Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.282
1.
Endocrinol Diabetes Metab ; 7(3): e488, 2024 May.
Article En | MEDLINE | ID: mdl-38718275

INTRODUCTION: This study aimed to assess and compare the prevalence of diabetes complications between men and women with Type 2 diabetes (T2D), as well as how gender relates to these complications. METHODS: In this cross-sectional study, complications of diabetes, including coronary artery disease (CAD), retinopathy, neuropathy and diabetic kidney disease (DKD), were evaluated in 1867 participants with T2D. Additionally, baseline characteristics of the individuals, including anthropometric measurements, metabolic parameters and the use of dyslipidaemia drugs and antihyperglycaemic agents, were assessed. Gender differences in complications were examined using the chi-squared test. Multivariate logistic regression was employed to investigate the relationship between gender and T2D complications, with and without adjusting for the characteristics of the studied population. RESULTS: In the studied population, 62.1% had at least one complication, and complications were 33.5% for DKD, 29.6% for CAD, 22.9% for neuropathy and 19.1% for retinopathy. The prevalence of CAD and neuropathy was higher in men. However, DKD and retinopathy were more prevalent among women. Odds ratios of experiencing any complication, CAD and retinopathy in men compared with women were 1.57 (95% CI: 1.27-2.03), 2.27 (95% CI: 1.72-2.99) and 0.72 (95% CI: 0.52-0.98), respectively, after adjusting for demographic factors, anthropometric measures, metabolic parameters and the consumption of dyslipidaemia drugs and antihyperglycaemic agents. CONCLUSION: The prevalence of diabetes complications was significantly higher in men with diabetes, highlighting the need for better treatment adherence. CAD was associated with the male gender, whereas retinopathy was associated with the female gender. Men and women with diabetes should be monitored closely for CAD and retinopathy, respectively, regardless of their age, diabetes duration, anthropometric measures, laboratory findings and medications.


Diabetes Mellitus, Type 2 , Diabetic Retinopathy , Humans , Male , Female , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Cross-Sectional Studies , Middle Aged , Aged , Prevalence , Sex Factors , Diabetic Retinopathy/etiology , Diabetic Retinopathy/epidemiology , Diabetes Complications/etiology , Diabetes Complications/epidemiology , Adult , Diabetic Neuropathies/etiology , Diabetic Neuropathies/epidemiology , Diabetic Nephropathies/etiology , Diabetic Nephropathies/epidemiology , Coronary Artery Disease/etiology
2.
Medicine (Baltimore) ; 103(19): e38051, 2024 May 10.
Article En | MEDLINE | ID: mdl-38728488

This study aimed to explore the potential correlation between atherosclerotic cardiovascular disease (ASCVD) and diabetic retinopathy (DR) in patients with type 2 diabetes mellitus (T2DM). We enrolled 6540 patients with T2DM who were receiving chronic disease management for hypertension, hyperglycemia, and hyperlipidemia in Chengyang District of Qingdao. Among them, 730 had ASCVD (ASCVD group), which 5810 did not (N-ASCVD group). The results showed significantly higher levels of age, blood glucose, glycosylated hemoglobin (HbA1c), systolic blood pressure, ASCVD family history, female proportion, and DR incidence in the N-ASCVD group. Additionally, the glomerular filtration rate was significantly lower in the ASCVD group. Logistic regression analysis revealed a positive correlation between DR and ASCVD risk. DR was further categorized into 2 subtypes, nonproliferative DR (NPDR) and proliferative DR (PDR), based on e lesion severity. Interestingly, only the PDR was associated with ASCVD. Even after accounting for traditional ASCVD risk factors such as age, sex, and family history, PDR remained associated with ASCVD, with a staggering 718% increase in the risk for patients with PDR. Therefore, there is a strong association between ASCVD and DR in individuals with T2DM, with PDR particularly exhibiting an independent and positive correlation with increased ASCVD risk.


Atherosclerosis , Diabetes Mellitus, Type 2 , Diabetic Retinopathy , Humans , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Female , Male , Diabetic Retinopathy/epidemiology , Diabetic Retinopathy/etiology , Middle Aged , Atherosclerosis/epidemiology , Atherosclerosis/etiology , Aged , Risk Factors , China/epidemiology , Glycated Hemoglobin/analysis , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/etiology , Blood Glucose/analysis , Blood Glucose/metabolism , Incidence
3.
BMC Endocr Disord ; 24(1): 65, 2024 May 10.
Article En | MEDLINE | ID: mdl-38730329

