1
|
Yang Y, Zhao B, Wang Y, Lan H, Liu X, Hu Y, Cao P. Diabetic neuropathy: cutting-edge research and future directions. Signal Transduct Target Ther 2025; 10:132. [PMID: 40274830 PMCID: PMC12022100 DOI: 10.1038/s41392-025-02175-1] [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] [Received: 08/19/2024] [Revised: 12/12/2024] [Accepted: 02/08/2025] [Indexed: 04/26/2025] Open
Abstract
Diabetic neuropathy (DN) is a prevalent and debilitating complication of diabetes mellitus, significantly impacting patient quality of life and contributing to morbidity and mortality. Affecting approximately 50% of patients with diabetes, DN is predominantly characterized by distal symmetric polyneuropathy, leading to sensory loss, pain, and motor dysfunction, often resulting in diabetic foot ulcers and lower-limb amputations. The pathogenesis of DN is multifaceted, involving hyperglycemia, dyslipidemia, oxidative stress, mitochondrial dysfunction, and inflammation, which collectively damage peripheral nerves. Despite extensive research, disease-modifying treatments remain elusive, with current management primarily focusing on symptom control. This review explores the complex mechanisms underlying DN and highlights recent advances in diagnostic and therapeutic strategies. Emerging insights into the molecular and cellular pathways have unveiled potential targets for intervention, including neuroprotective agents, gene and stem cell therapies, and innovative pharmacological approaches. Additionally, novel diagnostic tools, such as corneal confocal microscopy and biomarker-based tests, have improved early detection and intervention. Lifestyle modifications and multidisciplinary care strategies can enhance patient outcomes. While significant progress has been made, further research is required to develop therapies that can effectively halt or reverse disease progression, ultimately improving the lives of individuals with DN. This review provides a comprehensive overview of current understanding and future directions in DN research and management.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, China.
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Bing Zhao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuanzhe Wang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongli Lan
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinyu Liu
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yue Hu
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Peng Cao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, China.
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
2
|
Tao Y, Zhang HY, MacGilchrist C, Kirwan E, McIntosh C. Prevalence and risk factors of painful diabetic neuropathy: A systematic review and meta-analysis. Diabetes Res Clin Pract 2025; 222:112099. [PMID: 40107621 DOI: 10.1016/j.diabres.2025.112099] [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: 12/13/2024] [Revised: 02/24/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Painful diabetes-related peripheral neuropathy (PDPN) is a common and debilitating complication of diabetes, contributing significantly to morbidity and healthcare costs. This systematic review and meta-analysis aim to determine the global prevalence of PDPN among individuals with diabetic peripheral neuropathy (DPN) and to identify associated risk factors. A comprehensive search of four English and three Chinese databases was conducted for observational studies on PDPN prevalence up to June 22, 2024. Of the 41 studies included, the pooled global prevalence of PDPN was 46.7 % (95 % CI, 41.8-51.7). In subgroup analysis, significant statistical differences were observed in prevalence estimates between different diagnostic methods for neuropathic pain, with neuropathic-specific pain scales indicating higher rates (P = 0.03). Studies with mean diabetes duration of less than 10 years or more than 15 years reported higher prevalence (P < 0.01). Significant risk factors for PDPN included older age (OR = 1.02, 95 % CI, 1.01-1.04), female gender (OR = 1.58, 95 % CI, 1.19-2.11), BMI ≥ 30 kg/m2 (OR = 1.62, 95 % CI, 1.43-1.83), longer diabetes duration (OR = 1.05, 95 % CI, 1.01-1.08), and nephropathy (OR = 1.32, 95 % CI, 1.24-1.40). Targeted screening and standardized diagnostic tools are urgently needed to enhance PDPN management and mitigate its burden globally.
Collapse
Affiliation(s)
- Y Tao
- Discipline of Podiatric Medicine, School of Health Sciences, University of Galway, Galway H91 TK33, Ireland
| | - H Y Zhang
- Xiangya Nursing School, Central South University, Changsha, Hunan Province, China
| | - C MacGilchrist
- Discipline of Podiatric Medicine, School of Health Sciences, University of Galway, Galway H91 TK33, Ireland; Alliance for Research and Innovation in Wounds, College of Medicine, Nursing & Health Sciences, University of Galway, Galway H91 TK3, Ireland.
| | - E Kirwan
- Discipline of Podiatric Medicine, School of Health Sciences, University of Galway, Galway H91 TK33, Ireland
| | - C McIntosh
- Discipline of Podiatric Medicine, School of Health Sciences, University of Galway, Galway H91 TK33, Ireland; Alliance for Research and Innovation in Wounds, College of Medicine, Nursing & Health Sciences, University of Galway, Galway H91 TK3, Ireland
| |
Collapse
|
3
|
Smithiseth K, Leurcharusmee P, Sawaddiruk P, Chattipakorn N, Chattipakorn S. Unraveling the link between magnesium and diabetic neuropathy: Evidence from in vitro to clinical studies. Nutr Res 2025; 135:13-31. [PMID: 39891959 DOI: 10.1016/j.nutres.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 02/03/2025]
Abstract
Diabetic neuropathy (DN) is one of the major complications of diabetes and the most common cause of neuropathic pain. Although the underlying pathological mechanisms remain unclear, several studies have produced conflicting results regarding the link between magnesium (Mg) concentration and DN. This ambiguity raises questions about the potential benefits of Mg supplementation in individuals with DN. Therefore, this comprehensive review summarizes and discusses the evidence from clinical, in vitro, and in vivo studies on the association between Mg and DN. Several findings indicate that Mg depletion is linked to the presence of neuropathy in diabetic patients. Additionally, low Mg concentration may contribute to the onset or worsening of DN by promoting axonal degeneration through various pathways. Furthermore, multiple studies have shown that Mg supplementation can have neuroprotective effects. These findings suggest potential as an alternative or complementary therapy for preventing and treating DN in the future.
Collapse
Affiliation(s)
- Kannika Smithiseth
- Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Passakorn Sawaddiruk
- Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn Chattipakorn
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
4
|
Kuo H, Lin C, Tsai S, Chen C, Lyu R, Chu C, Ro L, Liao M, Chang H, Weng Y, Hwang J. Blood metabolomic profile in patients with type 2 diabetes mellitus with diabetic peripheral neuropathic pain. J Diabetes Investig 2025; 16:246-256. [PMID: 39548809 PMCID: PMC11786186 DOI: 10.1111/jdi.14355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/17/2024] [Accepted: 11/01/2024] [Indexed: 11/18/2024] Open
Abstract
AIMS This study aimed to identify metabolic markers for diabetic peripheral neuropathic pain (DPNP) in patients with type 2 diabetes mellitus (T2DM). MATERIALS AND METHODS Blood metabolite levels in the amino acid, biogenic amine, sphingomyelin, phosphatidylcholine (PC), carnitines, and hexose classes were analyzed in nondiabetic control (n = 27), T2DM without DPNP (n = 58), and T2DM with DPNP (n = 29) using liquid chromatography tandem mass spectrometry. Variable importance projection (VIP) evaluation by partial least squares discriminant analysis was performed on clinical parameters and metabolites. RESULTS Sixteen variables with VIP > 1.0 (P < 0.05) were identified across all patient groups, and 5 variables were identified to discriminate between the two T2DM groups. DPNP patients showed elevated fasting blood glucose, glutamate, PC aa C36:1, lysoPC a C18:1, and lysoPC a C18:2, while low-density lipoprotein cholesterol, phenylalanine, and tryptophan were reduced. Glutamate, lysoPC a C18:1, and lysoPC a C18:2 discriminated T2DM with DPNP from those without DPNP with an AUC of 0.671. The AUC was improved to 0.765 when ratios of metabolite pairs were considered. INTERPRETATION Blood metabolites include glutamate, and phospholipid-related metabolites implicated in neuropathic pain may have the potential as biomarkers for DPNP. Further investigation is required to understand the mechanism of action of these altered metabolites in DPNP.
Collapse
Affiliation(s)
- Hung‐Chou Kuo
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Chia‐Ni Lin
- Department of Laboratory MedicineChang Gung Memorial HospitalTaoyuan CityTaiwan
- Department of Medical Biotechnology and Laboratory Science, College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Sung‐Sheng Tsai
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
| | - Chiung‐Mei Chen
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Rong‐Kuo Lyu
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Chun‐Che Chu
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Long‐Sun Ro
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Ming‐Feng Liao
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Hong‐Shiu Chang
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Yi‐Ching Weng
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Jawl‐Shan Hwang
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
| |
Collapse
|
5
|
Goudman L, De Smedt A, Jansen J, Billot M, Roulaud M, Rigoard P, Moens M. Virtual Reality to Improve Sleep Quality in Patients Suffering from Painful Diabetic Polyneuropathy: A Proof of Concept Study. J Clin Med 2024; 13:7163. [PMID: 39685622 DOI: 10.3390/jcm13237163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/20/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: Sleep disturbance is often observed in the context of chronic pain. We hypothesize that, by providing an immersive Virtual Reality (VR) experience with a serious game to chronic pain patients an hour before bedtime, attention can be diverted from the pain condition, consequently leading to improved sleep quality. The aim is to evaluate the efficacy of VR compared to usual care in reducing the number of awakenings during the night and increasing sleep efficiency in patients suffering from painful diabetic polyneuropathy (PDPN). Methods: Eight patients with PDPN were randomized to either two weeks of VR or two weeks of usual care, followed by a cross-over. The primary outcome measurements were sleep efficiency and number of awakenings during the night. As secondary outcomes, self-reported sleep quality, insomnia, pain catastrophizing, anxiety, depression, pain intensity, side effects and impression of change were evaluated. Results: Data of seven patients were analysed. Actigraphy data, self-reported sleep quality, insomnia, pain catastrophizing, anxiety, depression and pain intensity scores did not differ between usual care and VR. As for impression of change, more patients improved after VR compared to usual care (V = 21, p = 0.03). Conclusions: A 2-week period of pain neuroscience education through VR did not result in increased sleep efficiency or fewer awakenings compared to usual care in patients with PDPN. These pilot results indicate that patients subjectively experience an improvement, yet this is not substantiated by either self-reported or objective measurements.
Collapse
Affiliation(s)
- Lisa Goudman
- STIMULUS Research Group, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Cluster Neurosciences, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
- Pain in Motion (PAIN) Research Group, Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Research Foundation-Flanders (FWO), 1090 Brussels, Belgium
| | - Ann De Smedt
- STIMULUS Research Group, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Cluster Neurosciences, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Department of Physical Medicine and Rehabilitation, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Julie Jansen
- STIMULUS Research Group, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Maxime Billot
- CHU de Poitiers, PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), F-86000 Poitiers, France
| | - Manuel Roulaud
- CHU de Poitiers, PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), F-86000 Poitiers, France
| | - Philippe Rigoard
- CHU de Poitiers, PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), F-86000 Poitiers, France
- CHU de Poitiers, service de Neurochirurgie du rachis, chirurgie de la douleur et du handicap, F-86000 Poitiers, France
- Pprime Institute UPR 3346, CNRS, ISAE-ENSMA, Université de Poitiers, F-86000 Poitiers, France
| | - Maarten Moens
- STIMULUS Research Group, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Cluster Neurosciences, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
- Pain in Motion (PAIN) Research Group, Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Research Foundation-Flanders (FWO), 1090 Brussels, Belgium
- Department of Radiology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| |
Collapse
|
6
|
Brask-Thomsen PK, Itani M, Karlsson P, Kristensen AG, Krøigård T, Jensen TS, Tankisi H, Sindrup SH, Finnerup NB, Gylfadottir SS. Development and Progression of Polyneuropathy Over 5 Years in Patients With Type 2 Diabetes. Neurology 2024; 103:e209652. [PMID: 39008800 DOI: 10.1212/wnl.0000000000209652] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND AND OBJECTIVES There is a need for knowledge regarding the natural course of diabetic polyneuropathy (DPN), a complication in type 2 diabetes (T2D). The aim of this study was to examine the development of DPN over time. METHODS Patients with newly diagnosed T2D, recruited from a national cohort, and controls without diabetes of similar age and sex, underwent sensory phenotyping in 2016-2018. The Toronto consensus criteria were used to classify patients into possible, probable, and confirmed DPN. For this 5-year, observational, follow-up, cohort study, all participants were invited to a reexamination combining bedside sensory examination, quantitative sensory testing (QST), nerve conduction studies (NCSs), and skin biopsies measuring intraepidermal nerve fiber density (IENFD) in order to compare phenotypic and diagnostic changes over time. RESULTS Of the baseline 389 patients and 97 controls, 184 patients (median [interquartile range] diabetes duration 5.9 [4.1-7.4] years, mean hemoglobin A1c [HbA1c] 51 ± 11 mmol/mol at baseline) and 43 controls completed follow-up (46.9%). Confirmed DPN was present in 35.8% and 50.3%, probable DPN in 27.2% and 14.6%, possible DPN in 17.2% and 16.6%, and no DPN in 15.2% and 17.9% at baseline and follow-up, respectively. The estimated prevalence (95% CI) of confirmed DPN was 33.5% (24.9-42.1) compared with 22.7% (17.5-28.0) at baseline. During the follow-up period, 43.9% of patients with probable DPN developed confirmed DPN. Progression of neuropathy occurred in 16.5% and 24.7% and regression in 5.9% and 18.6% of patients based on NCS and IENFD, respectively. Progression based on NCS and/or IENFD was associated with higher baseline waist circumference and triglycerides, and regression with lower baseline HbA1c. Patients with at least probable DPN at baseline but neither patients without DPN nor controls developed increased spread of hyposensitivity, more hyposensitivity on QST and lower NCS z-scores at follow-up, and worsening of nerve parameters at follow-up correlated with higher baseline triglycerides. DISCUSSION In patients with well-regulated T2D, the proportion of patients with confirmed DPN increased over 5 years driven by progression from probable DPN. A large proportion of patients progressed, and a smaller proportion regressed on nerve parameters. Higher triglycerides correlated with this progression and may constitute a risk factor.
