD98059, an ERK1/2 inhibitor, affects the function of aortas and DM MPs. MPs were obtained from streptozotocin-induced DM, DM after PD98059 treatment, and ICR mice as control. The mice and MPs were then analyzed on the basis of their vascular function and enzyme expressions. Compared with the controls, platelet-derived MPs and ERK1/2 levels in the MPs were significantly elevated in the DM but showed little change in PD98059-treated DM. PD98059 mainly decreased ERK1/2 phosphorylation in the MPs. In the aortas of DM and DM MPs the endothelium-dependent vascular function was impaired, and there was a significantly greater improvement in the vascular function in the PD98059-treated DM aortas and the aortas treated with PD98059-treated DM MPs than in DM aortas and the aortas treated with DM MPs. Furthermore, DM MPs increased ERK1/2 and intracellular adhesion molecule-1 (ICAM-1) expressions in the aortas, but PD98059-treated DM MPs did not show these effects. For the first time, these results indicate that PD98059 treatment improves endothelial dysfunction in DM, and adhesion properties of DM MPs can be partly blocked by PD98059 via ERK and ICAM-1. These effects may explain some of the vascular complications in diabetes.


Cell-Derived Microparticles/pathology , Diabetes Mellitus, Experimental/complications , Diabetic Angiopathies/drug therapy , Endothelium, Vascular/drug effects , Flavonoids/pharmacology , Animals , Cell-Derived Microparticles/drug effects , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/drug therapy , Diabetic Angiopathies/etiology , Diabetic Angiopathies/pathology , Endothelium, Vascular/pathology , Flavonoids/therapeutic use , Humans , Male , Mice , Mice, Inbred ICR
12.
J Diabetes Res ; 2021: 4461311, 2021.
Article En | MEDLINE | ID: mdl-34631895

Vascular calcification is the transformation of arterial wall mesenchymal cells, particularly smooth muscle cells (SMCs), into osteoblast phenotypes by various pathological factors. Additionally, vascular transformation mediates the abnormal deposition of calcium salts in the vascular wall, such as intimal and media calcification. Various pathological types have been described, such as calcification and valve calcification. The incidence of vascular calcification in patients with diabetes is much higher than that in nondiabetic patients, representing a critical cause of cardiovascular events in patients with diabetes. Because basic research on the clinical transformation of vascular calcification has yet to be conducted, this study systematically expounds on the risk factors for vascular calcification, vascular bed differences, sex differences, ethnic differences, diagnosis, severity assessments, and treatments to facilitate the identification of a new entry point for basic research and subsequent clinical transformation regarding vascular calcification and corresponding clinical evaluation strategies.


Diabetes Mellitus/epidemiology , Diabetic Angiopathies/epidemiology , Epidemiologic Research Design , Vascular Calcification/epidemiology , Animals , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Humans , Incidence , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Phenotype , Prevalence , Prognosis , Risk Assessment , Risk Factors , Vascular Calcification/metabolism , Vascular Calcification/pathology
13.
J Diabetes Complications ; 35(12): 108055, 2021 12.
Article En | MEDLINE | ID: mdl-34620556

BACKGROUND: Prospective studies describe a linkage between increased sodium intake and higher incidence of cardiovascular organ damage and end points. We analyzed whether tissue sodium content in the skin and muscles correlate with vascular hypertrophic remodeling, a risk factor for cardiovascular disease. METHODS: In patients with type 2 diabetes we assessed tissue sodium content and vascular structural parameters of the retinal arterioles. The structural parameters of retinal arterioles assessed by Scanning Laser Doppler Flowmetry were vessel (VD) and lumen diameter (LD), wall thickness (WT), wall-to-lumen ratio (WLR) and wall cross sectional area (WCSA). Tissue sodium content was measured with a 3.0 T clinical 23Sodium-Magnetic Resonance Imaging (23Na-MRI) system. RESULTS: In patients with type 2 diabetes (N = 52) we observed a significant correlation between muscle sodium content and VD (p = 0.005), WT (p = 0.003), WCSA (p = 0.002) and WLR (p = 0.013). With respect to skin sodium content a significant correlation has been found with VD (p = 0.042), WT (p = 0.023) and WCSA (p = 0.019). Further analysis demonstrated that tissue sodium content of skin and muscle is a significant determinant of hypertrophic vascular remodeling independent of age, gender, diuretic use and 24-hour ambulatory BP. CONCLUSION: With the 23Na-MRI technology we could demonstrate that high tissue sodium content is independently linked to hypertrophic vascular remodeling in type 2 diabetes. TRIAL REGISTRATION: Trial registration number: NCT02383238 Date of registration: March 9, 2015.