OBJECTIVE: Diabetic patients are often comorbid with dyslipidemia, however, the relationship between high-density lipoprotein cholesterol(HDL-C) and diabetic retinopathy (DR) in the adult diabetic population remains to be fully elucidated.The aim of this study is to evaluate the associations between HDL-C and DR in the United States adults with diabetes. METHODS: A total of 1708 participants from the National Health and Nutrition Examination Survey (NHANES) 2005-2008 were enrolled in the present study. Fundus images of all study subjects were captured and evaluated using a digital camera and an ophthalmic digital imaging system, and the diagnosis of DR was made by the severity scale of the Early Treatment Diabetic Retinopathy Study (ETDRS).Roche Diagnostics were used to measure serum HDL-C concentration. The relationship of DR with HDL-C was investigated using multivariable logistic regression. The potential non-line correlation was explored with smooth curve fitting approach. RESULTS: The fully-adjusted model showed that HDL-C positively correlated with DR(OR:1.69, 95%CI: 1.25-2.31).However, an inverted U-shaped association between them was observed by applying the smooth curve fitted method. The inflection point of HDL-C(1.99mmol/l) was calculated by utilizing the two-piecewise logistic regression model. In the subgroup analysis, the inverted U-shaped nonlinear correlation between HDL-C and DR was also found in female, Non-Hispanic White, and lower age groups. CONCLUSION: Our study revealed an inverted U-shaped positive relationship between HDL-C and DR.The findings may provide us with a more comprehensive understanding of the association between HDL-C and DR.


Cholesterol, HDL , Diabetic Retinopathy , Humans , Diabetic Retinopathy/blood , Diabetic Retinopathy/epidemiology , Diabetic Retinopathy/etiology , Female , Male , Cholesterol, HDL/blood , Cross-Sectional Studies , Middle Aged , Retrospective Studies , Nutrition Surveys , Adult , Aged , Risk Factors , United States/epidemiology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications
4.
BMJ Open Diabetes Res Care ; 12(3)2024 May 06.
Article En | MEDLINE | ID: mdl-38719508

INTRODUCTION: To study the HbA1c trajectory from the time of diagnosis to examine if patients at the greatest risk for severe microangiopathy can be identified early allowing clinicians to intervene as soon as possible to avoid complications. RESEARCH DESIGN AND METHODS: In a population-based observational study, 447 patients diagnosed with type 1 diabetes before 35 years of age, 1983-1987, were followed from diagnosis until 2019. Mean HbA1c was calculated each year for each patient. Severe diabetic microangiopathy was defined as proliferative diabetic retinopathy (PDR) or macroalbuminuria (nephropathy). RESULTS: After 32 years, 27% had developed PDR and 8% macroalbuminuria. Patients with weighted HbA1c (wHbA1c); <57 mmol/mol; <7.4% did not develop PDR or macroalbuminuria. The HbA1c trajectories for patients developing PDR and macroalbuminuria follow separate courses early on and stay separated for 32 years during the follow-up. Patients without severe complications show an initial dip, after which HbA1c slowly increases. HbA1c in patients with severe complications directly rises to a high level within a few years. Mean HbA1c calculated for the period 5-8 years after diabetes onset strongly predicts the development of severe complications. Females with childhood-onset diabetes exhibit a high peak in HbA1c during adolescence associated with higher wHbA1c and higher prevalence of PDR. CONCLUSIONS: The HbA1c trajectory from diabetes onset shows that mean HbA1c for the period 5-8 years after diagnosis strongly predicts severe microangiopathy. Females with childhood-onset diabetes exhibit a high peak in HbA1c during adolescence associated with higher wHbA1c and a higher prevalence of PDR.


Diabetes Mellitus, Type 1 , Diabetic Angiopathies , Glycated Hemoglobin , Humans , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/epidemiology , Female , Male , Glycated Hemoglobin/analysis , Adult , Adolescent , Diabetic Angiopathies/epidemiology , Diabetic Angiopathies/etiology , Young Adult , Follow-Up Studies , Child , Diabetic Retinopathy/epidemiology , Diabetic Retinopathy/etiology , Prognosis , Biomarkers/blood , Albuminuria/epidemiology , Risk Factors , Child, Preschool , Diabetic Nephropathies/epidemiology , Diabetic Nephropathies/etiology , Disease Progression , Severity of Illness Index
5.
Front Endocrinol (Lausanne) ; 15: 1340608, 2024.
Article En | MEDLINE | ID: mdl-38699385