Collapse
Affiliation(s)
- Peter Kolind Brask-Thomsen
- From the Danish Pain Research Center (P.K.B.-T., P.K., A.G.K., T.S.J., N.B.F., S.S.G.), Department of Clinical Medicine, Aarhus University; Steno Diabetes Center Aarhus (P.K.B.-T., T.S.J.), Aarhus University Hospital; Department of Neurology (M.I., T.K., S.H.S.), Odense University Hospital; Core Center for Molecular Morphology (P.K.), Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University; Department of Clinical Neurophysiology (A.G.K., H.T.), Aarhus University Hospital; Department of Neurophysiology (T.K.), Odense University Hospital; Department of Clinical Research (T.K.), University of Southern Denmark, Odense; Department of Clinical Medicine (H.T.), Aarhus University; and Department of Neurology (N.B.F., S.S.G.), Aarhus University Hospital, Denmark
| | - Mustapha Itani
- From the Danish Pain Research Center (P.K.B.-T., P.K., A.G.K., T.S.J., N.B.F., S.S.G.), Department of Clinical Medicine, Aarhus University; Steno Diabetes Center Aarhus (P.K.B.-T., T.S.J.), Aarhus University Hospital; Department of Neurology (M.I., T.K., S.H.S.), Odense University Hospital; Core Center for Molecular Morphology (P.K.), Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University; Department of Clinical Neurophysiology (A.G.K., H.T.), Aarhus University Hospital; Department of Neurophysiology (T.K.), Odense University Hospital; Department of Clinical Research (T.K.), University of Southern Denmark, Odense; Department of Clinical Medicine (H.T.), Aarhus University; and Department of Neurology (N.B.F., S.S.G.), Aarhus University Hospital, Denmark
| | - Pall Karlsson
- From the Danish Pain Research Center (P.K.B.-T., P.K., A.G.K., T.S.J., N.B.F., S.S.G.), Department of Clinical Medicine, Aarhus University; Steno Diabetes Center Aarhus (P.K.B.-T., T.S.J.), Aarhus University Hospital; Department of Neurology (M.I., T.K., S.H.S.), Odense University Hospital; Core Center for Molecular Morphology (P.K.), Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University; Department of Clinical Neurophysiology (A.G.K., H.T.), Aarhus University Hospital; Department of Neurophysiology (T.K.), Odense University Hospital; Department of Clinical Research (T.K.), University of Southern Denmark, Odense; Department of Clinical Medicine (H.T.), Aarhus University; and Department of Neurology (N.B.F., S.S.G.), Aarhus University Hospital, Denmark
| | - Alexander G Kristensen
- From the Danish Pain Research Center (P.K.B.-T., P.K., A.G.K., T.S.J., N.B.F., S.S.G.), Department of Clinical Medicine, Aarhus University; Steno Diabetes Center Aarhus (P.K.B.-T., T.S.J.), Aarhus University Hospital; Department of Neurology (M.I., T.K., S.H.S.), Odense University Hospital; Core Center for Molecular Morphology (P.K.), Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University; Department of Clinical Neurophysiology (A.G.K., H.T.), Aarhus University Hospital; Department of Neurophysiology (T.K.), Odense University Hospital; Department of Clinical Research (T.K.), University of Southern Denmark, Odense; Department of Clinical Medicine (H.T.), Aarhus University; and Department of Neurology (N.B.F., S.S.G.), Aarhus University Hospital, Denmark
| | - Thomas Krøigård
- From the Danish Pain Research Center (P.K.B.-T., P.K., A.G.K., T.S.J., N.B.F., S.S.G.), Department of Clinical Medicine, Aarhus University; Steno Diabetes Center Aarhus (P.K.B.-T., T.S.J.), Aarhus University Hospital; Department of Neurology (M.I., T.K., S.H.S.), Odense University Hospital; Core Center for Molecular Morphology (P.K.), Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University; Department of Clinical Neurophysiology (A.G.K., H.T.), Aarhus University Hospital; Department of Neurophysiology (T.K.), Odense University Hospital; Department of Clinical Research (T.K.), University of Southern Denmark, Odense; Department of Clinical Medicine (H.T.), Aarhus University; and Department of Neurology (N.B.F., S.S.G.), Aarhus University Hospital, Denmark
| | - Troels S Jensen
- From the Danish Pain Research Center (P.K.B.-T., P.K., A.G.K., T.S.J., N.B.F., S.S.G.), Department of Clinical Medicine, Aarhus University; Steno Diabetes Center Aarhus (P.K.B.-T., T.S.J.), Aarhus University Hospital; Department of Neurology (M.I., T.K., S.H.S.), Odense University Hospital; Core Center for Molecular Morphology (P.K.), Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University; Department of Clinical Neurophysiology (A.G.K., H.T.), Aarhus University Hospital; Department of Neurophysiology (T.K.), Odense University Hospital; Department of Clinical Research (T.K.), University of Southern Denmark, Odense; Department of Clinical Medicine (H.T.), Aarhus University; and Department of Neurology (N.B.F., S.S.G.), Aarhus University Hospital, Denmark
| | - Hatice Tankisi
- From the Danish Pain Research Center (P.K.B.-T., P.K., A.G.K., T.S.J., N.B.F., S.S.G.), Department of Clinical Medicine, Aarhus University; Steno Diabetes Center Aarhus (P.K.B.-T., T.S.J.), Aarhus University Hospital; Department of Neurology (M.I., T.K., S.H.S.), Odense University Hospital; Core Center for Molecular Morphology (P.K.), Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University; Department of Clinical Neurophysiology (A.G.K., H.T.), Aarhus University Hospital; Department of Neurophysiology (T.K.), Odense University Hospital; Department of Clinical Research (T.K.), University of Southern Denmark, Odense; Department of Clinical Medicine (H.T.), Aarhus University; and Department of Neurology (N.B.F., S.S.G.), Aarhus University Hospital, Denmark
| | - Søren H Sindrup
- From the Danish Pain Research Center (P.K.B.-T., P.K., A.G.K., T.S.J., N.B.F., S.S.G.), Department of Clinical Medicine, Aarhus University; Steno Diabetes Center Aarhus (P.K.B.-T., T.S.J.), Aarhus University Hospital; Department of Neurology (M.I., T.K., S.H.S.), Odense University Hospital; Core Center for Molecular Morphology (P.K.), Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University; Department of Clinical Neurophysiology (A.G.K., H.T.), Aarhus University Hospital; Department of Neurophysiology (T.K.), Odense University Hospital; Department of Clinical Research (T.K.), University of Southern Denmark, Odense; Department of Clinical Medicine (H.T.), Aarhus University; and Department of Neurology (N.B.F., S.S.G.), Aarhus University Hospital, Denmark
| | - Nanna B Finnerup
- From the Danish Pain Research Center (P.K.B.-T., P.K., A.G.K., T.S.J., N.B.F., S.S.G.), Department of Clinical Medicine, Aarhus University; Steno Diabetes Center Aarhus (P.K.B.-T., T.S.J.), Aarhus University Hospital; Department of Neurology (M.I., T.K., S.H.S.), Odense University Hospital; Core Center for Molecular Morphology (P.K.), Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University; Department of Clinical Neurophysiology (A.G.K., H.T.), Aarhus University Hospital; Department of Neurophysiology (T.K.), Odense University Hospital; Department of Clinical Research (T.K.), University of Southern Denmark, Odense; Department of Clinical Medicine (H.T.), Aarhus University; and Department of Neurology (N.B.F., S.S.G.), Aarhus University Hospital, Denmark
| | - Sandra Sif Gylfadottir
- From the Danish Pain Research Center (P.K.B.-T., P.K., A.G.K., T.S.J., N.B.F., S.S.G.), Department of Clinical Medicine, Aarhus University; Steno Diabetes Center Aarhus (P.K.B.-T., T.S.J.), Aarhus University Hospital; Department of Neurology (M.I., T.K., S.H.S.), Odense University Hospital; Core Center for Molecular Morphology (P.K.), Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University; Department of Clinical Neurophysiology (A.G.K., H.T.), Aarhus University Hospital; Department of Neurophysiology (T.K.), Odense University Hospital; Department of Clinical Research (T.K.), University of Southern Denmark, Odense; Department of Clinical Medicine (H.T.), Aarhus University; and Department of Neurology (N.B.F., S.S.G.), Aarhus University Hospital, Denmark
| |
Collapse
|
7
|
Elliott J, Sloan G, Stevens L, Selvarajah D, Cruccu G, Gandhi RA, Kempler P, Fuller JH, Chaturvedi N, Tesfaye S. Female sex is a risk factor for painful diabetic peripheral neuropathy: the EURODIAB prospective diabetes complications study. Diabetologia 2024; 67:190-198. [PMID: 37870649 PMCID: PMC10709240 DOI: 10.1007/s00125-023-06025-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/05/2023] [Indexed: 10/24/2023]
Abstract
AIMS/HYPOTHESIS While the risk factors for diabetic peripheral neuropathy (DPN) are now well recognised, the risk factors for painful DPN remain unknown. We performed analysis of the EURODIAB Prospective Complications Study data to elucidate the incidence and risk factors of painful DPN. METHODS The EURODIAB Prospective Complications Study recruited 3250 participants with type 1 diabetes who were followed up for 7.3±0.6 (mean ± SD) years. To evaluate DPN, a standardised protocol was used, including clinical assessment, quantitative sensory testing and autonomic function tests. Painful DPN (defined as painful neuropathic symptoms in the legs in participants with confirmed DPN) was assessed at baseline and follow-up. RESULTS At baseline, 234 (25.2%) out of 927 participants with DPN had painful DPN. At follow-up, incident DPN developed in 276 (23.5%) of 1172 participants. Of these, 41 (14.9%) had incident painful DPN. Most of the participants who developed incident painful DPN were female (73% vs 48% painless DPN p=0.003) and this remained significant after adjustment for duration of diabetes and HbA1c (OR 2.69 [95% CI 1.41, 6.23], p=0.004). The proportion of participants with macro- or microalbuminuria was lower in those with painful DPN compared with painless DPN (15% vs 34%, p=0.02), and this association remained after adjusting for HbA1c, diabetes duration and sex (p=0.03). CONCLUSIONS/INTERPRETATION In this first prospective study to investigate the risk factors for painful DPN, we definitively demonstrate that female sex is a risk factor for painful DPN. Additionally, there is less evidence of diabetic nephropathy in incident painful, compared with painless, DPN. Thus, painful DPN is not driven by cardiometabolic factors traditionally associated with microvascular disease. Sex differences may therefore play an important role in the pathophysiology of neuropathic pain in diabetes. Future studies need to look at psychosocial, genetic and other factors in the development of painful DPN.
Collapse
Affiliation(s)
- Jackie Elliott
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield, UK
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Gordon Sloan
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield, UK
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Lynda Stevens
- Department of Epidemiology and Public Health, University College, London, UK
| | - Dinesh Selvarajah
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield, UK
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Giorgio Cruccu
- Department of Neurological Sciences, La Sapienza University, Rome, Italy
| | - Rajiv A Gandhi
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield, UK
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Peter Kempler
- First Department of Medicine, Semmelweis University, Budapest, Hungary
| | - John H Fuller
- Epidemiology and Public Health, Imperial College of Science, Technology & Medicine, London, UK
| | - Nishi Chaturvedi
- MRC Unit for Lifelong Health & Ageing at UCL, Institute of Cardiovascular Sciences, University College London, London, UK
| | - Solomon Tesfaye
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield, UK.
