1
|
Savelieff MG, Elafros MA, Viswanathan V, Jensen TS, Bennett DL, Feldman EL. The global and regional burden of diabetic peripheral neuropathy. Nat Rev Neurol 2025; 21:17-31. [PMID: 39639140 DOI: 10.1038/s41582-024-01041-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2024] [Indexed: 12/07/2024]
Abstract
Diabetic peripheral neuropathy (DPN) is length-dependent peripheral nerve damage arising as a complication of type 1 or type 2 diabetes in up to 50% of patients. DPN poses a substantial burden on patients, who can experience impaired gait and loss of balance, predisposing them to falls and fractures, and neuropathic pain, which is frequently difficult to treat and reduces quality of life. Advanced DPN can lead to diabetic foot ulcers and non-healing wounds that often necessitate lower-limb amputation. From a socioeconomic perspective, DPN increases both direct health-care costs and indirect costs from loss of productivity owing to neuropathy-related disability. In this Review, we highlight the importance of understanding country-specific and region-specific variations in DPN prevalence to inform public health policy and allocate resources appropriately. We also explore how identification of DPN risk factors can guide treatment and prevention strategies and aid the development of health-care infrastructure for populations at risk. We review evidence that metabolic factors beyond hyperglycaemia contribute to DPN development, necessitating a shift from pure glycaemic control to multi-targeted metabolic control, including weight loss and improvements in lipid profiles.
Collapse
Affiliation(s)
- Masha G Savelieff
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Melissa A Elafros
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
| | - Vijay Viswanathan
- MV Hospital for Diabetes and Prof. M. Viswanathan Diabetes Research Centre, Royapuram, Chennai, India
| | - Troels S Jensen
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
- Danish Pain Research Center, Aarhus University, Aarhus, Denmark
| | - David L Bennett
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
2
|
Wu RL, Chen N, Chen Y, Wu X, Ko CY, Chen XY. Visceral Adiposity as an Independent Risk Factor for Diabetic Peripheral Neuropathy in Type 2 Diabetes Mellitus: A Retrospective Study. J Diabetes Res 2024; 2024:9912907. [PMID: 39559714 PMCID: PMC11573447 DOI: 10.1155/2024/9912907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024] Open
Abstract
Background: Diabetic peripheral neuropathy (DPN) impacts approximately 50% of individuals with Type 2 diabetes mellitus (T2DM), leading to severe complications such as foot ulcers and amputations. Notably, visceral adiposity is increasingly recognized as a pivotal factor in augmenting the risk of DPN. We aim to evaluate the correlation between obesity-related body composition, particularly visceral fat, and DPN to facilitate early identification of high-risk patients with T2DM. Methods: This cross-sectional analysis encompassed 113 T2DM patients from the Department of Endocrinology and Metabolism at the Second Affiliated Hospital of Fujian Medical University, conducted between September 2020 and January 2021. Patients were categorized into two cohorts: those with DPN (DPN group) and those without (NDPN group). We utilized bioelectrical impedance analysis (BIA) to determine body measurements, such as weight and visceral fat area, in addition to collecting clinical and biochemical data. Logistic regression was employed to analyze the data. Results: The study uncovered a statistically significant difference in the visceral fat area between the DPN and NDPN groups (p = 0.048). Through multivariate logistic regression analysis, the visceral fat area was identified as an independent risk factor for DPN among T2DM patients (OR 1.027; 95% CI 1.004-1.051, p = 0.022). Other significant risk factors included the duration of diabetes and the presence of diabetic retinopathy. Conclusion: The visceral fat area serves as an independent risk factor for DPN in individuals with T2DM. Implementing measures to assess and manage visceral obesity could be vital in the prevention and management of DPN. This underscores the value of technologies such as BIA in clinical and community settings for early intervention.
Collapse
Affiliation(s)
- Rui-Ling Wu
- Department of Clinical Nutrition, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Niyao Chen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yanni Chen
- Department of Endocrinology, Shishi General Hospital, Quanzhou, China
| | - Xiaohong Wu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Chih-Yuan Ko
- Department of Clinical Nutrition, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xiao-Yu Chen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
3
|
Elafros MA, Reynolds EL, Callaghan BC. Obesity-related neuropathy: the new epidemic. Curr Opin Neurol 2024; 37:467-477. [PMID: 38864534 PMCID: PMC11371529 DOI: 10.1097/wco.0000000000001292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
PURPOSE OF REVIEW To examine the evidence evaluating the association between obesity and neuropathy as well as potential interventions. RECENT FINDINGS Although diabetes has long been associated with neuropathy, additional metabolic syndrome components, including obesity, are increasingly linked to neuropathy development, regardless of glycemic status. Preclinical rodent models as well as clinical studies are shedding light on the mechanisms of obesity-related neuropathy as well as challenges associated with slowing progression. Dietary and surgical weight loss and exercise interventions are promising, but more data is needed. SUMMARY High-fat-diet rodent models have shown that obesity-related neuropathy is a product of excess glucose and lipid accumulation leading to inflammation and cell death. Clinical studies consistently demonstrate obesity is independently associated with neuropathy; therefore, likely a causal risk factor. Dietary weight loss improves neuropathy symptoms but not examination scores. Bariatric surgery and exercise are promising interventions, but larger, more rigorous studies are needed. Further research is also needed to determine the utility of weight loss medications and ideal timing for obesity interventions to prevent neuropathy.
Collapse
Affiliation(s)
| | - Evan Lee Reynolds
- Department of Epidemiology and Biostatistics, Michigan State University College of Human Medicine, East Lansing, Michigan, USA
| | | |
Collapse
|
4
|
Annicchiarico A, Barile B, Buccoliero C, Nicchia GP, Brunetti G. Alternative therapeutic strategies in diabetes management. World J Diabetes 2024; 15:1142-1161. [PMID: 38983831 PMCID: PMC11229975 DOI: 10.4239/wjd.v15.i6.1142] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/17/2024] [Accepted: 04/12/2024] [Indexed: 06/11/2024] Open
Abstract
Diabetes is a heterogeneous metabolic disease characterized by elevated blood glucose levels resulting from the destruction or malfunction of pancreatic β cells, insulin resistance in peripheral tissues, or both, and results in a non-sufficient production of insulin. To adjust blood glucose levels, diabetic patients need exogenous insulin administration together with medical nutrition therapy and physical activity. With the aim of improving insulin availability in diabetic patients as well as ameliorating diabetes comorbidities, different strategies have been investigated. The first approaches included enhancing endogenous β cell activity or transplanting new islets. The protocol for this kind of intervention has recently been optimized, leading to standardized procedures. It is indicated for diabetic patients with severe hypoglycemia, complicated by impaired hypoglycemia awareness or exacerbated glycemic lability. Transplantation has been associated with improvement in all comorbidities associated with diabetes, quality of life, and survival. However, different trials are ongoing to further improve the beneficial effects of transplantation. Furthermore, to overcome some limitations associated with the availability of islets/pancreas, alternative therapeutic strategies are under evaluation, such as the use of mesenchymal stem cells (MSCs) or induced pluripotent stem cells for transplantation. The cotransplantation of MSCs with islets has been successful, thus providing protection against proinflammatory cytokines and hypoxia through different mechanisms, including exosome release. The use of induced pluripotent stem cells is recent and requires further investigation. The advantages of MSC implantation have also included the improvement of diabetes-related comorbidities, such as wound healing. Despite the number of advantages of the direct injection of MSCs, new strategies involving biomaterials and scaffolds have been developed to improve the efficacy of mesenchymal cell delivery with promising results. In conclusion, this paper offered an overview of new alternative strategies for diabetes management while highlighting some limitations that will need to be overcome by future approaches.