Arterioles/diagnostic imaging , Diabetes Mellitus, Type 2/physiopathology , Diabetic Angiopathies/physiopathology , Retina , Sodium/analysis , Vascular Remodeling/physiology , Aged , Arterioles/pathology , Arterioles/physiopathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnostic imaging , Diabetes Mellitus, Type 2/pathology , Diabetic Angiopathies/diagnostic imaging , Diabetic Angiopathies/etiology , Diabetic Angiopathies/pathology , Eye/blood supply , Female , Humans , Hypertrophy/diagnostic imaging , Hypertrophy/physiopathology , Laser-Doppler Flowmetry , Magnetic Resonance Imaging , Male , Middle Aged , Muscles/chemistry , Prospective Studies , Retina/diagnostic imaging , Retina/pathology , Retina/physiopathology , Skin/chemistry
14.
Cells ; 10(9)2021 09 04.
Article En | MEDLINE | ID: mdl-34571964

Oxidative stress within the vascular endothelium, due to excess generation of reactive oxygen species (ROS), is thought to be fundamental to the initiation and progression of the cardiovascular complications of type 2 diabetes mellitus. The term ROS encompasses a variety of chemical species including superoxide anion (O2•-), hydroxyl radical (OH-) and hydrogen peroxide (H2O2). While constitutive generation of low concentrations of ROS are indispensable for normal cellular function, excess O2•- can result in irreversible tissue damage. Excess ROS generation is catalysed by xanthine oxidase, uncoupled nitric oxide synthases, the mitochondrial electron transport chain and the nicotinamide adenine dinucleotide phosphate (NADPH) oxidases. Amongst enzymatic sources of O2•- the Nox2 isoform of NADPH oxidase is thought to be critical to the oxidative stress found in type 2 diabetes mellitus. In contrast, the transcriptionally regulated Nox4 isoform, which generates H2O2, may fulfil a protective role and contribute to normal glucose homeostasis. This review describes the key roles of Nox2 and Nox4, as well as Nox1 and Nox5, in glucose homeostasis, endothelial function and oxidative stress, with a key focus on how they are regulated in health, and dysregulated in type 2 diabetes mellitus.


Blood Glucose/metabolism , Diabetes Mellitus, Type 2/enzymology , Diabetic Angiopathies/enzymology , Endothelial Cells/enzymology , NADPH Oxidases/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Animals , Biomarkers/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/pathology , Diabetic Angiopathies/blood , Diabetic Angiopathies/pathology , Endothelial Cells/pathology , Homeostasis , Humans , Isoenzymes , Signal Transduction
15.
Front Endocrinol (Lausanne) ; 12: 731273, 2021.
Article En | MEDLINE | ID: mdl-34489872

Vascular aging is characterized by alterations in the constitutive properties and biological functions of the blood vessel wall. Endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) are indispensability elements in the inner layer and the medial layer of the blood vessel wall, respectively. Dipeptidyl peptidase-4 (DPP4) inhibitors, as a hypoglycemic agent, play a protective role in reversing vascular aging regardless of their effects in meliorating glycemic control in humans and animal models of type 2 diabetes mellitus (T2DM) through complex cellular mechanisms, including improving EC dysfunction, promoting EC proliferation and migration, alleviating EC senescence, obstructing EC apoptosis, suppressing the proliferation and migration of VSMCs, increasing circulating endothelial progenitor cell (EPC) levels, and preventing the infiltration of mononuclear macrophages. All of these showed that DPP4 inhibitors may exert a positive effect against vascular aging, thereby preventing vascular aging-related diseases. In the current review, we will summarize the cellular mechanism of DPP4 inhibitors regulating vascular aging; moreover, we also intend to compile the roles and the promising therapeutic application of DPP4 inhibitors in vascular aging-related diseases.