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK.
| |
Collapse
|
8
|
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]
|
9
|
Staehelin Jensen T. The pathogenesis of painful diabetic neuropathy and clinical presentation. Diabetes Res Clin Pract 2023; 206 Suppl 1:110753. [PMID: 38245319 DOI: 10.1016/j.diabres.2023.110753] [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: 05/01/2023] [Accepted: 05/30/2023] [Indexed: 01/22/2024]
Abstract
Diabetic neuropathy is a common complication of diabetes that affects up to 50% of patients during the course of the disease; 20-30% of the patients also develop neuropathic pain. The mechanisms underlying neuropathy are not known in detail, but both metabolic and vascular factors may contribute to the development of neuropathy. The development of the most common type of neuropathy is insidious, often starting distally in the toes and feet and gradually ascending up the leg and later also involving fingers and hands. The symptoms are mainly sensory with either sensory loss or positive symptoms with different types of paresthesia or painful sensations. In more advanced cases motor dysfunction may occur, causing gait disturbances and falls. The diagnosis of neuropathy is based on history and a careful examination, which includes a sensory examination of both large and small sensory nerve fiber function, as well as an examination of motor function and deep tendon reflexes of the lower limbs. Attention needs to be paid to the feet including examination of the skin, joints, and vascular supply. Nerve conduction studies are rarely needed to make a diagnosis of neuropathy. In patients with clear motor deficit or with an asymmetrical presentation, additional electrophysiological examination may be necessary. Early detection of diabetic neuropathy is important to avoid further irreversible injury to the peripheral nerves.
Collapse
Affiliation(s)
- Troels Staehelin Jensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
10
|
Tesfaye S, Brill S, Eerdekens M, Labrador MM, Petersen G, de Rooij Peek A, Reta A, Ryan D, Schaper N, Tölle T, Truini A, Ziegler D. Diagnosis, management and impact of painful diabetic peripheral neuropathy: A patient survey in four European countries. J Diabetes Complications 2023; 37:108417. [PMID: 36905720 DOI: 10.1016/j.jdiacomp.2023.108417] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/16/2023] [Accepted: 02/01/2023] [Indexed: 02/12/2023]
Abstract
AIMS The aim of this study was to assess patient perspectives and experiences of the impact of neuropathic pain, painful diabetic neuropathy (pDPN) diagnosis and treatment, and the patient-healthcare professional (HCP) relationship. METHODS We conducted a quantitative online survey in Germany, the Netherlands, Spain, and the UK among adults with diabetes who responded "yes" to at least four of ten questions of in the Douleur Neuropathique en 4 Questions (DN4) questionnaire. RESULTS Of 3626 respondents, 576 met the eligibility criteria. Daily pain was rated as moderate or severe by 79 % of respondents. Most participants reported a negative impact of their pain on sleep (74 %), mood (71 %), exercise (69 %), concentration (64 %) and daily activities (62 %), and 75 % of those in employment had missed work because of their pain in the past year. Overall, 22 % of respondents avoided discussing pain with their HCP, 50 % had not received formal pDPN diagnosis, and 56 % had not used prescribed pain medications. Although two-thirds (67 %) of respondents reported feeling satisfied or very satisfied with treatment, 82 % of these patients still experienced daily moderate or severe pain. CONCLUSIONS Neuropathic pain in people with diabetes affects daily life and remains underdiagnosed and undertreated in clinical practice.
Collapse
Affiliation(s)
| | - Silviu Brill
- Institute of Pain Medicine, Tel Aviv Medical Center, Israel
| | | | | | | | | | | | | | - Nicolaas Schaper
- Division of Endocrinology, Maastricht University Hospital, Netherlands
| | - Thomas Tölle
- Neurologische Klinik und Poliklinik der TU, München, Germany
| | - Andrea Truini
- Department of Human Neuroscience, Università Sapienza Rome, Italy
| | - Dan Ziegler
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
11
|
Galosi E, Leonardi L, Falco P, Di Pietro G, Fasolino A, Esposito N, Leone C, Di Stefano G, Inghilleri M, Luigetti M, Giovanni A, Truini A. Functional and morphometric assessment of small-fibre damage in late-onset hereditary transthyretin amyloidosis with polyneuropathy: the controversial relation between small-fibre-related symptoms and diagnostic test findings. Amyloid 2023; 30:59-66. [PMID: 36094793 DOI: 10.1080/13506129.2022.2120799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
INTRODUCTION We aimed at investigating whether functional and morphometric tests assessing small-fibre damage, ie quantitative sensory testing, Sudoscan and skin biopsy, reliably reflect neuropathic pain and autonomic symptoms in patients with late-onset hereditary transthyretin amyloidosis with polyneuropathy (ATTRv-PN). METHODS In 30 patients with late-onset ATTRv-PN, we collected quantitative sensory testing, Sudoscan and skin biopsy with assessment of intraepidermal, piloerector muscle and sweat gland nerve fibre density. We then correlated these functional and morphometric parameters with neuropathic pain and autonomic symptoms as assessed with the Neuropathic Pain Symptom Inventory (NPSI) and Composite Autonomic Symptom Score-31 (COMPASS-31). RESULTS 50% of patients showed small-fibre damage in the form of a pure small-fibre neuropathy, 47% in the context of a mixed fibre neuropathy with small and large fibre involvement. All patients complained of at least one autonomic symptom and 60% had neuropathic pain. Whereas quantitative sensory testing and Sudoscan parameters correlated with neuropathic pain and autonomic symptoms as assessed by NPSI and COMPASS-31, intraepidermal, piloerector muscle and sweat gland nerve fibre density quantification did not. CONCLUSIONS Our findings indicate that functional test parameters reliably reflect neuropathic pain and autonomic symptoms related to small-fibre damage. These findings might help to identify clinically useful biomarkers to assess patient follow-up.
Collapse
Affiliation(s)
- Eleonora Galosi
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | - Luca Leonardi
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Sapienza University, Rome, Italy
| | - Pietro Falco
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | | | | | | | - Caterina Leone
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | | | | | - Marco Luigetti
- Dipartimento Universitario di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy.,UOC Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonini Giovanni
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Sapienza University, Rome, Italy
| | - Andrea Truini
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| |
Collapse
|
12
|
Ang L, Mizokami-Stout K, Eid SA, Elafros M, Callaghan B, Feldman EL, Pop-Busui R. The conundrum of diabetic neuropathies-Past, present, and future. J Diabetes Complications 2022; 36:108334. [PMID: 36306721 PMCID: PMC10202025 DOI: 10.1016/j.jdiacomp.2022.108334] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/01/2022] [Accepted: 10/01/2022] [Indexed: 10/31/2022]
Abstract
Diabetic neuropathy (DN) remains arguably the most prevalent chronic complication in people with both type 1 and type 2 diabetes, including in youth, despite changes in the current standards of clinical care. Additionally, emerging evidence demonstrates that neuropathy affects a large proportion of people with undiagnosed diabetes and/or prediabetes, as well as those with obesity. Here we summarize the latest epidemiology of DN, recent findings regarding the pathophysiology of the disease, as well as current outcome measures for screening and diagnosis, in research and clinical settings. The authors discuss novel perspectives on the impact of social determinants of health in DN development and management, and the latest evidence on effective therapies, including pharmacological and nonpharmacological therapies for neuropathic pain. Throughout the publication, we identify knowledge gaps and the need for future funding to address these gaps, as well as needs to advocate for a personalized care approach to reduce the burden of DN and optimize quality of life for all affected individuals.
Collapse
Affiliation(s)
- Lynn Ang
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, United States of America
| | - Kara Mizokami-Stout
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, United States of America; Ann Arbor Veteran Affairs Hospital, Ann Arbor, MI, United States of America
| | - Stephanie A Eid
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States of America
| | - Melissa Elafros
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States of America
| | - Brian Callaghan
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States of America
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States of America
| | - Rodica Pop-Busui
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, United States of America.
| |
Collapse
|
13
|
Zhu GC, Chen YW, Tsai KL, Wang JJ, Hung CH, Schmid AB. Effects of Neural Mobilization on Sensory Dysfunction and Peripheral Nerve Degeneration in Rats With Painful Diabetic Neuropathy. Phys Ther 2022; 102:pzac104. [PMID: 35913760 PMCID: PMC7613682 DOI: 10.1093/ptj/pzac104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 03/16/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022]
Abstract
OBJECTIVE This study aims to evaluate the effectiveness of neural mobilization (NM) in the management of sensory dysfunction and nerve degeneration related to experimental painful diabetic neuropathy (PDN). METHODS This is a pre-clinical animal study performed in the streptozocin-induced diabetic rat model. Three groups were included: a treatment group of rats with PDN receiving NM under anesthesia (PDN-NM, n = 10), a sham treatment group of rats with PDN that received only anesthesia (PDN-Sham, n = 9), and a vehicle control group with nondiabetic animals (Vehicle, n = 10). Rats in the PDN-NM and PDN-Sham groups received 1 treatment session on days 10, 12, and 14 after streptozocin injection, with a 48-hour rest period between sessions. Behavioral tests were performed using von Frey and Plantar tests. Evaluation for peripheral nerve degeneration was performed through measuring protein gene product 9.5-positive intra-epidermal nerve fiber density in hind-paw skin biopsies. All measurements were performed by a blinded investigator. RESULTS The behavioral tests showed that a single NM session could reduce hyperalgesia, which was maintained for 48 hours. The second treatment session further improved this treatment effect, and the third session maintained it. These results suggest that it requires multiple treatment sessions to produce and maintain hypoalgesic effects. Skin biopsy analysis showed that the protein gene product 9.5-positive intra-epidermal nerve fiber density was higher on the experimental side of the PDN-NM group compared with the PDN-Sham group, suggesting NM may mitigate the degeneration of peripheral nerves. CONCLUSION This study demonstrated that NM may be an effective method to manage experimentally induced PDN, potentially through mitigation of nerve degeneration. Further studies are needed to develop standardized protocols for clinical use. IMPACT These findings provide neurophysiological evidence for the use of NM in PDN and can form the basis for the development of physical therapy-based programs in clinics.
Collapse
Affiliation(s)
- Guan-Cheng Zhu
- Department of Physical Therapy, National Cheng Kung University, Tainan, Taiwan (R.O.C.)
| | - Yu-Wen Chen
- Department of Physical Therapy, China Medical University, Taichung, Taiwan (R.O.C.)
| | - Kun-Ling Tsai
- Department of Physical Therapy, National Cheng Kung University, Tainan, Taiwan (R.O.C.)
| | - Jhi-Joung Wang
- Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan (R.O.C.)
| | - Ching-Hsia Hung
- Department of Physical Therapy, National Cheng Kung University, Tainan, Taiwan (R.O.C.)
| | - Annina B Schmid
- Nuffield Department of Clinical Neuroscience, University of Oxford, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
14
|
Blonde L, Umpierrez GE, Reddy SS, McGill JB, Berga SL, Bush M, Chandrasekaran S, DeFronzo RA, Einhorn D, Galindo RJ, Gardner TW, Garg R, Garvey WT, Hirsch IB, Hurley DL, Izuora K, Kosiborod M, Olson D, Patel SB, Pop-Busui R, Sadhu AR, Samson SL, Stec C, Tamborlane WV, Tuttle KR, Twining C, Vella A, Vellanki P, Weber SL. American Association of Clinical Endocrinology Clinical Practice Guideline: Developing a Diabetes Mellitus Comprehensive Care Plan-2022 Update. Endocr Pract 2022; 28:923-1049. [PMID: 35963508 PMCID: PMC10200071 DOI: 10.1016/j.eprac.2022.08.002] [Citation(s) in RCA: 221] [Impact Index Per Article: 73.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The objective of this clinical practice guideline is to provide updated and new evidence-based recommendations for the comprehensive care of persons with diabetes mellitus to clinicians, diabetes-care teams, other health care professionals and stakeholders, and individuals with diabetes and their caregivers. METHODS The American Association of Clinical Endocrinology selected a task force of medical experts and staff who updated and assessed clinical questions and recommendations from the prior 2015 version of this guideline and conducted literature searches for relevant scientific papers published from January 1, 2015, through May 15, 2022. Selected studies from results of literature searches composed the evidence base to update 2015 recommendations as well as to develop new recommendations based on review of clinical evidence, current practice, expertise, and consensus, according to established American Association of Clinical Endocrinology protocol for guideline development. RESULTS This guideline includes 170 updated and new evidence-based clinical practice recommendations for the comprehensive care of persons with diabetes. Recommendations are divided into four sections: (1) screening, diagnosis, glycemic targets, and glycemic monitoring; (2) comorbidities and complications, including obesity and management with lifestyle, nutrition, and bariatric surgery, hypertension, dyslipidemia, retinopathy, neuropathy, diabetic kidney disease, and cardiovascular disease; (3) management of prediabetes, type 2 diabetes with antihyperglycemic pharmacotherapy and glycemic targets, type 1 diabetes with insulin therapy, hypoglycemia, hospitalized persons, and women with diabetes in pregnancy; (4) education and new topics regarding diabetes and infertility, nutritional supplements, secondary diabetes, social determinants of health, and virtual care, as well as updated recommendations on cancer risk, nonpharmacologic components of pediatric care plans, depression, education and team approach, occupational risk, role of sleep medicine, and vaccinations in persons with diabetes. CONCLUSIONS This updated clinical practice guideline provides evidence-based recommendations to assist with person-centered, team-based clinical decision-making to improve the care of persons with diabetes mellitus.