Collapse
Affiliation(s)
- Alessia Annicchiarico
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| | - Barbara Barile
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| | - Cinzia Buccoliero
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| | - Grazia Paola Nicchia
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| | - Giacomina Brunetti
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| |
Collapse
|
5
|
Määttä LL, Andersen ST, Parkner T, Hviid CVB, Bjerg L, Kural MA, Charles M, Søndergaard E, Kuhle J, Tankisi H, Witte DR, Jensen TS. Longitudinal Change in Serum Neurofilament Light Chain in Type 2 Diabetes and Early Diabetic Polyneuropathy: ADDITION-Denmark. Diabetes Care 2024; 47:986-994. [PMID: 38502878 DOI: 10.2337/dc23-2208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/22/2024] [Indexed: 03/21/2024]
Abstract
OBJECTIVE To investigate the longitudinal development of neurofilament light chain (NfL) levels in type 2 diabetes with and without diabetic polyneuropathy (+/-DPN) and to explore the predictive potential of NfL as a biomarker for DPN. RESEARCH DESIGN AND METHODS We performed retrospective longitudinal case-control analysis of data from 178 participants of the Anglo-Danish-Dutch Study of Intensive Treatment in People with Screen-Detected Diabetes in Primary Care-Denmark (ADDITION-Denmark) cohort of people with screen-detected type 2 diabetes. Biobank samples acquired at the ADDITION-Denmark 5- and 10-year follow-ups were analyzed for serum NfL (s-NfL) using single-molecule array, and the results were compared with established reference material to obtain NfL z-scores. DPN was diagnosed according to Toronto criteria for confirmed DPN at the 10-year follow-up. RESULTS s-NfL increased over time in +DPN (N = 39) and -DPN participants (N = 139) at levels above normal age-induced s-NfL increase. Longitudinal s-NfL change was greater in +DPN than in -DPN participants (17.4% [95% CI 4.3; 32.2] or 0.31 SD [95% CI 0.03; 0.60] higher s-NfL or NfL z-score increase in +DPN compared with -DPN). s-NfL at the 5-year follow-up was positively associated with nerve conduction studies at the 10-year follow-up (P = 0.02 to <0.001), but not with DPN risk. Areas under the curve (AUCs) for s-NfL were not inferior to AUCs for the Michigan Neuropathy Screening Instrument questionnaire score or vibration detection thresholds. Higher yearly s-NfL increase was associated with higher DPN risk (odds ratio 1.36 [95% CI 1.08; 1.71] per 1 ng/L/year). CONCLUSIONS Our findings suggest that preceding s-NfL trajectories differ slightly between those with and without DPN and imply a possible biomarker value of s-NfL trajectories in DPN.
Collapse
Affiliation(s)
- Laura L Määttä
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Signe T Andersen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Tina Parkner
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Claus V B Hviid
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Lasse Bjerg
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Mustafa A Kural
- Department of Clinical Neurophysiology, Aarhus University Hospital, Aarhus, Denmark
| | - Morten Charles
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Esben Søndergaard
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Kuhle
- Department of Neurology, University of Basel, Basel, Switzerland
- Multiple Sclerosis Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel, University Hospital Basel, Basel, Switzerland
| | - Hatice Tankisi
- Department of Clinical Neurophysiology, Aarhus University Hospital, Aarhus, Denmark
| | - Daniel R Witte
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Troels S Jensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
6
|
Erandathi MA, Wang WYC, Mayo M, Lee CC. Comprehensive Factors for Predicting the Complications of DiabetesMellitus: A Systematic Review. Curr Diabetes Rev 2024; 20:e040124225240. [PMID: 38178670 PMCID: PMC11327746 DOI: 10.2174/0115733998271863231116062601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND This article focuses on extracting a standard feature set for predicting the complications of diabetes mellitus by systematically reviewing the literature. It is conducted and reported by following the guidelines of PRISMA, a well-known systematic review and meta-analysis method. The research articles included in this study are extracted using the search engine "Web of Science" over eight years. The most common complications of diabetes, diabetic neuropathy, retinopathy, nephropathy, and cardiovascular diseases are considered in the study. METHOD The features used to predict the complications are identified and categorised by scrutinising the standards of electronic health records. RESULT Overall, 102 research articles have been reviewed, resulting in 59 frequent features being identified. Nineteen attributes are recognised as a standard in all four considered complications, which are age, gender, ethnicity, weight, height, BMI, smoking history, HbA1c, SBP, eGFR, DBP, HDL, LDL, total cholesterol, triglyceride, use of insulin, duration of diabetes, family history of CVD, and diabetes. The existence of a well-accepted and updated feature set for health analytics models to predict the complications of diabetes mellitus is a vital and contemporary requirement. A widely accepted feature set is beneficial for benchmarking the risk factors of complications of diabetes. CONCLUSION This study is a thorough literature review to provide a clear state of the art for academicians, clinicians, and other stakeholders regarding the risk factors and their importance.
Collapse
Affiliation(s)
| | | | | | - Ching-Chi Lee
- National Chen Kung University Hospital, Tainan, Taiwan
| |
Collapse
|
7
|
Spallone V. Diabetic neuropathy: Current issues in diagnosis and prevention. CHRONIC COMPLICATIONS OF DIABETES MELLITUS 2024:117-163. [DOI: 10.1016/b978-0-323-88426-6.00016-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
8
|
Li J, Zhang X, Zhang Y, Dan X, Wu X, Yang Y, Chen X, Li S, Xu Y, Wan Q, Yan P. Increased Systemic Immune-Inflammation Index Was Associated with Type 2 Diabetic Peripheral Neuropathy: A Cross-Sectional Study in the Chinese Population. J Inflamm Res 2023; 16:6039-6053. [PMID: 38107379 PMCID: PMC10723178 DOI: 10.2147/jir.s433843] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/08/2023] [Indexed: 12/19/2023] Open
Abstract
Background Systemic immune-inflammation index (SII), a novel inflammatory marker, has been demonstrated to be associated with type 2 diabetes mellitus (T2DM) and its vascular complications, however, the relation between SII and diabetic peripheral neuropathy (DPN) has been never reported. We aimed to explore whether SII is associated with DPN in Chinese population. Methods A cross-sectional study was conducted among 1460 hospitalized patients with T2DM. SII was calculated as the platelet count × neutrophil count/lymphocyte count, and its possible association with DPN was investigated by correlation and multivariate logistic regression analysis, and subgroup analyses. Results Patients with higher SII quartiles had higher vibration perception threshold and prevalence of DPN (all P<0.01), and SII was independently positively associated with the prevalence of DPN (P<0.01). Multivariate logistic regression analysis showed that the risk of prevalence of DPN increased progressively across SII quartiles (P for trend <0.01), and participants in the highest quartile of SII was at a significantly increased risk of prevalent DPN compared to those in the lowest quartile after adjustment for potential confounding factors (odds rate: 1.211, 95% confidence intervals 1.045-1.404, P<0.05). Stratified analysis revealed positive associations of SII quartiles with risk of prevalent DPN only in men, people less than 65 years old, with body mass index <24 kg/m2, duration of diabetes >5 years, hypertension, dyslipidaemia, poor glycaemic control, and estimated glomerular filtration rate <90 mL/min/1.73 m2 (P for trend <0.01 or P for trend <0.05). The receiver operating characteristic curve analysis revealed that the optimal cut-off point of SII for predicting DPN was 617.67 in patients with T2DM, with a sensitivity of 45.3% and a specificity of 73%. Conclusion The present study showed that higher SII is independently associated with increased risk of DPN, and SII might serve as a new risk biomarker for DPN in Chinese population.
Collapse
Affiliation(s)
- Jia Li
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, China, Luzhou, People’s Republic of China
| | - Xing Zhang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, China, Luzhou, People’s Republic of China
| | - Yi Zhang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, China, Luzhou, People’s Republic of China
| | - Xiaofang Dan
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, China, Luzhou, People’s Republic of China
| | - Xian Wu
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, China, Luzhou, People’s Republic of China
| | - Yuxia Yang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, China, Luzhou, People’s Republic of China
| | - Xiping Chen
- Clinical medical college, Southwest Medical University, Luzhou, People’s Republic of China
| | - Shengxi Li
- Basic Medical College, Southwest Medical University, Luzhou, People’s Republic of China
| | - Yong Xu
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, China, Luzhou, People’s Republic of China
| | - Qin Wan
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, China, Luzhou, People’s Republic of China
| | - Pijun Yan
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, China, Luzhou, People’s Republic of China
| |
Collapse
|
9
|
Määttä LL, Andersen ST, Parkner T, Hviid CVB, Bjerg L, Kural MA, Charles M, Søndergaard E, Sandbæk A, Tankisi H, Witte DR, Jensen TS. Serum neurofilament light chain - A potential biomarker for polyneuropathy in type 2 diabetes? Diabetes Res Clin Pract 2023; 205:110988. [PMID: 38349953 DOI: 10.1016/j.diabres.2023.110988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 02/15/2024]
Abstract
AIMS To investigate the relationship between neurofilament light chain (NfL) and the presence and severity of diabetic polyneuropathy (DPN). METHODS We performed cross-sectional analysis of data from 178 participants of the ADDITION-Denmark cohort of people with screen-detected type 2 diabetes and 32 healthy controls. Biobank serum samples were analyzed for NfL using single-molecule array. DPN was defined by Toronto criteria for confirmed DPN. Original and axonal nerve conduction study (NCS) sum z-scores were used as indicators of the severity of DPN and peripheral nerve damage. RESULTS 39 (21.9%) participants had DPN. Serum NfL (s-NfL) was significantly higher in participants with DPN (18.8 ng/L [IQR 14.4; 27.9]) than in participants without DPN (15.4 ng/L [IQR 11.7; 20.1]). There were no unadjusted s-NfL differences between controls (17.6 ng/L [IQR 12.7; 19.8]) and participants with or without DPN. Higher original and axonal NCS sum z-scores were associated with 10% higher s-NfL (10.2 and 12.1% [95% CI's 4.0; 16.8 and 6.6; 17.9] per 1 SD). The AUC of s-NfL for DPN was 0.63 (95% CI 0.52; 0.73). CONCLUSIONS S-NfL is unlikely to be a reliable biomarker for the presence of DPN. S-NfL is however associated tothe severity of the nerve damage underlying DPN.