Objectives: This study aims to investigate the causal relationship between Alzheimer's Disease (AD) and Diabetic Retinopathy (DR). Methods: Employing Mendelian Randomization (MR), Generalized Summary-data-based Mendelian Randomization (GSMR), and the MR-Steiger test, this study scrutinizes the genetic underpinnings of the hypothesized causal association between AD and DR, as well as its Proliferative DR (PDR) and Non-Proliferative DR (NPDR) subtypes. Comprehensive data from Genome-Wide Association Studies (GWAS) were analyzed, specifically AD data from the Psychiatric Genomics Consortium (71,880 cases/383,378 controls), and DR, PDR, and NPDR data from both the FinnGen consortium (FinnGen release R8, DR: 5,988 cases/314,042 controls; PDR: 8,383 cases/329,756 controls; NPDR: 3,446 cases/314,042 controls) and the IEU OpenGWAS (DR: 14,584 cases/176,010 controls; PDR: 8,681 cases/204,208 controls; NPDR: 2,026 cases/204,208 controls). The study also incorporated Functional Mapping and Annotation (FUMA) for an in-depth analysis of the GWAS results. Results: The MR analyses revealed that genetic susceptibility to AD significantly increases the risk of DR, as evidenced by GWAS data from the FinnGen consortium (OR: 2.5090; 95% confidence interval (CI):1.2102-5.2018, false discovery rate P-value (PFDR)=0.0201; GSMR: bxy=0.8936, bxy_se=0.3759, P=0.0174), NPDR (OR: 2.7455; 95% CI: 1.3178-5.7197, PFDR=0.0166; GSMR: bxy=0.9682, bxy_se=0.3802, P=0.0126), and PDR (OR: 2.3098; 95% CI: 1.2411-4.2986, PFDR=0.0164; GSMR: bxy=0.7962, bxy_se=0.3205, P=0.0129) using DR GWAS from FinnGen consortium. These results were corroborated by DR GWAS datasets from IEU OpenGWAS. The MR-Steiger test confirmed a significant association of all identified instrumental variables (IVs) with AD. While a potential causal effect of DR and its subtypes on AD was identified, the robustness of these results was constrained by a low power value. FUMA analysis identified OARD1, NFYA, TREM1 as shared risk genes between DR and AD, suggesting a potential genetic overlap between these complex diseases. Discussion: This study underscores the contribution of AD to an increased risk of DR, as well as NPDR and PDR subtypes, underscoring the necessity of a holistic approach in the management of patients affected by these conditions.


Alzheimer Disease , Diabetic Retinopathy , Genome-Wide Association Study , Mendelian Randomization Analysis , Humans , Alzheimer Disease/genetics , Risk Factors , Diabetic Retinopathy/genetics , Diabetic Retinopathy/epidemiology , Diabetic Retinopathy/etiology , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide
6.
Nutrients ; 16(9)2024 May 05.
Article En | MEDLINE | ID: mdl-38732638

As the most serious of the many worse new pathological changes caused by diabetes, there are many risk factors for the occurrence and development of diabetic retinopathy (DR). They mainly include hyperglycemia, hypertension, hyperlipidemia and so on. Among them, hyperglycemia is the most critical cause, and plays a vital role in the pathological changes of DR. High-sucrose diets (HSDs) lead to elevated blood glucose levels in vivo, which, through oxidative stress, inflammation, the production of advanced glycation end products (AGEs) and vascular endothelial growth factor (VEGF), cause plenty of pathological damages to the retina and ultimately bring about loss of vision. The existing therapies for DR primarily target the terminal stage of the disease, when irreversible visual impairment has appeared. Therefore, early prevention is particularly critical. The early prevention of DR-related vision loss requires adjustments to dietary habits, mainly by reducing sugar intake. This article primarily discusses the risk factors, pathophysiological processes and molecular mechanisms associated with the development of DR caused by HSDs. It aims to raise awareness of the crucial role of diet in the occurrence and progression of DR, promote timely changes in dietary habits, prevent vision loss and improve the quality of life. The aim is to make people aware of the importance of diet in the occurrence and progression of DR. According to the dietary modification strategies that we give, patients can change their poor eating habits in a timely manner to avoid theoretically avoidable retinopathy and obtain an excellent prognosis.