Collapse
Affiliation(s)
| | | | - S Sethu Reddy
- Central Michigan University, Mount Pleasant, Michigan
| | | | | | | | | | | | - Daniel Einhorn
- Scripps Whittier Diabetes Institute, La Jolla, California
| | | | | | - Rajesh Garg
- Lundquist Institute/Harbor-UCLA Medical Center, Torrance, California
| | | | | | | | | | | | - Darin Olson
- Colorado Mountain Medical, LLC, Avon, Colorado
| | | | | | - Archana R Sadhu
- Houston Methodist; Weill Cornell Medicine; Texas A&M College of Medicine; Houston, Texas
| | | | - Carla Stec
- American Association of Clinical Endocrinology, Jacksonville, Florida
| | | | - Katherine R Tuttle
- University of Washington and Providence Health Care, Seattle and Spokane, Washington
| | | | | | | | - Sandra L Weber
- University of South Carolina School of Medicine-Greenville, Prisma Health System, Greenville, South Carolina
| |
Collapse
|
15
|
Russo GT, Manicardi V, Rossi MC, Orsi E, Solini A. Sex- and gender-differences in chronic long-term complications of type 1 and type 2 diabetes mellitus in Italy. Nutr Metab Cardiovasc Dis 2022; 32:2297-2309. [PMID: 36064685 DOI: 10.1016/j.numecd.2022.08.011] [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: 05/04/2022] [Revised: 08/02/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
AIMS This review summarizes the contribution of Italian diabetologists devoted to a better understanding of the complex relationship linking sex/gender and long-term complications of type 1 (T1DM) and type 2 diabetes (T2DM) over the last fifteen years. DATA SYNTHESIS Microvascular and macrovascular complications of diabetes show sex- and gender-related differences, involving pathophysiological mechanisms, epidemiological features and clinical presentation, due to the interaction between biological and psychosocial factors. These differences greatly impact on the progression of diabetes and its long-term complications, especially in the cardiovascular, renal and liver districts. CONCLUSION A better knowledge of such sex- and gender-related characteristics is required for a more precise patient phenotypization, and for the choice of a personalized antihyperglycemic treatment. Despite such mounting evidence, current diabetes clinical guidelines do not as yet adequately consider sex/gender differences.
Collapse
Affiliation(s)
- G T Russo
- Department of Clinical and Experimental Medicine, University of Messina, Italy.
| | | | - M C Rossi
- CORESEARCH - Center for Outcomes Research and Clinical Epidemiology, Pescara, Italy
| | - E Orsi
- IRCCS Foundation Cà Grande Ospedale Maggiore, Milan, Italy
| | - A Solini
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Italy.
| |
Collapse
|
16
|
Muacevic A, Adler JR. Diabetic Polyneuropathy in Type 1 and Type 2 Diabetes Mellitus: A Cross-Sectional Study. Cureus 2022; 14:e30004. [PMID: 36348847 PMCID: PMC9637022 DOI: 10.7759/cureus.30004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is one of the three most common chronic diseases worldwide. This study aimed to determine the prevalence of diabetic polyneuropathy (DPN) among patients with diabetes. METHOD This cross-sectional study was carried out on DM patients who visited King Abdulaziz University Hospital (KAUH) between August 2021 and February 2022. We used the Michigan Neuropathy Screening Instrument (MNSI) questionnaire to determine if the patients had DN. In addition, we used the Global Physical Activity Questionnaire (GPAQ) to assess the level of physical activity (PA) in these DM patients. RESULTS A total of 336 patients consented to participate in the study. We found a DN prevalence of 23.8% amongst DM patients treated at the KAUH. In addition, the prevalence of DN amongst T1DM and T2DM patients was found to be 16% and 24.4%, respectively. Furthermore, we found that 65% of DM patients developed complications, with a significant correlation observed between the duration of DM and the development of complications. However, patient age and sex were non-statistically significantly correlated with the development of complications. Analysis of the GPAQ showed that among the 249 patients who completed the questionnaire, none had a high physical activity level, while 4% and 96% had moderate and low physical activity levels, respectively. No association was found between physical activity and patients' age, sex, type of DM, duration of DM, and development of complications. CONCLUSION DN prevalence amongst DM patients treated at KAUH was 23.8%. The duration of diabetes was found to be a risk factor for DN. However, patient age and sex were non-statistically significantly associated with DN.
Collapse
|
17
|
D’Egidio F, Lombardozzi G, Kacem Ben Haj M’Barek HE, Mastroiacovo G, Alfonsetti M, Cimini A. The Influence of Dietary Supplementations on Neuropathic Pain. Life (Basel) 2022; 12:1125. [PMID: 36013304 PMCID: PMC9410423 DOI: 10.3390/life12081125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
Neuropathic pain is defined as pain caused by a lesion or disease of the somatosensory nervous system and affects 7-10% of the worldwide population. Neuropathic pain can be induced by the use of drugs, including taxanes, thus triggering chemotherapy-induced neuropathic pain or as consequence of metabolic disorders such as diabetes. Neuropathic pain is most often a chronic condition, and can be associated with anxiety and depression; thus, it negatively impacts quality of life. Several pharmacologic approaches exist; however, they can lead numerous adverse effects. From this perspective, the use of nutraceuticals and diet supplements can be helpful in relieve neuropathic pain and related symptoms. In this review, we discuss how diet can radically affect peripheral neuropathy, and we focus on the potential approaches to ameliorate this condition, such as the use of numerous nutritional supplements or probiotics.
Collapse
Affiliation(s)
- Francesco D’Egidio
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.D.); (G.L.); (H.E.K.B.H.M.); (G.M.); (M.A.)
| | - Giorgia Lombardozzi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.D.); (G.L.); (H.E.K.B.H.M.); (G.M.); (M.A.)
| | - Housem E. Kacem Ben Haj M’Barek
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.D.); (G.L.); (H.E.K.B.H.M.); (G.M.); (M.A.)
| | - Giada Mastroiacovo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.D.); (G.L.); (H.E.K.B.H.M.); (G.M.); (M.A.)
| | - Margherita Alfonsetti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.D.); (G.L.); (H.E.K.B.H.M.); (G.M.); (M.A.)
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.D.); (G.L.); (H.E.K.B.H.M.); (G.M.); (M.A.)
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
18
|
Kim YJ, Lee JM, Lee CW, Chung CH, Lee JH, Ko SH, Cho DH, Lee JH, Cho HC, Kim SS, Kim CH, Won JC, Park TS. Analysis of clinical phenotypes of neuropathic symptoms in patients with type 2 diabetes: A multicenter study. J Diabetes Investig 2022; 13:1852-1860. [PMID: 35779248 PMCID: PMC9623523 DOI: 10.1111/jdi.13880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/17/2022] [Accepted: 06/28/2022] [Indexed: 12/03/2022] Open
Abstract
Aims/Introduction We investigated the classification of diabetic peripheral neuropathy (DPN) patients by subjective symptoms, and identification of the relationship between the patterns and intensities of symptoms and the clustered groups of DPN patients. Materials and Methods This multicenter study analyzed epidemiological data and sensory symptoms of 649 patients with DPN. Cluster analysis was carried out to identify subgroups of patients with characteristic symptom profiles. Factor analysis was carried out to investigate the symptom patterns of the clustered groups of DPN patients. Results Three clusters of patients with DPN were identified: severe symptoms with decreased quality of life (cluster 1, n = 119, 18.3%), predominantly insensate symptoms with relatively good quality of life (cluster 2, n = 318, 49.0%), and moderate pain intensity and decreased quality of life (cluster 3, n = 204, 31.4%). The frequency of symptoms on each item of the Michigan Neuropathy Screening Instrument questionnaire showed a similar distribution according to pain intensities along with the three clusters. Conclusions Our study supports the hypothesis that diversity in sensory symptoms exists in patients with DPN. Heterogeneity in DPN patients should be taken into account for a more stratified or individualized treatment approach. Based on a multicenter study, we identified three clusters of patients with DPN. Our research supports the hypothesis that diversity in sensory symptoms exists in patients with DPN. Heterogeneity in DPN patients should be taken into account for a more stratified or individualized treatment approach.
Collapse
Affiliation(s)
- Yu Ji Kim
- Department of Internal Medicine, Research Institute of Clinical Medicine of Jeonbuk National University Medical School-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju
| | - Jeong Min Lee
- Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, the Catholic University of Korea, Seoul
| | - Chang Won Lee
- Department of Internal Medicine, Busan St. Mary's Hospital, Busan
| | - Choon Hee Chung
- Department of Internal Medicine, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju
| | - Jae Hyuk Lee
- Department of Internal Medicine, Myongji Hospital, Hanyang University College of Medicine, Goyang
| | - Seung Hyun Ko
- Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon
| | - Dong Hyeok Cho
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju
| | - Ji Hyun Lee
- Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu
| | - Ho Chan Cho
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu
| | - Sang Soo Kim
- Department of Internal Medicine, Pusan National University Hospital, Busan
| | - Chong Hwa Kim
- Department of Internal Medicine, Sejong General Hospital, Bucheon
| | - Jong Chul Won
- Department of Internal Medicine, Cardiovascular and Metabolic Disease Center, Inje University Sanggye Paik Hospital, Inje University College of Medicine, Seoul
| | - Tae Sun Park
- Department of Internal Medicine, Research Institute of Clinical Medicine of Jeonbuk National University Medical School-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju
| |
Collapse
|
19
|
Fang XX, Wang H, Song HL, Wang J, Zhang ZJ. Neuroinflammation Involved in Diabetes-Related Pain and Itch. Front Pharmacol 2022; 13:921612. [PMID: 35795572 PMCID: PMC9251344 DOI: 10.3389/fphar.2022.921612] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/12/2022] [Indexed: 12/25/2022] Open
Abstract
Diabetes mellitus (DM) is a global epidemic with increasing incidence, which results in diverse complications, seriously affects the patient quality of life, and brings huge economic burdens to society. Diabetic neuropathy is the most common chronic complication of DM, resulting in neuropathic pain and chronic itch. The precise mechanisms of diabetic neuropathy have not been fully clarified, hindering the exploration of novel therapies for diabetic neuropathy and its terrible symptoms such as diabetic pain and itch. Accumulating evidence suggests that neuroinflammation plays a critical role in the pathophysiologic process of neuropathic pain and chronic itch. Indeed, researchers have currently made significant progress in knowing the role of glial cells and the pro-inflammatory mediators produced from glial cells in the modulation of chronic pain and itch signal processing. Here, we provide an overview of the current understanding of neuroinflammation in contributing to the sensitization of the peripheral nervous system (PNS) and central nervous system (CNS). In addition, we also summarize the inflammation mechanisms that contribute to the pathogenesis of diabetic itch, including activation of glial cells, oxidative stress, and pro-inflammatory factors. Targeting excessive neuroinflammation may provide potential and effective therapies for the treatment of chronic neuropathic pain and itch in DM.
Collapse
Affiliation(s)
- Xiao-Xia Fang
- Department of Human Anatomy, School of Medicine, Nantong University, Nantong, China
- Department of Medical Functional Laboratory, School of Medicine, Nantong University, Nantong, China
| | - Heng Wang
- Department of Human Anatomy, School of Medicine, Nantong University, Nantong, China
| | - Hao-Lin Song
- Department of Human Anatomy, School of Medicine, Nantong University, Nantong, China
| | - Juan Wang
- Department of Human Anatomy, School of Medicine, Nantong University, Nantong, China
| | - Zhi-Jun Zhang
- Department of Human Anatomy, School of Medicine, Nantong University, Nantong, China
| |
Collapse
|
20
|
Granovsky Y, Shafran Topaz L, Laycock H, Zubiedat R, Crystal S, Buxbaum C, Bosak N, Hadad R, Domany E, Khamaisi M, Sprecher E, Bennett DL, Rice A, Yarnitsky D. Conditioned pain modulation is more efficient in patients with painful diabetic polyneuropathy than those with nonpainful diabetic polyneuropathy. Pain 2022; 163:827-833. [PMID: 34371518 PMCID: PMC9009321 DOI: 10.1097/j.pain.0000000000002434] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/20/2021] [Accepted: 06/22/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Endogenous pain modulation, as tested by the conditioned pain modulation (CPM) protocol, is typically less efficient in patients with chronic pain compared with healthy controls. We aimed to assess whether CPM is less efficient in patients with painful diabetic polyneuropathy (DPN) compared with those with nonpainful DPN. Characterization of the differences in central pain processing between these 2 groups might provide a central nervous system explanation to the presence or absence of pain in diabetic neuropathy in addition to the peripheral one. Two hundred seventy-one patients with DPN underwent CPM testing and clinical assessment, including quantitative sensory testing. Two modalities of the test stimuli (heat and pressure) conditioned to cold noxious water were assessed and compared between patients with painful and nonpainful DPN. No significant difference was found between the groups for pressure pain CPM; however, patients with painful DPN demonstrated unexpectedly more efficient CPMHEAT (-7.4 ± 1.0 vs -2.3 ± 1.6; P = 0.008). Efficient CPMHEAT was associated with higher clinical pain experienced in the 24 hours before testing (r = -0.15; P = 0.029) and greater loss of mechanical sensation (r = -0.135; P = 0.042). Moreover, patients who had mechanical hypoesthesia demonstrated more efficient CPMHEAT (P = 0.005). More efficient CPM among patients with painful DPN might result from not only central changes in pain modulation but also from altered sensory messages coming from tested affected body sites. This calls for the use of intact sites for proper assessment of pain modulation in patients with neuropathy.