Collapse
Affiliation(s)
- Laura L Määttä
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard, 165, J109, 8200 Aarhus, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard, 11, 8200 Aarhus, Denmark.
| | - Signe T Andersen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard, 165, J109, 8200 Aarhus, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard, 11, 8200 Aarhus, Denmark.
| | - Tina Parkner
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensens Boulevard, 99, 8200 Aarhus, Denmark Aarhus, Denmark.
| | - Claus V B Hviid
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensens Boulevard, 99, 8200 Aarhus, Denmark Aarhus, Denmark; Department of Clinical Biochemistry, Aalborg University Hospital, Hobrovej 18-22, 9000 Aalborg, Denmark.
| | - Lasse Bjerg
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard, 11, 8200 Aarhus, Denmark; Department of Public Health, Aarhus University, Batholins Allé 2, 8000 Aarhus, Denmark.
| | - Mustafa A Kural
- Department of Clinical Neurophysiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard, 165, J209, 8200 Aarhus, Denmark.
| | - Morten Charles
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard, 11, 8200 Aarhus, Denmark; Department of Public Health, Aarhus University, Batholins Allé 2, 8000 Aarhus, Denmark.
| | - Esben Søndergaard
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard, 11, 8200 Aarhus, Denmark.
| | - Annelli Sandbæk
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard, 11, 8200 Aarhus, Denmark; Department of Public Health, Aarhus University, Batholins Allé 2, 8000 Aarhus, Denmark.
| | - Hatice Tankisi
- Department of Clinical Neurophysiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard, 165, J209, 8200 Aarhus, Denmark.
| | - Daniel R Witte
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard, 11, 8200 Aarhus, Denmark; Department of Public Health, Aarhus University, Batholins Allé 2, 8000 Aarhus, Denmark.
| | - Troels S Jensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard, 165, J109, 8200 Aarhus, Denmark.
| |
Collapse
|
10
|
Chang KC, Pai YW, Lin CH, Lee IT, Chang MH. The association between hyperlipidemia, lipid-lowering drugs and diabetic peripheral neuropathy in patients with type 2 diabetes mellitus. PLoS One 2023; 18:e0287373. [PMID: 37319238 PMCID: PMC10270586 DOI: 10.1371/journal.pone.0287373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 06/04/2023] [Indexed: 06/17/2023] Open
Abstract
AIMS Previous studies showed conflicting relationship between hyperlipidemia, lipid-lowering therapy and diabetic peripheral neuropathy (DPN). As most of these works emerges from the Western and Australian countries, our study aims to investigate whether hyperlipidemia or lipid-lowering therapy (LLT) is associated with DPN in Taiwanese patients with type 2 diabetes (T2D). METHODS A cross-sectional, hospital-based observation study in adults with T2D was conducted from January to October 2013. DPN was screened using the Michigan Neuropathy Screening Instrument. Data were obtained at the time of enrollment, including medication usage, anthropometric measurements and laboratory examinations. RESULTS 2,448 participants were enrolled, 524 (21.4%) of whom had DPN. Patients with DPN had significantly lower plasma total cholesterol (185.6 ± 38.6 vs 193.4 ± 42.3 mg/dL) and low-density lipoprotein cholesterol levels (114.6 ± 32.7 vs 119 ± 30.8 mg/dL). Multivariate analysis demonstrated that neither hyperlipidemia (adjusted OR (aOR), 0.81; 95% confidence interval (CI), 0.49-1.34) nor LLT (aOR, 1.10; 95% CI, 0.58-2.09) was associated with DPN. Subgroup analysis revealed that neither total cholesterol (aOR, 0.72; 95% CI, 0.2-2.62), low-density lipoprotein cholesterol levels (aOR, 0.75; 95% CI, 0.2-2.79), statin (aOR, 1.09; 95% CI, 0.59-2.03) nor fibrate (aOR, 1.73; 95% CI, 0.33-1.61) was associated with DPN. CONCLUSION Our results suggest that neither hyperlipidemia nor lipid-lowering medication was associated with DPN in adults with T2D. DPN is a multifactorial disease, and our findings indicate that lipid metabolism may play a minor role in its pathogenesis.
Collapse
Affiliation(s)
- Kuo-Cheng Chang
- Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yen-Wei Pai
- Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Ching-Heng Lin
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - I-Te Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ming-Hong Chang
- Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
11
|
Wu Y, Wan Q, Xu Y, Li J, Li K, Zhang Z, Tang Q, Miao Y, Yan P. Lower Visceral Fat Area in Patients with Type 2 Diabetic Peripheral Neuropathy. Diabetes Metab Syndr Obes 2022; 15:3639-3654. [PMID: 36439295 PMCID: PMC9694982 DOI: 10.2147/dmso.s388330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/10/2022] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE There is preliminary evidence that visceral fat area (VFA) was associated with the presence of type 2 diabetic peripheral neuropathy (DPN) in the Korean population; however, no studies have reported the association in Chinese population. The purpose of this study was to explore the possible correlation of VFA with DPN in such a population. METHODS A total of 2498 hospitalized patients with type 2 diabetes mellitus (T2DM) undergone VFA measurement, and were divided into DPN group (n=900) and non-DPN group (n=1594). The association of VFA with the presence of DPN was evaluated by correlation and multiple logistic regression analyses, generalized additive model with a smooth curve fitting, and receiver operating characteristic (ROC) curve analysis. RESULTS The VFA was significantly lower in the DPN group than in the non-DPN group (P < 0.001). VFA was significantly and positively associated with sural nerve conduction velocity (SNCV) and amplitude potential (SNAP) and negatively associated with the presence of DPN (all P< 0.001); there was no significant difference in the curve fitting (P = 0.344). Multivariate logistic regression analysis showed that the risk of presence of DPN decreased progressively across the VFA quartiles (P for trend < 0.001) and was significantly lower in patients in the highest VFA quartile than in those in the lowest quartile (OR: 0.382, 95% CI 0.151-0.968, P< 0.001) after multivariate adjustment. The ROC analysis revealed that the best cut-off value of VFA for predicting the presence of DPN was 50.5cm2 (sensitivity 84.40%; specificity 34.00%). CONCLUSION These results suggest that lower VFA level may be associated with increased risk of the presence of DPN in T2DM patients.
Collapse
Affiliation(s)
- Yuru Wu
- Department of Endocrinology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| | - Qin Wan
- Department of Endocrinology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| | - Yong Xu
- Department of Endocrinology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| | - Jia Li
- Department of Endocrinology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| | - Ke Li
- Department of Endocrinology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| | - Zhihong Zhang
- Department of General Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Qian Tang
- Department of Endocrinology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| | - Ying Miao
- Department of Endocrinology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| | - Pijun Yan
- Department of Endocrinology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
- Correspondence: Pijun Yan, Department of Endocrinology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China, Tel/Fax +86-830-3165361, Email
| |
Collapse
|
12
|
Tommerdahl KL, Shapiro ALB, Nehus EJ, Bjornstad P. Early microvascular complications in type 1 and type 2 diabetes: recent developments and updates. Pediatr Nephrol 2022; 37:79-93. [PMID: 33852054 PMCID: PMC8527882 DOI: 10.1007/s00467-021-05050-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/01/2021] [Accepted: 03/09/2021] [Indexed: 12/30/2022]
Abstract
The prevalence of youth-onset diabetes is progressing rapidly worldwide, and poor glycemic control, in combination with prolonged diabetes duration and comorbidities including hypertension, has led to the early development of microvascular complications including diabetic kidney disease, retinopathy, and neuropathy. Pediatric populations with type 1 (T1D) and type 2 (T2D) diabetes are classically underdiagnosed with microvascular complications, and this leads to both undertreatment and insufficient attention to the mitigation of risk factors that could help attenuate further progression of complications and decrease the likelihood for long-term morbidity and mortality. This narrative review aims to present a comprehensive summary of the epidemiology, risk factors, symptoms, screening practices, and treatment options, including future opportunities for treatment advancement, for microvascular complications in youth with T1D and T2D. We seek to uniquely focus on the inherent challenges of managing pediatric populations with diabetes and discuss the similarities and differences between microvascular complications in T1D and T2D, while presenting a strong emphasis on the importance of early identification of at-risk youth. Further investigation of possible treatment mechanisms for microvascular complications in youth with T1D and T2D through dedicated pediatric outcome trials is necessary to target the brief window where early pathological vascular changes may be significantly attenuated.