Diabetic Retinopathy , Disease Progression , Humans , Diabetic Retinopathy/etiology , Diabetic Retinopathy/prevention & control , Risk Factors , Dietary Sucrose/adverse effects , Oxidative Stress , Blood Glucose/metabolism , Diet/adverse effects , Feeding Behavior , Glycation End Products, Advanced/metabolism , Glycation End Products, Advanced/adverse effects
7.
Cell Biochem Funct ; 42(4): e4037, 2024 Jun.
Article En | MEDLINE | ID: mdl-38736204

Diabetes mellitus is associated with secondary complications such as diabetic retinopathy (DR), nephropathy (DN), and cardiomyopathy (DCM), all of which significantly impact patient health. Intercellular adhesion molecule-1 (ICAM-1) has been implicated in inflammatory responses and endothelial dysfunction, both crucial in the pathogenesis of these complications. The goal of this review is to investigate at potential therapy methods that target ICAM-1 pathways and to better understand the multifaceted role of ICAM-1 in secondary diabetic problems. A meticulous analysis of scholarly literature published globally was conducted to examine ICAM-1involvement in inflammatory processes, endothelial dysfunction, and oxidative stress related to diabetes and its complications. Elevated ICAM-1 levels are strongly associated with augmented leukocyte adhesion, compromised microvascular function, and heightened oxidative stress in diabetes. These pathways contribute significantly to DR, DN, and DCM pathogenesis, highlighting ICAM-1 as a key player in their progression. Understanding ICAM-1 role in secondary diabetic complications offers insights into novel therapeutic strategies. Targeting ICAM-1 pathways may mitigate inflammation, improve endothelial function, and ultimately attenuate diabetic complications, thereby enhancing patient health outcomes. Continued research in this area is crucial for developing effective targeted therapies.


Intercellular Adhesion Molecule-1 , Humans , Intercellular Adhesion Molecule-1/metabolism , Diabetes Complications/metabolism , Oxidative Stress , Animals , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Inflammation/metabolism , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Diabetic Retinopathy/etiology
8.
Diabetes Metab Res Rev ; 40(4): e3812, 2024 May.
Article En | MEDLINE | ID: mdl-38738481

AIMS: To evaluate the effectiveness of optical coherence tomography angiography (OCTA) in detecting early intraocular microvascular changes in diabetic patients. MATERIALS AND METHODS: A systematic study search was performed on PubMed, Medline, Embase, and the Cochrane Library, ranging from January 2012 to March 2023. Controlled studies compared diabetes mellitus (DM) patients with non-diabetic retinopathy (NDR) or patients with mild non-proliferative diabetic retinopathy (mild NPDR) to healthy people. These studies included parameters of OCTA such as foveal avascular zone (FAZ), vessel density of superficial capillary plexus (VDscp), vessel density of deep capillary plexus (VDdcp), and peripapillary VD. The relevant effect model was used according to the heterogeneity, and the mean difference and 95% confidence intervals were calculated. RESULTS: A total of 18 studies with 2101 eyes were eventually included in this meta-analysis. Our results demonstrated that early alterations of VDscp, VDdcp, and peripapillary VD in NDR patients had a significant difference compared with healthy people by OCTA (VDscp: WMD = -1.34, 95% CI: -1.99 to -0.68, P < 0.0001. VDdcp: WMD = -2.00, 95% CI: -2.95 to -1.04, P < 0.0001. Peripapillary VD: WMD = -1.07, 95% CI: -1.70 to -0.43, P = 0.0010). However, there was no statistically significant difference in total FAZ between them (WMD = -0.00, 95% CI: -0.02-0.01, P = 0.84). In addition, for patients with mild NPDR, OCTA could illustrate prominent changes in VDscp, VDdcp, and total FAZ compared with healthy people (VDscp: WMD = -6.11, 95% CI: -9.90 to -2.32, P = 0.002. VDdcp: WMD = -4.26, 95% CI: -5.95 to -2.57, P < 0.00001. FAZ: WMD = 0.06, 95% CI: 0.01-0.11, P = 0.03). CONCLUSIONS: In diabetic patients with or without retinopathy, the parameters of OCTA such as VDscp, VDdcp, and peripapillary vessel density were demonstrated as potential biomarkers in monitoring the early alterations of retinal microangiopathy, while total FAZ may have no significant changes in diabetic patients without retinopathy.


Diabetic Retinopathy , Retinal Vessels , Tomography, Optical Coherence , Humans , Tomography, Optical Coherence/methods , Diabetic Retinopathy/diagnostic imaging , Diabetic Retinopathy/etiology , Retinal Vessels/diagnostic imaging , Retinal Vessels/pathology , Fluorescein Angiography/methods , Microvessels/diagnostic imaging , Microvessels/pathology , Diabetes Mellitus/diagnostic imaging , Prognosis
9.
BMJ Open Diabetes Res Care ; 12(2)2024 Apr 11.
Article En | MEDLINE | ID: mdl-38604732