Collapse
Affiliation(s)
- Yelena Granovsky
- Laboratory of Clinical Neurophysiology, Bruce Rappaport Faculty of Medicine, Technion, Israel
- Department of Neurology, Rambam Health Care Campus, Haifa, Israel
| | - Leah Shafran Topaz
- Laboratory of Clinical Neurophysiology, Bruce Rappaport Faculty of Medicine, Technion, Israel
| | - Helen Laycock
- Pain Research, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Rabab Zubiedat
- Laboratory of Clinical Neurophysiology, Bruce Rappaport Faculty of Medicine, Technion, Israel
| | - Shoshana Crystal
- Laboratory of Clinical Neurophysiology, Bruce Rappaport Faculty of Medicine, Technion, Israel
| | - Chen Buxbaum
- Department of Neurology, Rambam Health Care Campus, Haifa, Israel
| | - Noam Bosak
- Department of Neurology, Rambam Health Care Campus, Haifa, Israel
| | - Rafi Hadad
- Department of Neurology, Rambam Health Care Campus, Haifa, Israel
| | - Erel Domany
- Department of Neurology, Rambam Health Care Campus, Haifa, Israel
| | - Mogher Khamaisi
- Department of Internal Medicine D, Rambam Health Care Campus, Haifa, Israel
- Endocrinology, Diabetes, and Metabolism Institute, Rambam Health Care Campus, Haifa, Israel
| | - Elliot Sprecher
- Department of Neurology, Rambam Health Care Campus, Haifa, Israel
| | - David L. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Andrew Rice
- Pain Research, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - David Yarnitsky
- Laboratory of Clinical Neurophysiology, Bruce Rappaport Faculty of Medicine, Technion, Israel
- Department of Neurology, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
21
|
Yuan P, Song F, Zhu P, Fan K, Liao Q, Huang L, Liu Z. Poly (ADP-ribose) polymerase 1-mediated defective mitophagy contributes to painful diabetic neuropathy in the db/db model. J Neurochem 2022; 162:276-289. [PMID: 35263449 DOI: 10.1111/jnc.15606] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 11/29/2022]
Abstract
Studies have shown that poly (ADP-ribose) polymerase 1 (PARP1) was involved in the pathological process of diabetes. Mitophagy is widely acknowledged to be a key regulatory process in maintaining reactive oxygen species homeostasis via lysosome degradation of damaged mitochondria. However, the regulatory role of PARP1 in mitophagy-related mitochondrial oxidative injury and progression of painful diabetic neuropathy (PDN) is unclear. In this study, we studied the in vitro and in vivo mechanisms of PARP1-mediated mitophagy blockade in a leptin gene-mutation (db/db) mouse model of PDN. Db/db mice models of PDN were established by assessing the sciatic nerve conduction velocity (SNCV), mechanical withdrawal threshold (MWT), and thermal withdrawal latency (TWL). The results showed that PARP1 activity and mitochondrial injury of dorsal root ganglion (DRG) neurons were increased, and mitophagy was impaired in PDN mice. PARP1 was found to mediate the impairment of mitophagy in DRG neurons isolated from PDN mice. PARP1 inhibitors (PJ34 or AG14361) attenuated diabetes-induced peripheral nerve hyperalgesia, restored DRG neuron mitophagy function and decreased mitochondrial oxidative injury. Mitophagy impairment induced by lysosome deacidificant (DC661) aggravated diabetes-induced DRG neuron mitochondrial oxidative stress and injury. Taken together, our data revealed that PARP1 induced defective mitophagy of DRG neurons is a key mechanism in diabetes-induced peripheral neuropathic injury. Inhibition of PARP1 and restoration of mitophagy function are potential therapeutic targets for PDN.
Collapse
Affiliation(s)
- Pengfei Yuan
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Industrial Avenue Central 253, Guangzhou, 510282, Guangdong Province, China
| | - Fuhu Song
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Pian Zhu
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Industrial Avenue Central 253, Guangzhou, 510282, Guangdong Province, China
| | - Keke Fan
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Industrial Avenue Central 253, Guangzhou, 510282, Guangdong Province, China
| | - Qinming Liao
- Department of Neurosurgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Lijin Huang
- Department of Neurosurgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Zhongjie Liu
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Industrial Avenue Central 253, Guangzhou, 510282, Guangdong Province, China
| |
Collapse
|
22
|
Liau YJ, Lin SF, Lee IT. Scores of peripheral neuropathic pain predicting long-term mortality in patients with type 2 diabetes: A retrospective cohort study. Front Endocrinol (Lausanne) 2022; 13:969149. [PMID: 36051389 PMCID: PMC9424503 DOI: 10.3389/fendo.2022.969149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Diabetic peripheral neuropathic pain (DPNP) is a prevalent chronic complication in patients with diabetes. Using a questionnaire is helpful for DPNP screening in outpatients. In this retrospective cohort, we aimed to examine whether DPNP diagnosed based on scoring questionnaires could predict long-term mortality in outpatients with type 2 diabetes. METHODS We enrolled 2318 patients who had joined the diabetes pay-for-performance program and completed the annual assessments, including both the identification pain questionnaire (ID pain) and Douleur Neuropathique en 4 questionnaire (DN4), between January 2013 and October 2013. Information on registered deaths was collected up to August 2019. RESULTS There was high consistency in the scores between the ID pain and DN4 (r = 0.935, P < 0.001). During the median follow-up of 6.2 years (interquartile range: 5.9-6.4 years), 312 patients deceased. Patients with an ID pain score of ≥ 2 had a higher mortality risk than those with a score of < 2 (hazard ratio [HR] = 1.394, 95%CI: 1.090-1.782), and patients with a DN4 score of ≥ 4 had a higher mortality risk than those with a score of < 4 (HR = 1.668, 95% confidence interval [CI]: 1.211-2.297). Patients consistently diagnosed with DPNP by the ID pain and DN4 had a significantly higher mortality risk (HR = 1.713, 95% CI: 1.223-2.398, P = 0.002), but not those discrepantly diagnosed with DPNP (P = 0.107), as compared with those without DPNP. CONCLUSIONS Both the ID pain and DN4 for DPNP screening were predictive of long-term mortality in patients with type 2 diabetes. However, a discrepancy in the diagnosis of DPNP weakened the power of mortality prediction.
Collapse
Affiliation(s)
- Yi-Ju Liau
- Department of Psychiatry, Jen-Ai Hospital, Taichung, Taiwan
| | - Shu-Fan Lin
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - I-Te Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- *Correspondence: I-Te Lee,
| |
Collapse
|
23
|
Galosi E, Hu X, Michael N, Nyengaard JR, Truini A, Karlsson P. Redefining distal symmetrical polyneuropathy features in type 1 diabetes: a systematic review. Acta Diabetol 2022; 59:1-19. [PMID: 34213655 PMCID: PMC8758619 DOI: 10.1007/s00592-021-01767-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022]
Abstract
Diabetic neuropathy is among the most frequent complications of both type 1 (T1DM) and type 2 diabetes (T2DM) and commonly manifests as a distal symmetrical polyneuropathy (DSPN). Despite evidence that T1DM- and T2DM-related DSPN are separate entities, most of our knowledge on diabetic DSPN derives from studies focused on type 2 diabetes. This systematic review provides an overview of current evidence on DSPN in T1DM, including its epidemiological, pathophysiological and clinical features, along with principal diagnostic tests findings. This review included 182 clinical and preclinical studies. The results indicate that DSPN is a less frequent complication in T1DM compared with T2DM and that distinctive pathophysiological mechanisms underlie T1DM-related DSPN development, with hyperglycemia as a major determinant. T1DM-related DSPN more frequently manifests with non-painful than painful symptoms, with lower neuropathic pain prevalence compared with T2DM-associated DSPN. The overt clinical picture seems characterized by a higher prevalence of large fiber-related clinical signs (e.g., ankle reflexes reduction and vibration hypoesthesia) and to a lesser extent small fiber damage (e.g., thermal or pinprick hypoesthesia). These findings as a whole suggest that large fibers impairment plays a dominant role in the clinical picture of symptomatic T1DM-related DSPN. Nevertheless, small fiber diagnostic testing shows high diagnostic accuracy in detecting early nerve damage and may be an appropriate diagnostic tool for disease monitoring and screening.
Collapse
Affiliation(s)
- Eleonora Galosi
- Department of Human Neuroscience, Sapienza University, Rome, Italy.
| | - Xiaoli Hu
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Aarhus University, Aarhus, Denmark
| | - Nivatha Michael
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jens Randel Nyengaard
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Aarhus University, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Andrea Truini
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | - Páll Karlsson
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Aarhus University, Aarhus, Denmark
- Department of Clinical Medicine, Danish Pain Research Center, Aarhus University, Aarhus, Denmark
| |
Collapse
|
24
|
Nemenov MI, Singleton JR, Premkumar LS. Role of Mechanoinsensitive Nociceptors in Painful Diabetic Peripheral Neuropathy. Curr Diabetes Rev 2022; 18:e081221198649. [PMID: 34879806 DOI: 10.2174/1573399818666211208101555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/08/2021] [Accepted: 07/09/2021] [Indexed: 11/22/2022]
Abstract
The cutaneous mechanisms that trigger spontaneous neuropathic pain in diabetic peripheral neuropathy (PDPN) are far from clear. Two types of nociceptors are found within the epidermal and dermal skin layers. Small-diameter lightly myelinated Aδ and unmyelinated C cutaneous mechano and heat-sensitive (AMH and CMH) and C mechanoinsensitive (CMi) nociceptors transmit pain from the periphery to central nervous system. AMH and CMH fibers are mainly located in the epidermis, and CMi fibers are distributed in the dermis. In DPN, dying back intra-epidermal AMH and CMH fibers leads to reduced pain sensitivity, and the patients exhibit significantly increased pain thresholds to acute pain when tested using traditional methods. The role of CMi fibers in painful neuropathies has not been fully explored. Microneurography has been the only tool to access CMi fibers and differentiate AMH, CMH, and CMi fiber types. Due to the complexity, its use is impractical in clinical settings. In contrast, a newly developed diode laser fiber selective stimulation (DLss) technique allows to safely and selectively stimulate Aδ and C fibers in the superficial and deep skin layers. DLss data demonstrate that patients with painful DPN have increased Aδ fiber pain thresholds, while C-fiber thresholds are intact because, in these patients, CMi fibers are abnormally spontaneously active. It is also possible to determine the involvement of CMi fibers by measuring the area of DLss-induced neurogenic axon reflex flare. The differences in AMH, CMH, and CMi fibers identify patients with painful and painless neuropathy. In this review, we will discuss the role of CMi fibers in PDPN.
Collapse
Affiliation(s)
- Mikhail I Nemenov
- Department of Anesthesia, Stanford University, Palo Alto, CA, USA
- Lasmed LLC, Mountain View, CA, USA
| | | | - Louis S Premkumar
- Department of Pharmacology, SIU School of Medicine, Springfield, Illinois, USA and Ion Channel Pharmacology LLC, Springfield, IL, USA
| |
Collapse
|
25
|
Jin Y, Wang W, Chen W, Guo S, Li C, Zhu D, Bi Y. Corneal confocal microscopy: A useful tool for diagnosis of small fiber neuropathy in type 2 diabetes. J Diabetes Investig 2021; 12:2183-2189. [PMID: 34134175 PMCID: PMC8668068 DOI: 10.1111/jdi.13616] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/08/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022] Open
Abstract
AIM To investigate the diagnostic utility of corneal confocal microscopy (CCM) for small fiber neuropathy in type 2 diabetes. MATERIALS AND METHODS There were 186 participants with type 2 diabetes enrolled in this cross-sectional research. Pure small fiber neuropathy and mixed fiber neuropathy were defined using clinical examination, electromyography, and quantitative sensory testing. Demographics and clinical data, corneal confocal microscopy parameters, and other neuropathy measures were compared among the groups. The diagnostic utility of corneal confocal microscopy for small fiber neuropathy was assessed by the receiver operating curve. RESULTS Of the 186 patients, 24.7% had a pure small fiber neuropathy and 17.2% of patients were diagnosed with mixed fiber neuropathy. The corneal nerve fiber density (CNFD), corneal nerve fiber branch density (CNBD), and corneal nerve fiber length (CNFL) were significantly lower in subjects with pure small fiber neuropathy compared with those without diabetic peripheral neuropathy (all P < 0.05). The receiver operating curve analysis for corneal confocal microscopy diagnosing small fiber neuropathy demonstrated the area under the curve for CNFD of 0.791, CNFL of 0.778, CNBD of 0.710. CONCLUSIONS Patients with type 2 diabetes with pure small fiber neuropathy showed more corneal nerve loss compared with those without diabetic peripheral neuropathy. It was revealed that corneal confocal microscopy can be a reasonable marker in the diagnosis of small fiber neuropathy in type 2 diabetes.