Collapse
Affiliation(s)
- Kalie L Tommerdahl
- Department of Pediatrics, Section of Pediatric Endocrinology, Children's Hospital Colorado and University of Colorado Anschutz Medical Campus, 13123 E. 16th Avenue, Box B265, Aurora, CO, 80045, USA
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
- Center for Women's Health Research, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Allison L B Shapiro
- Department of Pediatrics, Section of Pediatric Endocrinology, Children's Hospital Colorado and University of Colorado Anschutz Medical Campus, 13123 E. 16th Avenue, Box B265, Aurora, CO, 80045, USA
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Edward J Nehus
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Petter Bjornstad
- Department of Pediatrics, Section of Pediatric Endocrinology, Children's Hospital Colorado and University of Colorado Anschutz Medical Campus, 13123 E. 16th Avenue, Box B265, Aurora, CO, 80045, USA.
- Center for Women's Health Research, University of Colorado School of Medicine, Aurora, CO, USA.
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
13
|
García-Mesa Y, Feito J, González-Gay M, Martínez I, García-Piqueras J, Martín-Cruces J, Viña E, Cobo T, García-Suárez O. Involvement of Cutaneous Sensory Corpuscles in Non-Painful and Painful Diabetic Neuropathy. J Clin Med 2021; 10:jcm10194609. [PMID: 34640627 PMCID: PMC8509589 DOI: 10.3390/jcm10194609] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/02/2021] [Accepted: 10/05/2021] [Indexed: 12/18/2022] Open
Abstract
Distal diabetic sensorimotor polyneuropathy (DDSP) is the most prevalent form of diabetic neuropathy, and some of the patients develop gradual pain. Specialized sensory structures present in the skin encode different modalities of somatosensitivity such as temperature, touch, and pain. The cutaneous sensory structures responsible for the qualities of mechanosensitivity (fine touch, vibration) are collectively known as cutaneous mechanoreceptors (Meissner corpuscles, Pacinian corpuscles, and Merkel cell-axonal complexes), which results are altered during diabetes. Here, we used immunohistochemistry to analyze the density, localization within the dermis, arrangement of corpuscular components (axons and Schwann-like cells), and expression of putative mechanoproteins (PIEZO2, ASIC2, and TRPV4) in cutaneous mechanoreceptors of subjects suffering clinically diagnosed non-painful and painful distal diabetic sensorimotor polyneuropathy. The number of Meissner corpuscles, Pacinian corpuscles, and Merkel cells was found to be severely decreased in the non-painful presentation of the disease, and almost disappeared in the painful presentation. Furthermore, there was a marked reduction in the expression of axonal and Schwann-like cell markers (with are characteristics of corpuscular denervation) as well as of all investigated mechanoproteins in the non-painful distal diabetic sensorimotor polyneuropathy, and these were absent in the painful form. Taken together, these alterations might explain, at least partly, the impairment of mechanosensitivity system associated with distal diabetic sensorimotor polyneuropathy. Furthermore, our results support that an increasing severity of DDSP may increase the risk of developing painful neuropathic symptoms. However, why the absence of cutaneous mechanoreceptors is associated with pain remains to be elucidated.
Collapse
Affiliation(s)
- Yolanda García-Mesa
- Grupo SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Spain; (Y.G.-M.); (J.F.); (J.G.-P.); (J.M.-C.); (E.V.)
| | - Jorge Feito
- Grupo SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Spain; (Y.G.-M.); (J.F.); (J.G.-P.); (J.M.-C.); (E.V.)
- Servicio de Anatomía Patológica, Complejo Asistencial Universitario de Salamanca, 37007 Salamanca, Spain
| | - Mario González-Gay
- Sercivio de Angiología y Cirugía Vascular, Fundación Hospital de Jove, 33290 Gijón, Spain;
| | - Irene Martínez
- Sercivio de Cirugía Plástica y Reparadora, Fundación Hospital de Jove, 33290 Gijón, Spain;
| | - Jorge García-Piqueras
- Grupo SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Spain; (Y.G.-M.); (J.F.); (J.G.-P.); (J.M.-C.); (E.V.)
| | - José Martín-Cruces
- Grupo SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Spain; (Y.G.-M.); (J.F.); (J.G.-P.); (J.M.-C.); (E.V.)
| | - Eliseo Viña
- Grupo SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Spain; (Y.G.-M.); (J.F.); (J.G.-P.); (J.M.-C.); (E.V.)
- Servicio de Cardiología, Unidad de Hemodinámica y Cardiología Intervencionista, Hospital de Cabueñes, 33206 Gijón, Spain
| | - Teresa Cobo
- Departamento de Cirugía y Especialidades Médico-Quirúrgicas, Universidad de Oviedo, 33006 Oviedo, Spain;
| | - Olivia García-Suárez
- Grupo SINPOS, Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Spain; (Y.G.-M.); (J.F.); (J.G.-P.); (J.M.-C.); (E.V.)
- Correspondence:
| |
Collapse
|
14
|
Ziegler D, Bönhof GJ, Strom A, Straßburger K, Karusheva Y, Szendroedi J, Roden M. Progression and regression of nerve fibre pathology and dysfunction early in diabetes over 5 years. Brain 2021; 144:3251-3263. [PMID: 34499110 DOI: 10.1093/brain/awab330] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/24/2021] [Accepted: 08/08/2021] [Indexed: 12/17/2022] Open
Abstract
It has been traditionally suggested that the early development of diabetic sensorimotor polyneuropathy (DSPN) is characterized by a predominant and progressive injury to small nerve fibres followed by large fibre impairment. We alternatively hypothesized that small and large fibre damage due to DSPN in type 1 and type 2 diabetes could develop in parallel and may not only be progressive but also reversible. Participants from the German Diabetes Study baseline cohort with recent-onset type 1/type 2 diabetes (n = 350/570) and age-matched glucose-tolerant control individuals (Control 1/Control 2: n = 114/190) were assessed by nerve conduction studies (NCS), thermal detection thresholds (TDT), vibration perception threshold (VPT), Neuropathy Symptom Score (NSS), Neuropathy Disability Score (NDS), and intraepidermal nerve fibre density (IENFD) in skin biopsies (type 1/type 2 diabetes: n = 102/226; Control 1/Control 2: n = 109/208). Subsets of participants with type 1/type 2 diabetes were followed for 5 years (n = 184/307; IENFD subset: n = 18/69). DSPN was defined by the Toronto Consensus criteria. At baseline, DSPN was present in 8.1 and 13.3% of the type 1 and type 2 diabetes groups, respectively. The most frequently abnormal tests in the lower limbs below or above the 2.5th and 97.5th centile of the controls were IENFD (13.7%) and individual NCS (up to 9.4%) in type 1 diabetes participants and IENFD (21.8%), malleolar VPT (17.5%), and individual NCS (up to 11.8%) in those with type 2 diabetes, whereas TDT abnormalities did not differ between the control and diabetes groups. After 5 years in type 2 diabetes participants, the highest progression rates from the normal to the abnormal range were found for IENFD (18.8%) by -4.1 ± 2.8 fibres/mm, malleolar VPT (18.6%) by 9.1 ± 20.2 µm, and NDS (15.0%) by 3.7 ± 1.5 points, while vice versa the highest regression rates were observed for NDS (11.2%) by -3.1 ± 1.3 points, sural nerve amplitude (9.1%) by 4.7 ± 3.0 µV, IENFD (8.7%) by 1.4 ± 1.3 fibres/mm, and NSS (8.2%) by -5.8 ± 1.6 points. In type 1 diabetes participants, no major progression was seen after 5 years, but subclinical DSPN regressed in 10.3%. These findings point to an early parallel damage to both small and large nerve fibres in well-controlled recent-onset type 2 and, to a lesser extent, type 1 diabetes. After 5 years peripheral nerve morphology and function and clinical measures progress to the abnormal range in type 2 diabetes, but initial nerve alterations are also reversible to a meaningful degree.
Collapse
Affiliation(s)
- Dan Ziegler
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany.,Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany.,German Center for Diabetes Research, Partner Düsseldorf, 85764 München-Neuherberg, Germany
| | - Gidon J Bönhof
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany.,Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| | - Alexander Strom
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany.,German Center for Diabetes Research, Partner Düsseldorf, 85764 München-Neuherberg, Germany
| | - Klaus Straßburger
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Yanislava Karusheva
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| | - Julia Szendroedi
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany.,Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany.,German Center for Diabetes Research, Partner Düsseldorf, 85764 München-Neuherberg, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany.,Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany.,German Center for Diabetes Research, Partner Düsseldorf, 85764 München-Neuherberg, Germany
| | | |
Collapse
|
15
|
Wu B, Niu Z, Hu F. Study on Risk Factors of Peripheral Neuropathy in Type 2 Diabetes Mellitus and Establishment of Prediction Model. Diabetes Metab J 2021; 45:526-538. [PMID: 34352988 PMCID: PMC8369209 DOI: 10.4093/dmj.2020.0100] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/12/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Diabetic peripheral neuropathy (DPN) is one of the most serious complications of type 2 diabetes mellitus (T2DM). DPN increases the risk of ulcers, foot infections, and noninvasive amputations, ultimately leading to long-term disability. METHODS Seven hundred patients with T2DM were investigated from 2013 to 2017 in the Sanlin community by obtaining basic data from the electronic medical record system (EMRS). From September 2018 to July 2019, 681 patients (19 missing) were investigated using a questionnaire, physical examination, biochemical index test, and follow-up Toronto clinical scoring system (TCSS) test. Patients with a TCSS score ≥6 points were diagnosed with DPN. After removing missing values, 612 patients were divided into groups in a 3:1 ratio for external validation. Using different Lasso analyses (misclassification error, mean squared error, -2log-likelihood, and area under curve) and a logistic regression analysis of the training set, models A, B, C, and D were established. The receiver operating characteristic (ROC) curve, calibration plot, dynamic component analysis (DCA) measurements, net classification improvement (NRI) and integrated discrimination improvement (IDI) were used to validate discrimination and clinical practicality of the model. RESULTS Through data analysis, model A (containing four factors), model B (containing five factors), model C (containing seven factors), and model D (containing seven factors) were built. After calibration, ROC curve, DCA, NRI and IDI, models C and D exhibited better accuracy and greater predictive power. CONCLUSION Four prediction models were established to assist with the early screening of DPN in patients with T2DM. The influencing factors in model C and D are more important factors for patients with T2DM diagnosed with DPN.