Aging/pathology , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Vascular Diseases/drug therapy , Vascular Diseases/etiology , Aging/drug effects , Animals , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Diabetic Angiopathies/etiology , Diabetic Angiopathies/pathology , Diabetic Angiopathies/physiopathology , Diabetic Angiopathies/prevention & control , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelial Cells/physiology , Humans , Signal Transduction/drug effects
16.
Int J Mol Sci ; 22(17)2021 Aug 26.
Article En | MEDLINE | ID: mdl-34502144

Impaired wound healing in people with diabetes has multifactorial causes, with insufficient neovascularization being one of the most important. Hypoxia-inducible factor-1 (HIF-1) plays a central role in the hypoxia-induced response by activating angiogenesis factors. As its activity is under precise regulatory control of prolyl-hydroxylase domain 2 (PHD-2), downregulation of PHD-2 by small interfering RNA (siRNA) could stabilize HIF-1α and, therefore, upregulate the expression of pro-angiogenic factors as well. Intracellular delivery of siRNA can be achieved with nanocarriers that must fulfill several requirements, including high stability, low toxicity, and high transfection efficiency. Here, we designed and compared the performance of layer-by-layer self-assembled siRNA-loaded gold nanoparticles with two different outer layers-Chitosan (AuNP@CS) and Poly L-arginine (AuNP@PLA). Although both formulations have exactly the same core, we find that a PLA outer layer improves the endosomal escape of siRNA, and therefore, transfection efficiency, after endocytic uptake in NIH-3T3 cells. Furthermore, we found that endosomal escape of AuNP@PLA could be improved further when cells were additionally treated with desloratadine, thus outperforming commercial reagents such as Lipofectamine® and jetPRIME®. AuNP@PLA in combination with desloratadine was proven to induce PHD-2 silencing in fibroblasts, allowing upregulation of pro-angiogenic pathways. This finding in an in vitro context constitutes a first step towards improving diabetic wound healing with siRNA therapy.


Angiogenesis Inducing Agents/metabolism , Diabetic Angiopathies/metabolism , Gold , Hypoxia/metabolism , Lysosomes , Nanoparticles , RNA, Small Interfering/genetics , Animals , Cell Survival , Chemical Phenomena , Diabetic Angiopathies/etiology , Diabetic Angiopathies/pathology , Drug Compounding , Endosomes/metabolism , Gene Transfer Techniques , Hypoxia/genetics , Loratadine/analogs & derivatives , Loratadine/chemistry , Loratadine/pharmacology , Mice , NIH 3T3 Cells , Nanoparticles/chemistry , RNA, Small Interfering/administration & dosage
17.
Front Endocrinol (Lausanne) ; 12: 720466, 2021.
Article En | MEDLINE | ID: mdl-34456875

Diabetic vascular complications (DVC) including macrovascular and microvascular lesions, have a significant impact on public health, and lead to increased patient mortality. Disordered intercellular cascades play a vital role in diabetic systemic vasculopathy. Exosomes participate in the abnormal signal transduction of local vascular cells and mediate the transmission of metabolic disorder signal molecules in distant organs and cells through the blood circulation. They can store different signaling molecules in the membrane structure and release them into the blood, urine, and tears. In recent years, the carrier value and therapeutic effect of exosomes derived from stem cells have garnered attention. Exosomes are not only a promising biomarker but also a potential target and tool for the treatment of DVC. This review explored changes in the production process of exosomes in the diabetic microenvironment and exosomes' early warning role in DVC from different systems and their pathological processes. On the basis of these findings, we discussed the future direction of exosomes in the treatment of DVC, and the current limitations of exosomes in DVC research.