Collapse
Affiliation(s)
- Yu Jin
- Department of EndocrinologyNanjing Drum Tower Hospital Clinical College of Nanjing Medical UniversityNanjingChina
- Department of EndocrinologyDrum Tower Hospital Affiliated to Nanjing University Medical SchoolNanjingChina
- Department of OsteoporosisLianyungang Clinical College of Nanjing Medical UniversityLianyungangChina
| | - Weimin Wang
- Department of EndocrinologyDrum Tower Hospital Affiliated to Nanjing University Medical SchoolNanjingChina
| | - Wei Chen
- Department of EndocrinologyDrum Tower Hospital Affiliated to Nanjing University Medical SchoolNanjingChina
| | - Simin Guo
- Department of EndocrinologyDrum Tower Hospital Affiliated to Nanjing University Medical SchoolNanjingChina
| | - Chenxi Li
- Department of EndocrinologyDrum Tower Hospital Affiliated to Nanjing University Medical SchoolNanjingChina
| | - Dalong Zhu
- Department of EndocrinologyDrum Tower Hospital Affiliated to Nanjing University Medical SchoolNanjingChina
| | - Yan Bi
- Department of EndocrinologyNanjing Drum Tower Hospital Clinical College of Nanjing Medical UniversityNanjingChina
- Department of EndocrinologyDrum Tower Hospital Affiliated to Nanjing University Medical SchoolNanjingChina
| |
Collapse
|
26
|
Zhang Y, Zhang S, Pan L, Wang B, Sun Y, Gao L, Wang L, Cui L, Zhang Q, Shang H, Jin S, Qin X, Geng D, Yu X, Yang L, Li L, Li Z, Yan C, Sun H, Sun T, Du B, Cao J, Hu F, Ma J, Zhou S, Zhao F, Li W, Zheng J, Yi Y, Xu J, Hu B, Sheng B, Li Z, Zhao Z, Yang T, Wang N, Zhao H, Mima D, Qu H, Wang Y, Song F, Li X, Li N, Fan D. Painful Diabetic Peripheral Neuropathy Study of Chinese Outpatients (PDNSCOPE): A Multicentre Cross-Sectional Registry Study of Clinical Characteristics and Treatment in Mainland China. Pain Ther 2021; 10:1355-1373. [PMID: 34363598 PMCID: PMC8586277 DOI: 10.1007/s40122-021-00281-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/10/2021] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION This aim of this study was to delineate current clinical scenarios of painful diabetic peripheral neuropathy (PDN) and associated anxiety and depression among patients in Mainland China, and to report current therapy and clinical practices. METHODS A total of 1547 participants were enrolled in the study between 14 June 2018 and 11 November 2019. Recruitment was conducted using a multilevel sampling method. Participants' demographics, medical histories, glucose parameters, Douleur Neuropathique 4 Questionnaire (DN4) scores, visual analogue scale (VAS) pain scores, Patient Health Questionnaire 9 (PHQ-9) scores, Generalised Anxiety Disorder 7 (GAD-7) scores and therapies were recorded. RESULTS The male-to-female ratio was 1.09:1 (807:740), and the mean age at onset was 61.28 ± 11.23 years. The mean DN4 score (± standard deviation) was 4.91 ± 1.88. The frequencies of DN4 sub-item phenotypes were: numbness, 81%; tingling, 68.71%; pins and needles, 62.90%; burning, 53.59%; hypoaesthesia to touch, 50.16%; electronic shocks, 43.31%; hypoaesthesia to pinprick, 37.94%; brushing, 37.82%; painful cold, 29.61%; and itching, 25.86%. Age, diabetic duration, depression history, PHQ-9 score and GAD-7 score were identified as risk factors for VAS pain score. Peripheral artery disease (PAD) was a protective factor for VAS pain score. For all participants currently diagnosed with PDN and for those previously diagnosed PDN, fasting blood glucose (FBG) was a risk factor for VAS; there was no association between FBG and VAS pain score for PDN diagnosed within 3 months prior to recruitment. Utilisation rate of opium therapies among enrolled participants was 0.71% , contradiction of first-line guideline recommendation for pain relief accounted for 9.43% (33/350) and contradiction of second-line guideline recommendation for opium dosage form was 0.57% (2/350). CONCLUSION Moderate to severe neuropathic pain in PDN was identified in 73.11% of participants. Age, diabetic duration, depression history, PHQ-9 score, GAD-7 score and FBG were risk factors for VAS pain scores. PAD was protective factor. The majority of pain relief therapies prescribed were in accordance with guidelines. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT03520608, retrospectively registered, 2018-05-11.
Collapse
Affiliation(s)
- Yuanjin Zhang
- Department of Neurology, Peking University Third Hospital, 49th North Garden Road, Haidian District, Beijing, China
| | - Shaowei Zhang
- Department of Endocrinology, Shenyang Weikang Hospital, Shenyang, China
| | - Liya Pan
- Department of Neurology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Baojun Wang
- Department of Neurology, Baotou Central Hospital, Baotou, China
| | - Yuanlin Sun
- Department of Neurology, Panjin Central Hospital, Panjin, China
| | - Lijun Gao
- Department of Neurology, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Ling Wang
- Department of Endocrinology, Fukuang General Hospital of Liaoning Health Industry Group, Fushun, China
| | - Lijuan Cui
- Department of Endocrinology, Bengang General Hospital of Liaoning Health Industry Group, Benxi, China
| | - Qing Zhang
- Department of Neurology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Heng Shang
- Department of Gastroenterology, Cangzhou People's Hospital, Cangzhou, China
| | - Suqin Jin
- Department of Neurology, The Second Hospital of Shandong University, Jinan, China
| | - Xing Qin
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Deqin Geng
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiaorong Yu
- Department of Neurology, Linfen People's Hospital, Linfen, China
| | - Lin Yang
- Department of Neurology, The First Affiliated Hospital of Dali University, Dali, China
| | - Li Li
- Department of Gastroenterology, The First People's Hospital of Taian, Tai'an, China
| | - Zuoxiao Li
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chaoli Yan
- Department of Endocrinology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Hongbin Sun
- Department of Neurology, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu, China
| | - Tao Sun
- Pain Department, Shandong Provincial Hospital, Jinan, China
| | - Baoxin Du
- Department of Neurology, Guangdong Traditional Chinese Medicine Hospital, Guangzhou, China
| | - Junying Cao
- Department of Neurology, Tianjin Gangkou Hospital, Tianjin, China
| | - Fengyun Hu
- Department of Neurology, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Jianhua Ma
- Department of Neurology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Shengnian Zhou
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Fengli Zhao
- Department of Neurology, Yuncheng Central Hospital, Yuncheng, China
| | - Wei Li
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Jianming Zheng
- Department of Neurology, Mindong Hospital of Fujian Medical University, Ningde, China
| | - Yanhui Yi
- Department of Neurology, The Second People's Hospital of Hunan Province, Brain Hospital of Hunan Province, Changsha, China
| | - Jianguo Xu
- Department of Neurology, Guiyang Sixth Hospital, Guiyang, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical Collage of Huazhong University of Science and Technology, Wuhan, China
| | - Baoying Sheng
- Department of Neurology, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Zhaohui Li
- Department of Neurology, People's Hospital of Xinxing Country, Yunfu, China
| | - Zhong Zhao
- Department of Neurology, Suzhou Municipal Hospital, Suzhou, China
| | - Ting Yang
- Department of Neurology, Tianjin Xiqing Hospital, Tianjin, China
| | - Ni Wang
- Department of Neurology, Central Hospital of Wafangdian, Dalian, China
| | - Hongdong Zhao
- Department of Neurology, Nanjing First Hospital, Nanjing, China
| | - Dunzhu Mima
- Department of Neurology, Tibet Autonomous Region People's Hospital, Lhasa, China
| | - Huaiqian Qu
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, China
| | - Yi Wang
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, China
| | - Fuxia Song
- Department of Neurology, Yantai Yuhuangding Hospital, Yantai, China
| | - Xinyi Li
- Department of Neurology, Shanxi Bethune Hospital, Taiyuan, China
| | - Nan Li
- Research Center of Clinical Epidemiology, Peking University Third Hospital, 49th North Garden Road, Haidian District, Beijing, China.
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, 49th North Garden Road, Haidian District, Beijing, China.
| |
Collapse
|
27
|
Stucky CL, Mikesell AR. Cutaneous pain in disorders affecting peripheral nerves. Neurosci Lett 2021; 765:136233. [PMID: 34506882 PMCID: PMC8579816 DOI: 10.1016/j.neulet.2021.136233] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 08/16/2021] [Accepted: 09/02/2021] [Indexed: 02/08/2023]
Abstract
Our ability to quickly detect and respond to harmful environmental stimuli is vital for our safety and survival. This inherent acute pain detection is a "gift" because it both protects our body from harm and allows healing of damaged tissues [1]. Damage to tissues from trauma or disease can result in distorted or amplified nociceptor signaling and sensitization of the spinal cord and brain (Central Nervous System; CNS) pathways to normal input from light touch mechanoreceptors. Together, these processes can result in nagging to unbearable chronic pain and extreme sensitivity to light skin touch (allodynia). Unlike acute protective pain, chronic pain and allodynia serve no useful purpose and can severely reduce the quality of life of an affected person. Chronic pain can arise from impairment to peripheral neurons, a phenomenon called "peripheral neuropathic pain." Peripheral neuropathic pain can be caused by many insults that directly affect peripheral sensory neurons, including mechanical trauma, metabolic imbalance (e.g., diabetes), autoimmune diseases, chemotherapeutic agents, viral infections (e.g., shingles). These insults cause "acquired" neuropathies such as small-fiber neuropathies, diabetic neuropathy, chemotherapy-induced peripheral neuropathy, and post herpetic neuralgia. Peripheral neuropathic pain can also be caused by genetic factors and result in hereditary neuropathies that include Charcot-Marie-Tooth disease, rare channelopathies and Fabry disease. Many acquired and hereditary neuropathies affect the skin, our largest organ and protector of nearly our entire body. Here we review how cutaneous nociception (pain perceived from the skin) is altered following diseases that affect peripheral nerves that innervate the skin. We provide an overview of how noxious stimuli are detected and encoded by molecular transducers on subtypes of cutaneous afferent endings and conveyed to the CNS. Next, we discuss several acquired and hereditary diseases and disorders that cause painful or insensate (lack of sensation) cutaneous peripheral neuropathies, the symptoms and percepts patients experience, and how cutaneous afferents and other peripheral cell types are altered in function in these disorders. We highlight exciting new research areas that implicate non-neuronal skin cells, particularly keratinocytes, in cutaneous nociception and peripheral neuropathies. Finally, we conclude with ideas for innovative new directions, areas of unmet need, and potential opportunities for novel cutaneous therapeutics that may avoid CNS side effects, as well as ideas for improved translation of mechanisms identified in preclinical models to patients.
Collapse
Affiliation(s)
- Cheryl L Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States.
| | - Alexander R Mikesell
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| |
Collapse
|
28
|
Chen X, Dai J, Li D, Huang X, Qu C. Effects of Pulsed Radiofrequency with Different Temperature on Model Rats of Chronic Constriction Injury. PAIN MEDICINE 2021; 22:1612-1618. [PMID: 33620466 DOI: 10.1093/pm/pnab045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVES The treatment for neuropathic pain is still a big challenge. Pulsed radiofrequency technique has been widely used to relieve neuropathic pain in recent years. The purpose of this study is to optimize the temperature for pulsed radiofrequency therapy. DESIGN Animal, experimental study. METHODS Seventy-five male SD rats were randomly divided into five groups: Sham operation group (Sham group), chronic constriction injury group (CCI group), PRF 42°C group (P42 group), PRF 50°C group (P50 group), and PRF 60°C group (P60 group). The hindpaw withdrawal threshold (HWT), paw thermal withdrawal latency (PTWL), sciatic nerve structure, and the concentration of spinal methionine enkephalin(M-ENK) were detected to identify which temperature is the best for PRF treatment. RESULTS PRF at 42°C, 50°C and 60°C significantly alleviated the pain in CCI rats. The therapeutic effects of 50°C and 60°C were similar, and both were better than 42°C. In addition, PRF using 42°C, 50°C, and 60°C mediated nerve injury to sciatic nerve were grade 1, 1, and 2, respectively. The concentration of M-ENK in spinal cord increased accompanying with the increasing of the temperature of PRF. CONCLUSIONS PRF using 50°C could induce less damage while achieving better improvement of mechanical and thermal pain threshold than 42°C and 60°C in CCI rats, which may be achieved by promoting the expression of M-ENK in spinal cord.