Collapse
Affiliation(s)
- Birong Wu
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zheyun Niu
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fan Hu
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Corresponding author: Fan Hu https://orcid.org/0000-0002-7929-6953 School of Public Health, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China E-mail:
| |
Collapse
|
16
|
Rumora AE, Guo K, Alakwaa FM, Andersen ST, Reynolds EL, Jørgensen ME, Witte DR, Tankisi H, Charles M, Savelieff MG, Callaghan BC, Jensen TS, Feldman EL. Plasma lipid metabolites associate with diabetic polyneuropathy in a cohort with type 2 diabetes. Ann Clin Transl Neurol 2021; 8:1292-1307. [PMID: 33955722 PMCID: PMC8164865 DOI: 10.1002/acn3.51367] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE The global rise in type 2 diabetes is associated with a concomitant increase in diabetic complications. Diabetic polyneuropathy is the most frequent type 2 diabetes complication and is associated with poor outcomes. The metabolic syndrome has emerged as a major risk factor for diabetic polyneuropathy; however, the metabolites associated with the metabolic syndrome that correlate with diabetic polyneuropathy are unknown. METHODS We conducted a global metabolomics analysis on plasma samples from a subcohort of participants from the Danish arm of Anglo-Danish-Dutch study of Intensive Treatment of Diabetes in Primary Care (ADDITION-Denmark) with and without diabetic polyneuropathy versus lean control participants. RESULTS Compared to lean controls, type 2 diabetes participants had significantly higher HbA1c (p = 0.0028), BMI (p = 0.0004), and waist circumference (p = 0.0001), but lower total cholesterol (p = 0.0001). Out of 991 total metabolites, we identified 15 plasma metabolites that differed in type 2 diabetes participants by diabetic polyneuropathy status, including metabolites belonging to energy, lipid, and xenobiotic pathways, among others. Additionally, these metabolites correlated with alterations in plasma lipid metabolites in type 2 diabetes participants based on neuropathy status. Further evaluating all plasma lipid metabolites identified a shift in abundance, chain length, and saturation of free fatty acids in type 2 diabetes participants. Importantly, the presence of diabetic polyneuropathy impacted the abundance of plasma complex lipids, including acylcarnitines and sphingolipids. INTERPRETATION Our explorative study suggests that diabetic polyneuropathy in type 2 diabetes is associated with novel alterations in plasma metabolites related to lipid metabolism.
Collapse
Affiliation(s)
- Amy E. Rumora
- Department of NeurologyUniversity of MichiganAnn ArborMichigan
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichigan
| | - Kai Guo
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichigan
- Department of Biomedical SciencesUniversity of North DakotaGrand ForksNorth Dakota
| | - Fadhl M. Alakwaa
- Department of NeurologyUniversity of MichiganAnn ArborMichigan
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichigan
| | | | - Evan L. Reynolds
- Department of NeurologyUniversity of MichiganAnn ArborMichigan
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichigan
| | - Marit E. Jørgensen
- Steno Diabetes Center CopenhagenGentofteDenmark
- University of Southern DenmarkOdenseDenmark
| | - Daniel R. Witte
- Department of Public HealthAarhus UniversityAarhusDenmark
- Danish Diabetes AcademyOdenseDenmark
| | - Hatice Tankisi
- Department of Clinical NeurophysiologyAarhus UniversityAarhusDenmark
| | - Morten Charles
- Department of Public HealthAarhus UniversityAarhusDenmark
| | - Masha G. Savelieff
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichigan
| | - Brian C. Callaghan
- Department of NeurologyUniversity of MichiganAnn ArborMichigan
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichigan
| | - Troels S. Jensen
- Danish Pain Research CenterDepartment of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Eva L. Feldman
- Department of NeurologyUniversity of MichiganAnn ArborMichigan
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichigan
| |
Collapse
|
17
|
Maalmi H, Wouters K, Savelberg HHCM, van der Velde JHPM, Reulen JPH, Mess W, Schalkwijk CG, Stehouwer CDA, Roden M, Ziegler D, Herder C, Schaper NC. Associations of cells from both innate and adaptive immunity with lower nerve conduction velocity: the Maastricht Study. BMJ Open Diabetes Res Care 2021; 9:9/1/e001698. [PMID: 33431599 PMCID: PMC7802711 DOI: 10.1136/bmjdrc-2020-001698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 11/24/2020] [Accepted: 11/29/2020] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Distal sensorimotor polyneuropathy (DSPN) is common in people with diabetes but is also found in pre-diabetes. Peripheral nerve myelin damage, which can be assessed by reduced nerve conduction velocity (NCV), is an essential feature of DSPN. Emerging evidence indicates that the development of DSPN may involve the activation of the immune system. However, available studies have mainly investigated circulating immune mediators, whereas the role of immune cells remains unclear. Therefore, we aimed to test whether leukocyte subsets are associated with NCV. RESEARCH DESIGN AND METHODS This cross-sectional study analyzed data from 850 individuals (of whom 252 and 118 had type 2 diabetes and pre-diabetes, respectively) of the Maastricht Study. NCV was measured in the peroneal and tibial motor nerves and the sural sensory nerve and summed to calculate a standardized NCV sum score. Associations between percentages of leukocyte subsets and NCV sum scores were estimated using linear regression models adjusted for demographic, lifestyle, metabolic and clinical covariates. RESULTS After adjustment for covariates, higher percentages of basophils and CD4+ T cells were associated with lower NCV (p=0.014 and p=0.005, respectively). The percentage of CD8+ T cells was positively associated with NCV (p=0.022). These associations were not modified by glucose metabolism status (all pinteraction >0.05). No associations were found for monocytes, eosinophils, neutrophils, lymphocytes, total T cells, Treg cells and B cells. CONCLUSIONS The associations of basophils, CD4+ and CD8+ T cells with NCV suggest that cell types from both innate and adaptive immunity may be implicated in the development of DSPN.
Collapse
Affiliation(s)
- Haifa Maalmi
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Kristiaan Wouters
- Department of Internal Medicine, Maastricht University Medical Centre+ (MUMC+), Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - Hans H C M Savelberg
- Department of Nutrition and Movement Sciences, Maastricht University, Maastricht, The Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Jeroen H P M van der Velde
- Department of Nutrition and Movement Sciences, Maastricht University, Maastricht, The Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Jos P H Reulen
- Department of Clinical Neurophysiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Werner Mess
- Department of Clinical Neurophysiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, Maastricht University Medical Centre+ (MUMC+), Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - Coen D A Stehouwer
- Department of Internal Medicine, Maastricht University Medical Centre+ (MUMC+), Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Dan Ziegler
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Nicolaas C Schaper
- Department of Internal Medicine, Maastricht University Medical Centre+ (MUMC+), Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| |
Collapse
|
18
|
Herder C, Schneider A, Zhang S, Wolf K, Maalmi H, Huth C, Pickford R, Laxy M, Bönhof GJ, Koenig W, Rathmann W, Roden M, Peters A, Thorand B, Ziegler D. Association of Long-Term Air Pollution with Prevalence and Incidence of Distal Sensorimotor Polyneuropathy: KORA F4/FF4 Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2020; 128:127013. [PMID: 33356516 PMCID: PMC7757787 DOI: 10.1289/ehp7311] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 11/20/2020] [Accepted: 11/20/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Air pollution contributes to type 2 diabetes and cardiovascular diseases, but its relevance for other complications of diabetes, in particular distal sensorimotor polyneuropathy (DSPN), is unclear. Recent studies have indicated that DSPN is also increasingly prevalent in obesity. OBJECTIVES We aimed to assess associations of air pollutants with prevalent and incident DSPN in a population-based study of older individuals with high rates of type 2 diabetes and obesity. METHODS Cross-sectional analyses on prevalent DSPN were based on 1,075 individuals 62-81 years of age from the German Cooperative Health Research in the Region of Augsburg (KORA) F4 survey (2006-2008). Analyses on incident DSPN included 424 individuals without DSPN at baseline (KORA F4), of whom 188 had developed DSPN by the KORA FF4 survey (2013-2014). Associations of annual average air pollutant concentrations at participants' residences with prevalent and incident DSPN were estimated using Poisson regression models with a robust error variance adjusting for multiple confounders. RESULTS Higher particle number concentrations (PNCs) were associated with higher prevalence [risk ratio (RR) per interquartile range (IQR) increase=1.10 (95% CI: 1.01, 1.20)] and incidence [1.11 (95% CI: 0.99, 1.24)] of DSPN. In subgroup analyses, particulate (PNC, PM10, PMcoarse, PM2.5, and PM2.5abs) and gaseous (NOx, NO2) pollutants were positively associated with prevalent DSPN in obese participants, whereas corresponding estimates for nonobese participants were close to the null [e.g., for an IQR increase in PNC, RR=1.17 (95% CI: 1.05, 1.31) vs. 1.06 (95% CI: 0.95, 1.19); pinteraction=0.22]. With the exception of PM2.5abs, corresponding associations with incident DSPN were positive in obese participants but null or inverse for nonobese participants, with pinteraction≤0.13 [e.g., for PNC, RR=1.28 (95% CI: 1.08, 1.51) vs. 1.03 (95% CI: 0.90, 1.18); pinteraction=0.03]. DISCUSSION Both particulate and gaseous air pollutants were positively associated with prevalent and incident DSPN in obese individuals. Obesity and air pollution may have synergistic effects on the development of DSPN. https://doi.org/10.1289/EHP7311.