INTRODUCTION: Diabetic retinopathy (DR), diabetic kidney disease (DKD) and distal symmetric polyneuropathy (DSPN) share common pathophysiology and pose an additive risk of early mortality. RESEARCH DESIGN AND METHODS: In adults with type 1 diabetes, 49 metabolites previously associated with either DR or DKD were assessed in relation to presence of DSPN. Metabolites overlapping in significance with presence of all three complications were assessed in relation to microvascular burden severity (additive number of complications-ie, presence of DKD±DR±DSPN) using linear regression models. Subsequently, the same metabolites were assessed with progression to endpoints: soft microvascular events (progression in albuminuria grade, ≥30% estimated glomerular filtration rate (eGFR) decline, or any progression in DR grade), hard microvascular events (progression to proliferative DR, chronic kidney failure, or ≥40% eGFR decline), and hard microvascular or macrovascular events (hard microvascular events, cardiovascular events (myocardial infarction, stroke, or arterial interventions), or cardiovascular mortality), using Cox models. All models were adjusted for sex, baseline age, diabetes duration, systolic blood pressure, HbA1c, body mass index, total cholesterol, smoking, and statin treatment. RESULTS: The full cohort investigated consisted of 487 participants. Mean (SD) follow-up was 4.8 (2.9, 5.7) years. Baseline biothesiometry was available in 202 participants, comprising the cross-sectional cohort. Eight metabolites were significantly associated with presence of DR, DKD, and DSPN, and six with additive microvascular burden severity. In the full cohort longitudinal analysis, higher levels of 3,4-dihydroxybutanoic acid (DHBA), 2,4-DHBA, ribonic acid, glycine, and ribitol were associated with development of events in both crude and adjusted models. Adding 3,4-DHBA, ribonic acid, and glycine to a traditional risk factor model improved the discrimination of hard microvascular events. CONCLUSIONS: While prospective studies directly assessing the predictive ability of these markers are needed, our results strengthen the role of clinical metabolomics in relation to risk assessment of diabetic complications in chronic type 1 diabetes.


Diabetes Mellitus, Type 1 , Diabetic Neuropathies , Diabetic Retinopathy , Adult , Humans , Diabetes Mellitus, Type 1/complications , Prospective Studies , Cross-Sectional Studies , Diabetic Retinopathy/etiology , Diabetic Retinopathy/complications , Diabetic Neuropathies/complications , Glycine
10.
Diabetes Care ; 47(5): 873-880, 2024 May 01.
Article En | MEDLINE | ID: mdl-38470988

OBJECTIVE: The impact of the difference between cystatin C- and creatinine-based estimated glomerular filtration rate (eGFRdiff) on diabetic microvascular complications (DMCs) remains unknown. We investigated the associations of eGFRdiff with overall DMCs and subtypes, including diabetic retinopathy (DR), diabetic kidney disease (DKD), and diabetic neuropathy (DN). RESEARCH DESIGN AND METHODS: This prospective cohort study included 25,825 participants with diabetes free of DMCs at baseline (2006 to 2010) from the UK Biobank. eGFRdiff was calculated using both absolute difference (eGFRabdiff) and the ratio (eGFRrediff) between cystatin C- and creatinine-based calculations. Incidence of DMCs was ascertained using electronic health records. Cox proportional hazards regression models were used to evaluate the associations of eGFRdiff with overall DMCs and subtypes. RESULTS: During a median follow-up of 13.6 years, DMCs developed in 5,753 participants, including 2,752 cases of DR, 3,203 of DKD, and 1,149 of DN. Each SD decrease of eGFRabdiff was associated with a 28% higher risk of overall DMCs, 14% higher risk of DR, 56% higher risk of DKD, and 29% higher risk of DN. For each 10% decrease in eGFRrediff, the corresponding hazard ratios (95% CIs) were 1.16 (1.14, 1.18) for overall DMCs, 1.08 (1.05, 1.11) for DR, 1.29 (1.26, 1.33) for DKD, and 1.17 (1.12, 1.22) for DN. The magnitude of associations was not materially altered in any of the sensitivity analyses. CONCLUSIONS: Large eGFRdiff was independently associated with risk of DMCs and its subtypes. Our findings suggested monitoring eGFRdiff in the diabetes population has potential benefit for identification of high-risk patients.


Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Diabetic Neuropathies , Diabetic Retinopathy , Adult , Humans , Cystatin C , Creatinine , Cohort Studies , Prospective Studies , Glomerular Filtration Rate , Diabetic Nephropathies/etiology , Diabetic Retinopathy/etiology , Diabetic Retinopathy/complications , Diabetic Neuropathies/complications , Risk Factors , Diabetes Mellitus, Type 2/complications
11.
Vestn Oftalmol ; 140(1): 45-56, 2024.
Article Ru | MEDLINE | ID: mdl-38450466

In recent years, there has been a growing interest in the contribution of neuroretinal degeneration to the pathogenesis of diabetic retinopathy (DR). PURPOSE: This study assesses the effect of the drug Retinalamin on the functional state of the retina in patients with DR using the Diopsys NOVA Vision Testing System that utilizes electrophysiological (EP) technology. MATERIAL AND METHODS: The study included patients with type 1 and 2 diabetes mellitus (DM) with DR of any stage without macular edema. Patients underwent standard ophthalmological examination and objective functional examination of the retina using the Diopsys NOVA Vision Testing System. The control group consisted of patients with type 1 and 2 DM with DR who did not receive Retinalamin. RESULTS: Significant changes in pattern electroretinography and flash electroretinography parameters were recorded in patients who received a course of Retinalamin. Two clinical examples are presented, which can be designated as the first experience of objective functional monitoring of treatment of patients with DR with Retinalamin. CONCLUSION: Retinoprotective therapy is necessary already at the early stages of DR. Electroretinography is an objective tool for functional analysis of the earliest changes in retinal cells in DR. It is necessary to use the identified "therapeutic" window for the appointment of retinoprotective agents.


Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Diabetic Retinopathy , Humans , Diabetic Retinopathy/diagnosis , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/etiology , Retina/diagnostic imaging , Electroretinography
12.
J Pediatr Endocrinol Metab ; 37(4): 341-346, 2024 Apr 25.
Article En | MEDLINE | ID: mdl-38487852

OBJECTIVES: This study aimed to identify discrepancies in the retinal nerve fiber layer (RNFL) between type 1 diabetes mellitus (T1DM) children without retinopathy and healthy subjects in northern China. METHODS: This was a cross-sectional hospital-based study carried out from Jan 2019 until Jul 2021 at the department of pediatrics in Tianjin medical university general hospital. Children with T1DM but no retinal disease were screened. RNFL thickness was obtained via spectral domain optical coherence tomography. Disease duration, HbA1c, 25-hydroxyvitamin D level, insulin regimen, and diet control status were also collected. RESULTS: A total of 20 children with T1DM and 20 matched health participants were enrolled. The mean age in the T1DM group was 10.3 ± 2.8 years, and the median duration of diabetes was 1 (range 1-3) year. Children with T1DM had thinner average RNFL than control subjects (105 ± 6 vs. 110 ± 11 µm, p=0.008), especially in temporal and nasal parts. There was a significant negative association between HbA1c levels and the RNFL thickness in the T1DM group (B (95 % confidence interval): -4.313 (-7.055 to -1.571); p=0.005). CONCLUSIONS: In our study, the decreased thickness of RNFL was negatively associated with elevated HbA1c in children with early stages of T1DM.


Diabetes Mellitus, Type 1 , Diabetic Retinopathy , Humans , Child , Adolescent , Cross-Sectional Studies , Retinal Ganglion Cells , Diabetic Retinopathy/epidemiology , Diabetic Retinopathy/etiology , Nerve Fibers , Tomography, Optical Coherence/methods , China/epidemiology
15.
Front Public Health ; 12: 1347671, 2024.
Article En | MEDLINE | ID: mdl-38351959

Introduction: A few past experimental studies have indicated that exposure to volatile organic compounds (VOCs) might be a potential risk factor for diabetes retinopathy (DR). However, these findings lack substantial support from extensive epidemiological research. This large-scale cross-sectional study aimed to examine whether exposure to low levels of VOCs in the general population is associated with diabetes mellitus (DM) and DR. Methods: The analytical data was from the National Health and Nutrition Examination Survey (NHANES) dataset (2011-2018). To minimize the potential impact of gender and age on the findings, propensity score matching was utilized to align the data selection. Relationships between blood VOCs and DM and DR were assessed in a sample of 2,932 adults using the logistic regression models. Additionally, Bayesian kernel machine regression (BKMR) models and Weighted Quantile Sum (WQS) were conducted for mixture exposure analysis. Results: The result shows VOCs were positive associated with DM and DR in US adults, as assessed by WQS model, and the calculated odd ratios (ORs) [95% confidence interval (C.I)] were 53.91(34.11 ~ 85.22) and 7.38(3.65 ~ 14.92), respectively. Among the components of VOCs, 1,2-Dibromoethane, Carbon Tetrachloride and 2,5-Dimethylfuran were positive related with the DR, and ORs (95%C.I) were 2.91(2.29 ~ 3.70), 2.86(2.25 ~ 3.65) and 2.19(1.79 ~ 2.94), respectively. BKMR model shows that there was a dose-response relationship between combined VOCs and DR, although the relationship was non-linearly. Conclusion: This study suggested that exposure to VOCs may increase the risk of DR, which had important public health implications.