Collapse
Affiliation(s)
- Xun Chen
- Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, China.,Department of Anesthesiology Management, Chongqing Nan'an District People's Hospital, Chongqing, China.,Laboratory of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Jianbo Dai
- Department of surgery Management, Chongqing Nan'an District People's Hospital, Chongqing, China
| | - Dan Li
- Department of Stomatology Management, Chongqing Tongnan District People's Hospital, Chongqing, China
| | - Xingliang Huang
- Department of Respiratory and Critical Care Medicine Management, Chongqing Tongnan District People's Hospital, Chongqing, China
| | - Cehua Qu
- Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, China
| |
Collapse
|
29
|
Sloan G, Selvarajah D, Tesfaye S. Pathogenesis, diagnosis and clinical management of diabetic sensorimotor peripheral neuropathy. Nat Rev Endocrinol 2021; 17:400-420. [PMID: 34050323 DOI: 10.1038/s41574-021-00496-z] [Citation(s) in RCA: 222] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/15/2021] [Indexed: 02/08/2023]
Abstract
Diabetic sensorimotor peripheral neuropathy (DSPN) is a serious complication of diabetes mellitus and is associated with increased mortality, lower-limb amputations and distressing painful neuropathic symptoms (painful DSPN). Our understanding of the pathophysiology of the disease has largely been derived from animal models, which have identified key potential mechanisms. However, effective therapies in preclinical models have not translated into clinical trials and we have no universally accepted disease-modifying treatments. Moreover, the condition is generally diagnosed late when irreversible nerve damage has already taken place. Innovative point-of-care devices have great potential to enable the early diagnosis of DSPN when the condition might be more amenable to treatment. The management of painful DSPN remains less than optimal; however, studies suggest that a mechanism-based approach might offer an enhanced benefit in certain pain phenotypes. The management of patients with DSPN involves the control of individualized cardiometabolic targets, a multidisciplinary approach aimed at the prevention and management of foot complications, and the timely diagnosis and management of neuropathic pain. Here, we discuss the latest advances in the mechanisms of DSPN and painful DSPN, originating both from the periphery and the central nervous system, as well as the emerging diagnostics and treatments.
Collapse
Affiliation(s)
- Gordon Sloan
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Dinesh Selvarajah
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
- Department of Oncology and Human Metabolism, University of Sheffield, Sheffield, UK
| | - Solomon Tesfaye
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK.
| |
Collapse
|
30
|
Unal-Cevik I, Orhan D, Acar-Ozen NP, Mamak-Ekinci EB. Small Fiber Functionality in Patients with Diabetic Neuropathic Pain. PAIN MEDICINE 2021; 22:2068-2078. [PMID: 33892490 DOI: 10.1093/pm/pnab150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Diabetic neuropathic pain is associated with small fiber neuropathy. We aimed to assess the functionality of small fibers in patients with diabetes by using a practical method. DESIGN Patients with impaired glucose tolerance (IGT), diabetic neuropathic pain (DNP), type II diabetes mellitus without neuropathic pain, and healthy control were included. Axon-reflex flare responses were induced by the intradermal application of capsaicin and histamine at the distal leg. The associated flare characteristics (flare areas and flare intensities) were recorded by using Laser Speckle Contrast Analysis (LASCA). The pain and itch responses were rated while performing LASCA. To verify the structural properties of the small fibers, proximal and distal skin biopsies were performed. RESULTS DN4, MNSI, NRS, evoked-burning pain scores, and HbA1c levels were the highest in the DNP group. Compatible with length-dependent neuropathy, the distal skin PGP9.5-positive intraepidermal nerve fiber densities (IENFDs) were the lowest, whereas TRPV1-positive IENFDs were the highest in patients with DNP. The distal leg LASCA data showed hypo-functionality in both patients with IGT and DNP and association with disease severity. CONCLUSION There is an unmet need to practically assess the functionality of small fibers in patients with pain. In this study, a practical and objective method that does not need special expertise for the measurement of the functional properties of small fibers by using axon-flare responses is presented. The LASCA method could potentially facilitate a practical, quick (within 5 minutes), and very early diagnosis of small fiber hypo-functionality in both patients with IGT and DNP.
Collapse
Affiliation(s)
- Isin Unal-Cevik
- Hacettepe University Faculty of Medicine, Department of Neurology, Pain Unit
| | - Diclehan Orhan
- Hacettepe University Faculty of Medicine, Department of Pathology
| | - Nazire Pinar Acar-Ozen
- Hacettepe University Faculty of Medicine, Department of Neurology, Pain Unit.,Ataturk Education and Research Hospital, Department of Neurology
| | | |
Collapse
|
31
|
Small-fibre pathology has no impact on somatosensory system function in patients with fibromyalgia. Pain 2021; 161:2385-2393. [PMID: 32897040 DOI: 10.1097/j.pain.0000000000001920] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We aimed to investigate whether small-fibre pathology, a common skin biopsy finding in patients with fibromyalgia, implies clinically important abnormalities of somatosensory system function and verify whether it is associated with voltage-gated sodium channel variants. In 57 consecutively enrolled patients with fibromyalgia, we used skin biopsy to distinguish patients with and without small-fibre pathology. In all patients, we assessed somatosensory system function using quantitative sensory testing (QST) and laser-evoked potentials and investigated voltage-gated sodium channel genotyping. We then compared these variables in patients with and without small-fibre pathology. We found that clinical measures, QST, and laser-evoked potential variables did not differ between patients with and without small-fibre pathology. In most patients with small-fibre pathology, QST and laser-evoked potential variables fell within normative ranges commonly used in clinical practice. Of the 57 patients, one patient without small-fibre pathology and 2 patients with small-fibre pathology had rare variants of voltage-gated sodium channels, namely SCN11A, SCN9A, and SCN1A variants. The SCN9A variant, found in a patient with small-fibre pathology, was an already profiled gain-of-function mutation, previously reported in small-fibre neuropathy. Our findings suggest that small-fibre pathology has a negligible impact on somatosensory system function in fibromyalgia. The genetic analysis suggests that patients with rare small-fibre neuropathy due to voltage-gated sodium channel variants may be misdiagnosed as patients with fibromyalgia.
Collapse
|
32
|
Jensen TS, Karlsson P, Gylfadottir SS, Andersen ST, Bennett DL, Tankisi H, Finnerup NB, Terkelsen AJ, Khan K, Themistocleous AC, Kristensen AG, Itani M, Sindrup SH, Andersen H, Charles M, Feldman EL, Callaghan BC. Painful and non-painful diabetic neuropathy, diagnostic challenges and implications for future management. Brain 2021; 144:1632-1645. [PMID: 33711103 DOI: 10.1093/brain/awab079] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/15/2021] [Accepted: 02/21/2021] [Indexed: 12/12/2022] Open
Abstract
Peripheral neuropathy is one of the most common complications of both type 1 and type 2 diabetes. Up to half of patients with diabetes develop neuropathy during the course of their disease, which is accompanied by neuropathic pain in 30-40% of cases. Peripheral nerve injury in diabetes can manifest as progressive distal symmetric polyneuropathy, autonomic neuropathy, radiculo-plexopathies, and mononeuropathies. The most common diabetic neuropathy is distal symmetric polyneuropathy, which we will refer to as DN, with its characteristic glove and stocking like presentation of distal sensory or motor function loss. DN or its painful counterpart, painful DN, are associated with increased mortality and morbidity; thus, early recognition and preventive measures are essential. Nevertheless, it is not easy to diagnose DN or painful DN, particularly in patients with early and mild neuropathy, and there is currently no single established diagnostic gold standard. The most common diagnostic approach in research is a hierarchical system, which combines symptoms, signs, and a series of confirmatory tests. The general lack of long-term prospective studies has limited the evaluation of the sensitivity and specificity of new morphometric and neurophysiological techniques. Thus, the best paradigm for screening DN and painful DN both in research and in clinical practice remains uncertain. Herein, we review the diagnostic challenges from both clinical and research perspectives and their implications for managing patients with DN. There is no established DN treatment, apart from improved glycaemic control, which is more effective in type 1 than in type 2 diabetes, and only symptomatic management is available for painful DN. Currently, less than one-third of patients with painful DN derive sufficient pain relief with existing pharmacotherapies. A more precise and distinct sensory profile from patients with DN and painful DN may help identify responsive patients to one treatment versus another. Detailed sensory profiles will lead to tailored treatment for patient subgroups with painful DN by matching to novel or established DN pathomechanisms and also for improved clinical trials stratification. Large randomized clinical trials are needed to identify the interventions, i.e. pharmacological, physical, cognitive, educational, etc., which lead to the best therapeutic outcomes.
Collapse
Affiliation(s)
- Troels S Jensen
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark.,Danish Pain Research Center, Aarhus University, Aarhus, Denmark
| | - Pall Karlsson
- Danish Pain Research Center, Aarhus University, Aarhus, Denmark
| | - Sandra S Gylfadottir
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark.,Danish Pain Research Center, Aarhus University, Aarhus, Denmark
| | - Signe T Andersen
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark.,Department of Public Health, Aarhus University, Aarhus, Denmark
| | - David L Bennett
- Nuffield Department of Clinical Neuroscience, Oxford University, Oxford, UK
| | - Hatice Tankisi
- Department of Neurophysiology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Astrid J Terkelsen
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark.,Danish Pain Research Center, Aarhus University, Aarhus, Denmark
| | - Karolina Khan
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | - Mustapha Itani
- Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Søren H Sindrup
- Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Henning Andersen
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Morten Charles
- Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | | |
Collapse
|
33
|
Abstract
Neuropathy is a common complication of long-term diabetes that impairs quality of life by producing pain, sensory loss and limb amputation. The presence of neuropathy in both insulin-deficient (type 1) and insulin resistant (type 2) diabetes along with the slowing of progression of neuropathy by improved glycemic control in type 1 diabetes has caused the majority of preclinical and clinical investigations to focus on hyperglycemia as the initiating pathogenic lesion. Studies in animal models of diabetes have identified multiple plausible mechanisms of glucotoxicity to the nervous system including post-translational modification of proteins by glucose and increased glucose metabolism by aldose reductase, glycolysis and other catabolic pathways. However, it is becoming increasingly apparent that factors not necessarily downstream of hyperglycemia can also contribute to the incidence, progression and severity of neuropathy and neuropathic pain. For example, peripheral nerve contains insulin receptors that transduce the neurotrophic and neurosupportive properties of insulin, independent of systemic glucose regulation, while the detection of neuropathy and neuropathic pain in patients with metabolic syndrome and failure of improved glycemic control to protect against neuropathy in cohorts of type 2 diabetic patients has placed a focus on the pathogenic role of dyslipidemia. This review provides an overview of current understanding of potential initiating lesions for diabetic neuropathy and the multiple downstream mechanisms identified in cell and animal models of diabetes that may contribute to the pathogenesis of diabetic neuropathy and neuropathic pain.
Collapse
|
34
|
Diabetic polyneuropathy and pain, prevalence, and patient characteristics: a cross-sectional questionnaire study of 5,514 patients with recently diagnosed type 2 diabetes. Pain 2021; 161:574-583. [PMID: 31693539 PMCID: PMC7017941 DOI: 10.1097/j.pain.0000000000001744] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Supplemental Digital Content is Available in the Text. Diabetic polyneuropathy and painful diabetic polyneuropathy are frequent in early type 2 diabetes, associated with modifiable risk factors, and have major impact on mental health. Most studies of diabetic polyneuropathy (DPN) and painful DPN are conducted in persons with longstanding diabetes. This cross-sectional study aimed to estimate the prevalence of DPN and painful DPN, important risk factors, and the association with mental health in recently diagnosed type 2 diabetes. A total of 5514 (82%) patients (median diabetes duration 4.6 years) enrolled in the Danish Centre for Strategic Research in Type 2 Diabetes cohort responded to a detailed questionnaire on neuropathy and pain. A score ≥4 on the MNSI questionnaire determined possible DPN, whereas pain presence in both feet together with a score ≥3 on the DN4 questionnaire determined possible painful DPN. The prevalence of possible DPN and possible painful DPN was 18% and 10%, respectively. Female sex, age, diabetes duration, body mass index, and smoking were associated with possible DPN, whereas only smoking showed a clear association with possible painful DPN (odds ratio 1.52 [95% confidence interval: 1.20-1.93]). Possible DPN and painful DPN were independently and additively associated with lower quality of life, poorer sleep, and symptoms of depression and anxiety. Possible DPN itself had greater impact on mental health than neuropathic pain. This large study emphasizes the importance of careful screening for DPN and pain early in the course of type 2 diabetes.