Collapse
Affiliation(s)
- Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Munich-Neuherberg, Germany
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Alexandra Schneider
- German Center for Diabetes Research, Munich-Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Siqi Zhang
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Kathrin Wolf
- German Center for Diabetes Research, Munich-Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Haifa Maalmi
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Munich-Neuherberg, Germany
| | - Cornelia Huth
- German Center for Diabetes Research, Munich-Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Regina Pickford
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Michael Laxy
- German Center for Diabetes Research, Munich-Neuherberg, Germany
- Institute of Health Economics and Health Care Management, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Global Diabetes Research Center, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
- Department of Sport and Health Science, Technical University of Munich, Munich, Germany
| | - Gidon J. Bönhof
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Munich-Neuherberg, Germany
| | - Wolfgang Koenig
- German Heart Center Munich, Technical University of Munich, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
| | - Wolfgang Rathmann
- German Center for Diabetes Research, Munich-Neuherberg, Germany
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Munich-Neuherberg, Germany
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Annette Peters
- German Center for Diabetes Research, Munich-Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Barbara Thorand
- German Center for Diabetes Research, Munich-Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Dan Ziegler
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Munich-Neuherberg, Germany
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
19
|
Christensen DH, Knudsen ST, Gylfadottir SS, Christensen LB, Nielsen JS, Beck-Nielsen H, Sørensen HT, Andersen H, Callaghan BC, Feldman EL, Finnerup NB, Jensen TS, Thomsen RW. Metabolic Factors, Lifestyle Habits, and Possible Polyneuropathy in Early Type 2 Diabetes: A Nationwide Study of 5,249 Patients in the Danish Centre for Strategic Research in Type 2 Diabetes (DD2) Cohort. Diabetes Care 2020; 43:1266-1275. [PMID: 32295810 DOI: 10.2337/dc19-2277] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 03/17/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To investigate the association of metabolic and lifestyle factors with possible diabetic polyneuropathy (DPN) and neuropathic pain in patients with early type 2 diabetes. RESEARCH DESIGN AND METHODS We thoroughly characterized 6,726 patients with recently diagnosed diabetes. After a median of 2.8 years, we sent a detailed questionnaire on neuropathy, including the Michigan Neuropathy Screening Instrument questionnaire (MNSIq), to identify possible DPN (score ≥4) and the Douleur Neuropathique en 4 Questions (DN4) questionnaire for possible associated neuropathic pain (MNSIq ≥4 + pain in both feet + DN4 score ≥3). RESULTS Among 5,249 patients with data on both DPN and pain, 17.9% (n = 938) had possible DPN, including 7.4% (n = 386) with possible neuropathic pain. In regression analyses, central obesity (waist circumference, waist-to-hip ratio, and waist-to-height ratio) was markedly associated with DPN. Other important metabolic factors associated with DPN included hypertriglyceridemia ≥1.7 mmol/L, adjusted prevalence ratio (aPR) 1.36 (95% CI 1.17; 1.59); decreased HDL cholesterol <1.0/1.2 mmol/L (male/female), aPR 1.35 (95% CI 1.12; 1.62); hs-CRP ≥3.0 mg/L, aPR 1.66 (95% CI 1.42; 1.94); C-peptide ≥1,550 pmol/L, aPR 1.72 (95% CI 1.43; 2.07); HbA1c ≥78 mmol/mol, aPR 1.42 (95% CI 1.06; 1.88); and antihypertensive drug use, aPR 1.34 (95% CI 1.16; 1.55). Smoking, aPR 1.50 (95% CI 1.24; 1.81), and lack of physical activity (0 vs. ≥3 days/week), aPR 1.61 (95% CI 1.39; 1.85), were also associated with DPN. Smoking, high alcohol intake, and failure to increase activity after diabetes diagnosis associated with neuropathic pain. CONCLUSIONS Possible DPN was associated with metabolic syndrome factors, insulin resistance, inflammation, and modifiable lifestyle habits in early type 2 diabetes.
Collapse
Affiliation(s)
- Diana H Christensen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark .,International Diabetic Neuropathy Consortium (IDNC), Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Søren T Knudsen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Sandra S Gylfadottir
- International Diabetic Neuropathy Consortium (IDNC), Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Lotte B Christensen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Jens S Nielsen
- Danish Center for Strategic Research in Type 2 Diabetes, Steno Diabetes Center Odense, Odense, Denmark
| | - Henning Beck-Nielsen
- Danish Center for Strategic Research in Type 2 Diabetes, Steno Diabetes Center Odense, Odense, Denmark
| | - Henrik T Sørensen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Henning Andersen
- International Diabetic Neuropathy Consortium (IDNC), Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Brian C Callaghan
- International Diabetic Neuropathy Consortium (IDNC), Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Eva L Feldman
- International Diabetic Neuropathy Consortium (IDNC), Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Nanna B Finnerup
- International Diabetic Neuropathy Consortium (IDNC), Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Troels S Jensen
- International Diabetic Neuropathy Consortium (IDNC), Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Reimar W Thomsen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
20
|
Zaccardi F, Davies MJ, Khunti K. The present and future scope of real-world evidence research in diabetes: What questions can and cannot be answered and what might be possible in the future? Diabetes Obes Metab 2020; 22 Suppl 3:21-34. [PMID: 32250528 DOI: 10.1111/dom.13929] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/18/2019] [Accepted: 11/18/2019] [Indexed: 12/16/2022]
Abstract
The last decade has witnessed an exponential growth in the opportunities to collect and link health-related data from multiple resources, including primary care, administrative, and device data. The availability of these "real-world," "big data" has fuelled also an intense methodological research into methods to handle them and extract actionable information. In medicine, the evidence generated from "real-world data" (RWD), which are not purposely collected to answer biomedical questions, is commonly termed "real-world evidence" (RWE). In this review, we focus on RWD and RWE in the area of diabetes research, highlighting their contributions in the last decade; and give some suggestions for future RWE diabetes research, by applying well-established and less-known tools to direct RWE diabetes research towards better personalized approaches to diabetes care. We underline the essential aspects to consider when using RWD and the key features limiting the translational potential of RWD in generating high-quality and applicable RWE. Only if viewed in the context of other study designs and statistical methods, with its pros and cons carefully considered, RWE will exploit its full potential as a complementary or even, in some cases, substitutive source of evidence compared to the expensive evidence obtained from randomized controlled trials.