Diabetic Angiopathies/diagnosis , Diabetic Angiopathies/therapy , Exosomes/physiology , Biomarkers/analysis , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Humans , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends
18.
Diabetes Metab Syndr ; 15(5): 102241, 2021.
Article En | MEDLINE | ID: mdl-34390975

BACKGROUND: Diabetic microvascular complications are a major cause of morbidity and are related to glycaemic control and cardiovascular risk factors. AIMS: We sought to determine the association of microvascular complications in relation to control of glycemia, blood pressure and lipids in T2DM patients attending secondary care in Qatar. METHODS: This is a cross-sectional study undertaken in patients with T2DM attending Qatar's National Diabetes Centres. Patients underwent assessment of glycemia, blood pressure and lipids and prevalence of diabetic peripheral neuropathy (DPN), retinopathy and microalbuminuria. RESULTS: We included 1114 subjects aged 52.1 ± 11.3 years with a duration of diabetes 10.0 ± 7.6 years and had a prevalence of 25.8% for DPN, 34.3% for painful DPN, 36.8% for microalbuminuria and 25.1% for retinopathy. Patients who achieved an HbA1c ≤ 7.0% compared to >7% had a significantly lower prevalence of DPN (P < 0.01), painful DPN (P < 0.01), retinopathy (P < 0.01) and microalbuminuria (P < 0.007). Patients who achieved a systolic BP ≤ 140 mmHg compared to >140 mmHg had a significantly lower prevalence of DPN (P < 0.001), painful DPN (P < 0.001), retinopathy (P < 0.001) and microalbuminuria (P < 0.001). Patients who achieved an LDL ≤2.6 mmol/l compared to >2.6 mmol/l had a significantly higher prevalence of DPN (P < 0.03), but no difference in other outcomes. There was no difference in microvascular complications between those who achieved a HDL-C ≥ 1.02 mmol/l, and among those who achieved triglycerides ≤1.7 mmol/l. CONCLUSIONS: Optimal control of glycemia and blood pressure, but not lipids is associated with a lower prevalence of diabetic microvascular complications.


Blood Pressure , Diabetes Mellitus, Type 2/drug therapy , Diabetic Angiopathies/prevention & control , Diabetic Neuropathies/prevention & control , Diabetic Retinopathy/prevention & control , Glycemic Control/standards , Lipids/analysis , Biomarkers/blood , Blood Glucose/analysis , Cross-Sectional Studies , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetic Angiopathies/epidemiology , Diabetic Angiopathies/pathology , Diabetic Neuropathies/epidemiology , Diabetic Neuropathies/pathology , Diabetic Retinopathy/epidemiology , Diabetic Retinopathy/pathology , Female , Follow-Up Studies , Glycated Hemoglobin/analysis , Humans , Male , Middle Aged , Prevalence , Prognosis , Qatar/epidemiology , Triglycerides/metabolism
19.
Front Endocrinol (Lausanne) ; 12: 664183, 2021.
Article En | MEDLINE | ID: mdl-34367063

Background: Controlling blood glucose, blood pressure, and blood lipid is of great importance for patients with type 2 diabetes, not only for cardiovascular disease, but also for other complications. Previous studies mainly focused on the control rate of outpatients, and the results were suboptimal, but few studies aimed at the inpatients. Method: The present study involved 3,245 hospitalized patients with type 2 diabetes from 2013 to 2017 in the Department of Endocrinology and Metabolism of Tianjin Medical University General Hospital. The percentages of inpatients who attained the goals of the China Diabetes Society and the American Diabetes Association were calculated for major cardiovascular risk factors (HbA1c, blood pressure, and blood lipid). The prevalence of microvascular and macrovascular complications was also assessed. Result: The percentages of patients who met the Chinese Diabetes Society goals-HbA1c <7%, blood pressure <130/80 mmHg, normal lipids, and all three goals-were 26.7, 14.8, 10.4, and 0.2% in 2013 and 30.5, 16.2, 8.0, and 0.9% in 2017, respectively. The percentage of patients who met all three American Diabetes Association goals (HbA1c<7%, blood pressure <140/90 mmHg, low-density lipoprotein cholesterol <2.6 mmol/L) increased from 4.3% in 2013 to 9.0% in 2017. The prevalence of major diabetes complications including coronary heart disease (31.7 vs. 31.9%), stroke (16.7 vs. 14.8%), diabetic kidney disease (37.9 vs. 35.8%), diabetic retinopathy (48.0 vs. 46.5%), neuropathy (63.1 vs. 61.9%), and diabetic foot (0.8 vs. 1.2%) were stable from 2013 to 2017. Conclusion: During 2013 to 2017, control rates of major cardiovascular risk factors including HbA1c, blood pressure, and low-density lipoprotein cholesterol were improved among hospitalized patients in Tianjin, China.