Diabetes Mellitus , Diabetic Retinopathy , Volatile Organic Compounds , Adult , Humans , Nutrition Surveys , Cross-Sectional Studies , Volatile Organic Compounds/adverse effects , Environmental Exposure/adverse effects , Bayes Theorem , Diabetic Retinopathy/epidemiology , Diabetic Retinopathy/etiology , Risk Factors
16.
Diabetes Res Clin Pract ; 209: 111560, 2024 Mar.
Article En | MEDLINE | ID: mdl-38316188

AIMS: With growing concerns over complications in diabetes sufferers, this study sought to develop an interpretable machine learning model to offer enhanced diagnostic and treatment recommendations. METHODS: We assessed coronary heart disease, diabetic nephropathy, diabetic retinopathy, and fatty liver disease using logistic regression, decision tree, random forest, and CatBoost algorithms. The SHAP algorithm was employed to elucidate the model's predictions, offering a more in-depth understanding of influential features. RESULTS: The CatBoost model notably outperformed other algorithms in AUC, achieving an average AUC of 90.47 % for the four complications. Through SHAP analysis and visualization, we provided clear and actionable insights into risk factors, enabling better complication risk assessment. CONCLUSIONS: We introduced an innovative, interpretable complication risk model for people with diabetes. This not only offers a potent tool for healthcare professionals but also empowers sufferers with clearer self-assessment capabilities, encouraging earlier preventive actions. Further studies will underscore the model's clinical applicability.


Diabetes Mellitus , Diabetic Nephropathies , Diabetic Retinopathy , Humans , Diabetic Retinopathy/diagnosis , Diabetic Retinopathy/epidemiology , Diabetic Retinopathy/etiology , Algorithms , Asian People , China/epidemiology
18.
JMIR Public Health Surveill ; 10: e48120, 2024 Feb 06.
Article En | MEDLINE | ID: mdl-38319705

BACKGROUND: Visceral adipose tissue plays an active role in the pathogenesis of type 2 diabetes and vascular dysfunction. The lipid accumulation product (LAP), visceral adiposity index (VAI), and Chinese VAI (CVAI) have been proposed as simple and validated surrogate indices for measuring visceral adipose tissue. However, the evidence from prospective studies on the associations between these novel indices of visceral obesity and diabetic retinopathy (DR) remains scant. OBJECTIVE: This study aimed to investigate the longitudinal associations of LAP, VAI, and CVAI with incident DR in Chinese patients with diabetes. METHODS: This was a prospective cohort study conducted in Guangzhou in southern China. We collected baseline data between November 2017 and July 2020, while on-site follow-up visits were conducted annually until January 2022. The study participants consisted of 1403 patients with a clinical diagnosis of diabetes, referred from primary care, who were free of DR at baseline. The LAP, VAI, and CVAI levels were calculated by sex-specific equations based on anthropometric and biochemical parameters. DR was assessed using 7-field color stereoscopic fundus photographs and graded according to the modified Airlie House Classification scheme. Time-dependent Cox proportional hazard models were constructed to estimate the hazard ratios with 95% CIs. Restricted cubic spline curves were fitted to examine the dose-response relationship between the 3 indices of visceral obesity and new-onset DR. Subgroup analyses were performed to investigate the potential effect modifiers. RESULTS: The mean age of study participants was 64.5 (SD 7.6) years, and over half (816/1403, 58.2%) were female. During a median follow-up of 2.13 years, 406 DR events were observed. A 1-SD increment in LAP, VAI, or CVAI was consistently associated with increased risk for new-onset DR, with a multivariable­adjusted hazard ratio of 1.24 (95% CI 1.09-1.41; P=.001), 1.22 (95% CI 1.09-1.36; P<.001), and 1.48 (95% CI 1.19-1.85; P=.001), respectively. Similar patterns were observed across tertiles in LAP (P for trend=.001), VAI (P for trend<.001), and CVAI (P for trend=.009). Patients in the highest tertile of LAP, VAI, and CVAI had an 84%, 86%, and 82% higher hazard of DR, respectively, compared to those in the lowest tertile. A nonlinear dose-response relationship with incident DR was noted for LAP and VAI (both P for nonlinearity<.05), but not for CVAI (P for nonlinearity=.51). We did not detect the presence of effect modification by age, sex, duration of diabetes, BMI, or comorbidity (all P for interaction>.10). CONCLUSIONS: Visceral obesity, as measured by LAP, VAI, or CVAI, is independently associated with increased risk for new-onset DR in Chinese patients with diabetes. Our findings may suggest the necessity of incorporating regular monitoring of visceral obesity indices into routine clinical practice to enhance population-based prevention for DR.