Collapse
Affiliation(s)
- Francesco Zaccardi
- Diabetes Research Centre, Leicester Diabetes Centre, Leicester, UK
- Leicester Real World Evidence Unit, Leicester Diabetes Centre, Leicester, UK
| | - Melanie J Davies
- Diabetes Research Centre, Leicester Diabetes Centre, Leicester, UK
| | - Kamlesh Khunti
- Diabetes Research Centre, Leicester Diabetes Centre, Leicester, UK
- Leicester Real World Evidence Unit, Leicester Diabetes Centre, Leicester, UK
| |
Collapse
|
21
|
Määttä LL, Charles M, Witte DR, Bjerg L, Jørgensen ME, Jensen TS, Andersen ST. Prospective Study of Neuropathic Symptoms Preceding Clinically Diagnosed Diabetic Polyneuropathy: ADDITION-Denmark. Diabetes Care 2019; 42:2282-2289. [PMID: 31558545 DOI: 10.2337/dc19-0869] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/09/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To evaluate whether diabetic polyneuropathy (DPN) follows the hypothesis for the course of nerve fiber damage reflected by symptoms progressing from pure small through mixed to large nerve fiber symptoms with or without symptoms of loss of function of small nerve fibers. RESEARCH DESIGN AND METHODS Repeated assessments of nerve fiber-specific symptoms were obtained in 518 participants of the ADDITION-Denmark study from the time of a screening-based diagnosis of type 2 diabetes using specific items of the Michigan Neuropathy Screening Instrument questionnaire. DPN was clinically assessed 13 years after inclusion. The course of symptoms reflecting dysfunction of specific nerve fibers was evaluated, and the association between symptoms and DPN was estimated using logistic regression models. RESULTS An overall stable, yet heterogeneous course of symptoms was seen. According to the hypothesis of symptom progression, 205 (40%) participants remained free of symptoms and 56 (11%) had stable, 114 (23%) progressing, and 132 (26%) improving symptoms. Cross-sectional estimates showed a higher risk of DPN (odds ratios between 2.1 and 4.1) for participants with mixed or large nerve fiber symptoms with or without symptoms of loss of function of small nerve fibers compared with participants without symptoms. CONCLUSIONS There was no evidence for a progressive development of nerve fiber damage in DPN reflected by symptoms going from pure small through mixed to large nerve fiber symptoms with or without symptoms of loss of function of small nerve fibers. Yet overall, neuropathic symptoms were prospectively associated with a higher risk of DPN.
Collapse
Affiliation(s)
- Laura L Määttä
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Morten Charles
- Research Unit of General Practice, Aarhus University, Aarhus, Denmark.,Steno Diabetes Center Aarhus, Aarhus, Denmark
| | - Daniel R Witte
- Department of Public Health, Aarhus University, Aarhus, Denmark.,Danish Diabetes Academy, Odense, Denmark
| | - Lasse Bjerg
- Department of Public Health, Aarhus University, Aarhus, Denmark.,Danish Diabetes Academy, Odense, Denmark.,Clinical Epidemiology, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Marit E Jørgensen
- Clinical Epidemiology, Steno Diabetes Center Copenhagen, Gentofte, Denmark.,National Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Troels S Jensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Signe T Andersen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark .,Department of Public Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
22
|
Kurisu S, Sasaki H, Kishimoto S, Hirayasu K, Ogawa K, Matsuno S, Furuta H, Arita M, Naka K, Nanjo K, Akamizu T. Clinical polyneuropathy does not increase with prediabetes or metabolic syndrome in the Japanese general population. J Diabetes Investig 2019; 10:1565-1575. [PMID: 30980464 PMCID: PMC6825938 DOI: 10.1111/jdi.13058] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/25/2019] [Accepted: 04/09/2019] [Indexed: 12/17/2022] Open
Abstract
AIMS/INTRODUCTION The prevalence of clinical polyneuropathies (ClinPNs) or nerve conduction abnormality (NCA) in the groups stratified by glucose tolerance, individual components of metabolic syndrome (metabolic syndrome [MetS] components: hypertension, dyslipidemia, obesity) and MetS defined by the International Diabetes Federation consensus was investigated in the Japanese general population. Factors associated with ClinPN and NCA were also identified. MATERIALS AND METHODS A total of 625 examinees of regional medical checkup programs were recruited to this cross-sectional study. ClinPNs were diagnosed by the Toronto Consensus. NCA was judged by at least one bilateral abnormality of sural nerve action potential amplitude or conduction velocity measured by a point-of-care nerve conduction device (DPNCheck). Clinical factors associated with ClinPNs or NCA were examined by multiple logistic regression analysis. Deteriorating factors of sural nerve action potential amplitude or conduction velocity values were also investigated in participants without diabetes (n = 550). RESULTS As for glucose tolerance, ClinPNs or NCA significantly increased only in known diabetes patients compared with other groups. There was no difference between prediabetes and the normal group. The prevalence of ClinPNs and NCA was not significantly related to MetS or MetS' components, except for frequent NCA in obesity. The factors significantly associated with both NCA and ClinPNs were smoking and known diabetes. In non-diabetic participants, aging, tall height and hypertension were significant deteriorating factors of nerve conduction functions. CONCLUSIONS In Japan, ClinPNs and NCA were increased in known diabetes patients, but did not increase in participants with prediabetes, MetS and MetS' components. Smoking and known diabetes were factors significantly associated with ClinPNs or NCA. Hypertension might be a modifiable deteriorating factor of nerve function.
Collapse
Affiliation(s)
- Seigo Kurisu
- Department of MedicineKihoku HospitalWakayama Medical UniversityWakayamaJapan
- First Department of MedicineWakayama Medical UniversityWakayamaJapan
| | - Hideyuki Sasaki
- Division of Diabetes and MetabolismSatellite Clinic for Integrative and Anti‐Aging MedicineWakayama Medical UniversityWakayamaJapan
| | - Shohei Kishimoto
- First Department of MedicineWakayama Medical UniversityWakayamaJapan
- Wakayama Rosai HospitalWakayamaJapan
| | - Kazuhiro Hirayasu
- Department of MedicineKihoku HospitalWakayama Medical UniversityWakayamaJapan
| | | | - Shohei Matsuno
- First Department of MedicineWakayama Medical UniversityWakayamaJapan
| | - Hiroto Furuta
- First Department of MedicineWakayama Medical UniversityWakayamaJapan
| | - Mikio Arita
- Health‐promotion Research CenterWakayamaJapan
| | | | | | - Takashi Akamizu
- First Department of MedicineWakayama Medical UniversityWakayamaJapan
| |
Collapse
|
23
|
Herder C, Roden M, Ziegler D. Novel Insights into Sensorimotor and Cardiovascular Autonomic Neuropathy from Recent-Onset Diabetes and Population-Based Cohorts. Trends Endocrinol Metab 2019; 30:286-298. [PMID: 30935671 DOI: 10.1016/j.tem.2019.02.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/22/2019] [Accepted: 02/28/2019] [Indexed: 12/19/2022]
Abstract
The most prevalent chronic complications of diabetes are diabetic neuropathies, among which distal sensorimotor polyneuropathy (DSPN) and cardiovascular autonomic neuropathy (CAN) are the best studied. Their major clinical sequelae such as foot ulcers, neuropathic pain, and orthostatic hypotension are associated with lower quality of life and increased risk of mortality. Here we discuss the recent insights into DSPN and CAN focusing on two prospective cohorts; that is, the German Diabetes Study (GDS) including recent-onset diabetes patients and the population-based Cooperative Health Research in the Region of Augsburg, Germany (KORA) surveys. The insights from these studies investigating novel tools for early detection and prediction of (pre)diabetic neuropathy as well as biomarkers of oxidative stress and inflammation should ultimately culminate in improving the health care of patients affected by this serious condition.
Collapse
Affiliation(s)
- Christian Herder
- Institute of Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany; Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; These authors contributed equally.
| | - Michael Roden
- Institute of Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany; Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Dan Ziegler
- Institute of Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany; Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; These authors contributed equally.
| |
Collapse
|
24
|
Schlesinger S, Herder C, Kannenberg JM, Huth C, Carstensen-Kirberg M, Rathmann W, Bönhof GJ, Koenig W, Heier M, Peters A, Meisinger C, Roden M, Thorand B, Ziegler D. General and Abdominal Obesity and Incident Distal Sensorimotor Polyneuropathy: Insights Into Inflammatory Biomarkers as Potential Mediators in the KORA F4/FF4 Cohort. Diabetes Care 2019; 42:240-247. [PMID: 30523031 DOI: 10.2337/dc18-1842] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/04/2018] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To investigate the associations between different anthropometric measurements and development of distal sensorimotor polyneuropathy (DSPN) considering interaction effects with prediabetes/diabetes and to evaluate subclinical inflammation as a potential mediator. RESEARCH DESIGN AND METHODS This study was conducted among 513 participants from the Cooperative Health Research in the Region of Augsburg (KORA) F4/FF4 cohort (aged 62-81 years). Anthropometry was measured at baseline. Incident DSPN was defined by neuropathic impairments using the Michigan Neuropathy Screening Instrument at baseline and follow-up. Associations between anthropometric measurements and DSPN were estimated by multivariable logistic regression. Potential differences by diabetes status were assessed using interaction terms. Mediation analysis was conducted to determine the mediation effect of subclinical inflammation in these associations. RESULTS After a mean follow-up of 6.5 years, 127 cases with incident DSPN were detected. Both general and abdominal obesity were associated with development of DSPN. The odds ratios (95% CI) of DSPN were 3.06 (1.57; 5.97) for overweight, 3.47 (1.72; 7.00) for obesity (reference: normal BMI), and 1.22 (1.07; 1.38) for 5-cm differences in waist circumference, respectively. Interaction analyses did not indicate any differences by diabetes status. Two chemokines (C-C motif chemokine ligand 7 [CCL7] and C-X-C motif chemokine ligand 10 [CXCL10]) and one neuron-specific marker (Delta/Notch-like epidermal growth factor-related receptor [DNER]) were identified as potential mediators, which explained a proportion of the total effect up to 11% per biomarker. CONCLUSIONS General and abdominal obesity were associated with incident DSPN among individuals with and without diabetes, and this association was partly mediated by inflammatory markers. However, further mechanisms and biomarkers should be investigated as additional mediators to explain the remainder of this association.