Cardiovascular Diseases/pathology , Diabetes Mellitus, Type 2/complications , Diabetic Angiopathies/pathology , Hospitalization/statistics & numerical data , Hypertension/pathology , Biomarkers/blood , Blood Glucose/analysis , Cardiovascular Diseases/etiology , Diabetic Angiopathies/etiology , Female , Follow-Up Studies , Heart Disease Risk Factors , Humans , Hypertension/etiology , Male , Middle Aged , Prognosis
20.
Mol Pharm ; 18(9): 3302-3325, 2021 09 06.
Article En | MEDLINE | ID: mdl-34297586

Microvascular complications are among the major outcomes of patients with type II diabetes mellitus, which are the consequences of impaired physiological functioning of small blood vessels and angiogenic responses in these patients. Overproduction and accumulation of methylglyoxal (MGO), a highly reactive dicarbonyl byproduct of glycolysis pathway, has been acclaimed as the main inducer of impaired angiogenic responses and microvascular dysfunction in diabetic patients with uncontrolled hyperglycemia. Hence, an effective approach to overcome diabetes-associated microvascular complications is to neutralize the deleterious activity of enhanced the concentration of MGO in the body. Owing to the glycation inhibitory activity of Aloe vera whole extract, and capability of l-carnosine, an endogenous dipeptide, in attenuating MGO's destructive activity, we examined whether application of a combination of l-carnosine and A. vera could be an effective way of synergistically weakening this reactive dicarbonyl's impaired angiogenic effects. Additionally, overcoming the poor cellular uptake and internalization of l-carnosine and A. vera, a nanophytosomal formulation of the physical mixture of two compounds was also established. Although l-carnosine and A. vera at whole studied combination ratios could synergistically enhance viability of human umbilical vein endothelial cells (HUVECs) treated with MGO, the 25:1 w/w ratio was the most effective one among the others (27 ± 0.5% compared to 12 ± 0.3 to 18 ± 0.4%; F (4, 15) = 183.9, P < 0.0001). Developing dual nanophytosomes of l-carnosine/A. vera (25:1) combination ratio, we demonstrated superiority of the nanophytosomal formulation in protecting HUVECs against MGO-induced toxicity following a 24-72 h incubation period (17.3, 15.8, and 12.4% respectively). Moreover, 500 µg/mL concentration of dual l-carnosine/A. vera nanophytosomes exhibited a superior free radical scavenging potency (63 ± 4 RFU vs 83 ± 5 RFU; F (5, 12) = 54.81, P < 0.0001) and nitric oxide synthesizing capacity (26.11 ± 0.19 vs 5.1 ± 0.33; F (5, 12) = 2537, P < 0.0001) compared to their physical combination counterpart. Similarly, 500 µg/mL dual l-carnosine/A. vera nanophytosome-treated HUVECs demonstrated a superior tube formation capacity (15 ± 3 vs 2 ± 0.3; F (5, 12) = 30.87, P < 0.001), wound scratch healing capability (4.92 ± 0.3 vs 3.07 ± 0.3 mm/h; F (5, 12) = 39.21, P < 0.0001), and transwell migration (586 ± 32 vs 394 ± 18; F (5, 12) = 231.8, P < 0.001) and invasion (172 ± 9 vs 115 ± 5; F (5, 12) = 581.1, P < 0.0001) activities compared to the physical combination treated ones. Further confirming the proangiogenic activity of the dual l-carnosine/A. vera nanophytosomes, a significant shift toward expression of proangiogenic genes including HIF-1α, VEGFA, bFGF, KDR, and Ang II was reported in treated HUVECs. Overall, dual l-carnosine/A. vera nanophytosomes could be a potential candidate for attenuating type II DM-associated microvascular complications with an impaired angiogenesis background.


Carnosine/pharmacology , Diabetic Angiopathies/drug therapy , Nanoparticles/therapeutic use , Neovascularization, Physiologic/drug effects , Plant Extracts/pharmacology , Aloe/chemistry , Carnosine/therapeutic use , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Drug Synergism , Human Umbilical Vein Endothelial Cells , Humans , Microvessels/drug effects , Plant Extracts/therapeutic use , Pyruvaldehyde/metabolism , Pyruvaldehyde/toxicity
...