Diabetes Mellitus, Type 2 , Diabetic Retinopathy , Obesity, Abdominal , Female , Humans , Male , Middle Aged , Asian People , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Diabetic Retinopathy/diagnosis , Diabetic Retinopathy/epidemiology , Diabetic Retinopathy/etiology , Obesity, Abdominal/complications , Obesity, Abdominal/epidemiology , Prospective Studies , Aged , China
19.
Front Endocrinol (Lausanne) ; 15: 1292412, 2024.
Article En | MEDLINE | ID: mdl-38344659

Context: Diabetic retinopathy (DR) and diabetic nephropathy (DN), are major microvascular complications of diabetes. DR is an important predictor of DN, but the relationship between the severity of DR and the pathological severity of diabetic glomerulopathy remains unclear. Objective: To investigate the relationship between severity of diabetic retinopathy (DR) and histological changes and clinical indicators of diabetic nephropathy (DN) in patients with type 2 diabetes mellitus (T2DM). Methods: Patients with T2DM (n=272) who underwent a renal biopsy were eligible. Severity of DR was classified as non-diabetic retinopathy, non-proliferative retinopathy, and proliferative retinopathy (PDR). Relationship between DN and DR and the diagnostic efficacy of DR for DN were explored. Results: DN had a higher prevalence of DR (86.4%) and DR was more severe. The sensitivity and specificity of DR in DN were 86.4% and 78.8%, while PDR was 26.4% and 98.5%, respectively. In DN patients, the severity of glomerular lesions (p=0.001) and prevalence of KW nodules (p<0.001) significantly increased with increasing severity of DR. The presence of KW nodules, lower hemoglobin levels, and younger age were independent risk factors associated with more severe DR in patients with DN. Conclusion: DR was a good predictor of DN. In DN patients, the severity of DR was associated with glomerular injury, and presence of KW nodules, lower hemoglobin levels and younger age were independent risk factors associated with more severe DR. Trial registration: ClinicalTrails.gov, NCT03865914.


Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Diabetic Retinopathy , Humans , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/epidemiology , Diabetic Nephropathies/etiology , Diabetes Mellitus, Type 2/complications , Diabetic Retinopathy/epidemiology , Diabetic Retinopathy/etiology , Diabetic Retinopathy/diagnosis , Risk Factors , Hemoglobins
20.
Sci Rep ; 14(1): 4054, 2024 02 19.
Article En | MEDLINE | ID: mdl-38374169

Diabetic retinopathy is a commonly observed cause of blindness and is a common problem in individuals with diabetes. Recent investigations have showed the capability of serum α-Klotho and FGF 23 in mitigating the effects of diabetic retinopathy. This study aimed to discover the correlation between FGF 23, α-Klotho, and diabetic retinopathy in type 1 diabetics. This case-control study included 63 diabetic patients and 66 healthy controls. Following an overnight duration of fasting, morning blood samples were taken from both the patient and the control groups. The serum concentrations of α-Klotho and FGF 23 were quantified. An experienced ophthalmologist inspected the retinopathy. All participants in this study have moderate non-proliferative retinopathy. A p value under 0.05 was considered statistically significant. The mean α-Klotho level for retinopathic diabetic patients was 501.7 ± 172.2 pg/mL and 579.6 ± 312.1 pg/mL for non-retinopathic diabetic patients. In comparison, α-Klotho level of the control group was 523.2 ± 265.4 pg/mL (p = 0.531). The mean of FGF 23 level did not demonstrate a significant difference (p = 0.259). The mean FGF 23 level were 75.7 ± 14.0 pg/mL, 74.0 ± 14.8 pg/mL and 79.3 ± 14.4 pg/mL in groups, respectively. In conclusion, there was no significant difference in FGF 23 and α-Klotho levels between type 1 diabetics with and without retinopathy when compared to the control group.


Diabetes Mellitus, Type 1 , Diabetic Retinopathy , Fibroblast Growth Factor-23 , Klotho Proteins , Humans , Case-Control Studies , Diabetes Mellitus, Type 1/complications , Diabetic Retinopathy/etiology , Fibroblast Growth Factor-23/blood , Fibroblast Growth Factor-23/chemistry , Fibroblast Growth Factors/metabolism , Glucuronidase , Klotho Proteins/blood , Klotho Proteins/chemistry
...