Collapse
Affiliation(s)
- Sabrina Schlesinger
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany .,German Center for Diabetes Research, München-Neuherberg, Germany
| | - Christian Herder
- German Center for Diabetes Research, München-Neuherberg, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Julia M Kannenberg
- German Center for Diabetes Research, München-Neuherberg, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Cornelia Huth
- German Center for Diabetes Research, München-Neuherberg, Germany.,Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Maren Carstensen-Kirberg
- German Center for Diabetes Research, München-Neuherberg, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Wolfgang Rathmann
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research, München-Neuherberg, Germany.,Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Gidon J Bönhof
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany.,German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany.,Department of Internal Medicine II-Cardiology, University of Ulm Medical Center, Ulm, Germany
| | - Margit Heier
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Annette Peters
- German Center for Diabetes Research, München-Neuherberg, Germany.,Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Christa Meisinger
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.,Chair of Epidemiology, Ludwig-Maximilians-Universität München am UNIKA-T Augsburg, Augsburg, Germany
| | - Michael Roden
- German Center for Diabetes Research, München-Neuherberg, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Barbara Thorand
- German Center for Diabetes Research, München-Neuherberg, Germany.,Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Dan Ziegler
- German Center for Diabetes Research, München-Neuherberg, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
25
|
Bönhof GJ, Herder C, Strom A, Papanas N, Roden M, Ziegler D. Emerging Biomarkers, Tools, and Treatments for Diabetic Polyneuropathy. Endocr Rev 2019; 40:153-192. [PMID: 30256929 DOI: 10.1210/er.2018-00107] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 08/23/2018] [Indexed: 12/12/2022]
Abstract
Diabetic neuropathy, with its major clinical sequels, notably neuropathic pain, foot ulcers, and autonomic dysfunction, is associated with substantial morbidity, increased risk of mortality, and reduced quality of life. Despite its major clinical impact, diabetic neuropathy remains underdiagnosed and undertreated. Moreover, the evidence supporting a benefit for causal treatment is weak at least in patients with type 2 diabetes, and current pharmacotherapy is largely limited to symptomatic treatment options. Thus, a better understanding of the underlying pathophysiology is mandatory for translation into new diagnostic and treatment approaches. Improved knowledge about pathogenic pathways implicated in the development of diabetic neuropathy could lead to novel diagnostic techniques that have the potential of improving the early detection of neuropathy in diabetes and prediabetes to eventually embark on new treatment strategies. In this review, we first provide an overview on the current clinical aspects and illustrate the pathogenetic concepts of (pre)diabetic neuropathy. We then describe the biomarkers emerging from these concepts and novel diagnostic tools and appraise their utility in the early detection and prediction of predominantly distal sensorimotor polyneuropathy. Finally, we discuss the evidence for and limitations of the current and novel therapy options with particular emphasis on lifestyle modification and pathogenesis-derived treatment approaches. Altogether, recent years have brought forth a multitude of emerging biomarkers reflecting different pathogenic pathways such as oxidative stress and inflammation and diagnostic tools for an early detection and prediction of (pre)diabetic neuropathy. Ultimately, these insights should culminate in improving our therapeutic armamentarium against this common and debilitating or even life-threatening condition.
Collapse
Affiliation(s)
- Gidon J Bönhof
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research, Munich-Neuherberg, Neuherberg, Partner Düsseldorf, Düsseldorf, Germany.,Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Alexander Strom
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research, Munich-Neuherberg, Neuherberg, Partner Düsseldorf, Düsseldorf, Germany
| | - Nikolaos Papanas
- Second Department of Internal Medicine, Diabetes Center, Diabetic Foot Clinic, Democritus University of Thrace, Alexandroupolis, Greece
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research, Munich-Neuherberg, Neuherberg, Partner Düsseldorf, Düsseldorf, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Dan Ziegler
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research, Munich-Neuherberg, Neuherberg, Partner Düsseldorf, Düsseldorf, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
26
|
Kural MA, Andersen ST, Andersen NT, Andersen H, Charles M, Finnerup NB, Jensen TS, Tankisi H. The utility of a point-of-care sural nerve conduction device for detection of diabetic polyneuropathy: A cross-sectional study. Muscle Nerve 2018; 59:187-193. [PMID: 30582180 DOI: 10.1002/mus.26361] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 10/11/2018] [Accepted: 10/14/2018] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Rapid and accessible methods for diagnosing diabetic polyneuropathy (DPN) have been developed, but not validated, in large cohorts of people with diabetes. METHODS The performance of a point-of-care device (POCD) was studied in 168 patients with type 2 diabetes, estimating the sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) compared with conventional sural nerve conduction studies (NCS). RESULTS A POCD amplitude limit of 6 µV increased the sensitivity (96%) and NPV (98%), but decreased the specificity (71%) and PPV (54%) compared with the 4-µV limit, which had values of 78%, 92%, 89%, and 71%, respectively. POCD on both legs showed better performance than on 1 leg. POCD amplitudes and conduction velocities correlated significantly with conventional sural NCS, but POCD values were underestimated compared with NCS. DISCUSSION The POCD may be used as a suitable screening tool for detection of DPN. Patients with abnormal and borderline results should undergo conventional NCS. Muscle Nerve 59:187-193, 2019.
Collapse
Affiliation(s)
- Mustafa A Kural
- Department of Clinical Neurophysiology, Aarhus University Hospital, Nørrebrogade 44, DK-8000, Aarhus C, Denmark
| | | | - Niels T Andersen
- Department of Public Health-Section for Biostatistics, Aarhus University, Aarhus, Denmark
| | - Henning Andersen
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Morten Charles
- Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Nanna B Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Troels S Jensen
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark.,Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Hatice Tankisi
- Department of Clinical Neurophysiology, Aarhus University Hospital, Nørrebrogade 44, DK-8000, Aarhus C, Denmark
| |
Collapse
|
27
|
Rigalleau V, Lecocq M, Rigo M, Monlun M, Poupon P, Blanco L, Alexandre L, Haissaguerre M, Mohammedi K. Comment on Andersen et al. Risk-Factor Trajectories Preceding Diabetic Polyneuropathy: ADDITION-Denmark. Diabetes Care 2018;41:1955-1962. Diabetes Care 2018; 41:e147. [PMID: 30348848 DOI: 10.2337/dc18-1543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Vincent Rigalleau
- Department of Nutrition-Diabetology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Maxime Lecocq
- Department of Nutrition-Diabetology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Marine Rigo
- Department of Nutrition-Diabetology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Marie Monlun
- Department of Nutrition-Diabetology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Pauline Poupon
- Department of Nutrition-Diabetology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Laurence Blanco
- Department of Nutrition-Diabetology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Laure Alexandre
- Department of Nutrition-Diabetology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Magali Haissaguerre
- Department of Nutrition-Diabetology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Kamel Mohammedi
- Department of Nutrition-Diabetology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| |
Collapse
|
28
|
Andersen ST, Witte DR, Andersen H, Bjerg L, Bruun NH, Jørgensen ME, Finnerup NB, Lauritzen T, Jensen TS, Tankisi H, Charles M. Response to Comment on Andersen et al. Risk-Factor Trajectories Preceding Diabetic Polyneuropathy: ADDITION-Denmark. Diabetes Care 2018;41:1955-1962. Diabetes Care 2018; 41:e148-e149. [PMID: 30348849 DOI: 10.2337/dci18-0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Signe T Andersen
- Section for General Medical Practice, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Daniel R Witte
- Section for Epidemiology, Department of Public Health, Aarhus University, Aarhus, Denmark.,Danish Diabetes Academy, Odense, Denmark
| | - Henning Andersen
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Lasse Bjerg
- Section for General Medical Practice, Department of Public Health, Aarhus University, Aarhus, Denmark.,Danish Diabetes Academy, Odense, Denmark.,Clinical Epidemiology, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Niels Henrik Bruun
- Section for General Medical Practice, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Marit E Jørgensen
- Clinical Epidemiology, Steno Diabetes Center Copenhagen, Gentofte, Denmark.,National Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Nanna B Finnerup
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark.,Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Torsten Lauritzen
- Section for General Medical Practice, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Troels S Jensen
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark.,Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Hatice Tankisi
- Department of Clinical Neurophysiology, Aarhus University, Aarhus, Denmark
| | - Morten Charles
- Section for General Medical Practice, Department of Public Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|