Collapse
|
35
|
Neuropathic Pain Related to Peripheral Neuropathies According to the IASP Grading System Criteria. Brain Sci 2020; 11:brainsci11010001. [PMID: 33374929 PMCID: PMC7821938 DOI: 10.3390/brainsci11010001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/26/2022] Open
Abstract
Neuropathic pain is defined as pain caused by a lesion or disease of the somatosensory system. Neuropathic pain represents a broad category of pain conditions, common complications of peripheral neuropathies, which are characterized by a combination of positive symptoms, including paresthesia and/or dysesthesia and sensory deficits in the painful area. In the present paper, we aimed to assess neuropathic pain frequency and clinical characteristics of peripheral neuropathies due to different aetiologies according to grading system criteria of the International Association for the Study of Pain for a definitive diagnosis of neuropathic pain. Epidemiological studies applying these criteria have been conducted in patients with diabetes, brachial plexus injury, and other traumatic nerve injuries. Neuropathic pain was diagnosed in 37–42% of patients with diabetic peripheral neuropathy, 56% of patients with brachial plexus injury, and 22% of patients with intercostobrachial neuropathy. The most frequent neuropathic pain type was ongoing pain (described as burning or pressing), followed by paroxysmal pain (electric shock-like sensations) and allodynia (pain evoked by brushing and pressure). By providing information on the frequency, clinical signs, and variables associated with neuropathic pain due to different aetiologies, these studies contribute to improving the clinical management of this condition.
Collapse
|
36
|
Itani M, Gylfadottir SS, Krøigård T, Kristensen AG, Christensen DH, Karlsson P, Möller S, Andersen H, Tankisi H, Nielsen JS, Jensen TS, Thomsen RW, Finnerup NB, Sindrup SH. Small and large fiber sensory polyneuropathy in type 2 diabetes: Influence of diagnostic criteria on neuropathy subtypes. J Peripher Nerv Syst 2020; 26:55-65. [PMID: 33295647 DOI: 10.1111/jns.12424] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/16/2020] [Accepted: 12/01/2020] [Indexed: 12/24/2022]
Abstract
Diabetic polyneuropathy (DPN) can be classified based on fiber diameter into three subtypes: small fiber neuropathy (SFN), large fiber neuropathy (LFN), and mixed fiber neuropathy (MFN). We examined the effect of different diagnostic models on the frequency of polyneuropathy subtypes in type 2 diabetes patients with DPN. This study was based on patients from the Danish Center for Strategic Research in Type 2 Diabetes cohort. We defined DPN as probable or definite DPN according to the Toronto Consensus Criteria. DPN was then subtyped according to four distinct diagnostic models. A total of 277 diabetes patients (214 with DPN and 63 with no DPN) were included in the study. We found a considerable variation in polyneuropathy subtypes by applying different diagnostic models independent of the degree of certainty of DPN diagnosis. For probable and definite DPN, the frequency of subtypes across diagnostic models varied from: 1.4% to 13.1% for SFN, 9.3% to 21.5% for LFN, 51.4% to 83.2% for MFN, and 0.5% to 14.5% for non-classifiable neuropathy (NCN). For the definite DPN group, the frequency of subtypes varied from: 1.6% to 13.5% for SFN, 5.6% to 20.6% for LFN, 61.9% to 89.7% for MFN, and 0.0% to 6.3% for NCN. The frequency of polyneuropathy subtypes depends on the type and number of criteria applied in a diagnostic model. Future consensus criteria should clearly define sensory functions to be tested, methods of testing, and how findings should be interpreted for both clinical practice and research purpose.
Collapse
Affiliation(s)
- Mustapha Itani
- Research Unit for Neurology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Sandra Sif Gylfadottir
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Thomas Krøigård
- Research Unit for Neurology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | | | | | - Pall Karlsson
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Core Center for Molecular Morphology, Section for Stereology and Microscopy, Aarhus University, Denmark
| | - Sören Möller
- OPEN-Open Patient data Explorative Network, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Henning Andersen
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Hatice Tankisi
- Department of Clinical Neurophysiology, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Steen Nielsen
- Danish Center for Strategic Research in Type 2 Diabetes, Steno Diabetes Center Odense, Odense, Denmark
| | - Troels Staehelin Jensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Nanna Brix Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Søren Hein Sindrup
- Research Unit for Neurology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
37
|
Maiga Y, Diallo S, Konipo FDN, Sangho O, Sangaré M, Diallo SH, Mahamadou S, Péréon Y, Giumelli B, Coulibaly A, Daou M, Traoré Z, Sow Sylla D, Albakaye M, Guinto CO, Ouologem M, Sissoko AS, Traoré HA, Coulibaly SP, Damier P, Attal N, Nizard J. Diabetic polyneuropathy with/out neuropathic pain in Mali: A cross-sectional study in two reference diabetes treatment centers in Bamako (Mali), Western Africa. PLoS One 2020; 15:e0241387. [PMID: 33166296 PMCID: PMC7652324 DOI: 10.1371/journal.pone.0241387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 10/13/2020] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Diabetic polyneuropathy (DPN) with or without neuropathic pain is a frequent complication of diabetes. This work aimed to determine the prevalence of diabetic polyneuropathy, to describe its epidemiological aspects, and to analyze the therapeutic itinerary of patients with DPN. METHODS This was a cross-sectional, descriptive study performed synchronously over six months at two major follow-up sites for patients with diabetes in Mali. DPN was diagnosed based on the Michigan Neuropathy Screening Instrument (MNSI). The neuropathic nature of the pain and the quality of life of patients were evaluated by the DN4 and the ED-5D scale, respectively. We used three (3) different questionnaires to collect data from patients (one at inclusion and another during the follow-up consultation) and from the caregivers of patients with DPN. RESULTS We included 252 patients with diabetes, and DPN was found to have a healthcare facility-based prevalence of 69.8% (176/252). The sex ratio was approximately three females for every male patient. The patients were mostly 31 to 60 years of age, 83% had type 2 diabetes, and 86.9% had neuropathic pain Approximately half of the patients (48.3%) had autonomic neuropathy and they reported moderate to intense pain, which was mainly described as a burning sensation. The patients exhibited impaired exteroceptive and proprioceptive sensations in 51.7% of cases. The patients smoked tobacco in 3.4% of cases, while 36.6% of the patients were obese and had dyslipidemia. The caregivers clearly indicated that appropriate medications were not readily accessible or available for their patients with DPN. CONCLUSION The healthcare facility-based prevalence of DPN with or without neuropathic pain was high in our cohort. These inexpensive and easy-to-use tools (MNSI, DN4) can be used to adequately diagnose DPN in the African context. In Mali, screening and early treatment of patients at risk of DPN should allow for a reduction of the burden of the disease, while caregivers need to be adequately trained to manage DPN.
Collapse
Affiliation(s)
- Youssoufa Maiga
- Faculty of Medicine, University of Technical Sciences and Technologies, Bamako, Mali
- Department of Neurology, Gabriel Touré Teaching Hospital, Bamako, Mali
- Laboratory of Therapeutics (EA3826), Faculty of Medicine of Nantes, University of Nantes, Nantes, France
| | - Salimata Diallo
- Department of Neurology, Gabriel Touré Teaching Hospital, Bamako, Mali
| | | | - Oumar Sangho
- Faculty of Medicine, University of Technical Sciences and Technologies, Bamako, Mali
| | - Modibo Sangaré
- Faculty of Medicine, University of Technical Sciences and Technologies, Bamako, Mali
| | - Seybou H. Diallo
- Faculty of Medicine, University of Technical Sciences and Technologies, Bamako, Mali
- Department of Neurology, Gabriel Touré Teaching Hospital, Bamako, Mali
| | | | - Yann Péréon
- Reference Center of Neuromuscular Diseases Atlantique-Occitanie-Caraïbes, Hôtel-Dieu, UHC of Nantes, Nantes, France
| | | | - Awa Coulibaly
- Department of Neurology, Gabriel Touré Teaching Hospital, Bamako, Mali
| | - Mariam Daou
- Department of Neurology, Gabriel Touré Teaching Hospital, Bamako, Mali
| | | | - Djeneba Sow Sylla
- Faculty of Medicine, University of Technical Sciences and Technologies, Bamako, Mali
- Center for Combating Diabetes in Mali (CCD), Bamako, Mali
| | - Mohamed Albakaye
- Department of Neurology, Gabriel Touré Teaching Hospital, Bamako, Mali
| | - Cheick Oumar Guinto
- Faculty of Medicine, University of Technical Sciences and Technologies, Bamako, Mali
| | - Madani Ouologem
- Faculty of Dental Surgery, University of Nantes, Nantes, France
| | - Adama S. Sissoko
- Faculty of Medicine, University of Technical Sciences and Technologies, Bamako, Mali
| | - Hamar A. Traoré
- Faculty of Medicine, University of Technical Sciences and Technologies, Bamako, Mali
| | | | | | - Nadine Attal
- INSERM U 98, CET, CHU Ambroise Paré, 92100 Boulogne-Billancourt, France
| | - Julien Nizard
- Laboratory of Therapeutics (EA3826), Faculty of Medicine of Nantes, University of Nantes, Nantes, France
- Faculty of Medicine, University of Nantes, Nantes, France
- Federal Pain Palliative Care and Support, Laboratory of Therapeutics, Nantes UHC, Nantes, France
| |
Collapse
|
38
|
Galosi E, Di Pietro G, La Cesa S, Di Stefano G, Leone C, Fasolino A, Di Lionardo A, Leonetti F, Buzzetti R, Mollica C, Cruccu G, Truini A. Differential involvement of myelinated and unmyelinated nerve fibers in painful diabetic polyneuropathy. Muscle Nerve 2020; 63:68-74. [PMID: 32996600 DOI: 10.1002/mus.27080] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND We aimed at evaluating the differential involvement of large myelinated Aβ-, small myelinated Aδ-, and unmyelinated C-fibers in patients with diabetic polyneuropathy and how they contribute to neuropathic pain. METHODS We collected clinical and diagnostic test variables in 133 consecutive patients with diabetic polyneuropathy. All patients underwent Aβ-fiber mediated nerve conduction study, Aδ-fiber mediated laser-evoked potentials and skin biopsy mainly assessing unmyelinated C-fibers. RESULTS Pure large-fiber and small-fiber polyneuropathy were relatively uncommon; conversely mixed-fiber polyneuropathy was the most common type of diabetic polyneuropathy (74%). The frequency of neuropathic pain was similar in the three different polyneuropathies. Ongoing burning pain and dynamic mechanical allodynia were similarly associated with specific small-fiber related variables. CONCLUSIONS Diabetic polyneuropathy mainly manifests as a mixed-fiber polyneuropathy, simultaneously involving Aβ-, Aδ-, and C-fibers. In most patients, neuropathic pain is distinctly associated with small-fiber damage. The evidence that the frequency of neuropathic pain does not differ across pure large-, pure small-, and mixed-fiber polyneuropathy, raises the possibility that in patients with pure large-fiber polyneuropathy nociceptive nerve terminal involvement might be undetected by standard diagnostic techniques.
Collapse
Affiliation(s)
- Eleonora Galosi
- Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Di Pietro
- Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy
| | - Silvia La Cesa
- Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy
| | - Giulia Di Stefano
- Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy
| | - Caterina Leone
- Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy
| | | | - Andrea Di Lionardo
- Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy
| | - Frida Leonetti
- Diabetes Unit, Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Raffaella Buzzetti
- Experimental Medicine Department, Sapienza University of Rome, Rome, Italy
| | - Cristina Mollica
- Dipartimento di Metodi e Modelli per l'Economia, il Territorio e la Finanza, Sapienza University of Rome, Rome, Italy
| | - Giorgio Cruccu
- Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy
| | - Andrea Truini
- Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
39
|
Ständer S, Schmelz M. [Neuropathic pruritus]. Schmerz 2020; 34:525-535. [PMID: 33025226 DOI: 10.1007/s00482-020-00502-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 11/26/2022]
Abstract
In the past 10 years specific pathways for pruritus have been characterized on a cellular and molecular level but their exact role in the pathophysiology of neuropathic pruritus remains unclear. This also applies to the question which of the competing theories for pruritus, e.g. specificity, temporal/spatial pattern or intensity, would best apply. While experimental trials on mice have mostly confirmed the theory of specificity, the results on humans indicate a role of spatial and temporal patterns. The skin innervation is greatly reduced by the neuropathy and could provide a "spatial contrast pattern" and the axotomy could induce a de novo expression of gastrin-releasing peptide (GRP) in primarily afferent nociceptors and thus modulate spinal pruritus processing. In addition, the overlap of pruritus and pain in neuropathy patients complicates the direct translation from animal experiments and requires collaboration at the clinical level between pain medicine and dermatology.
Collapse
Affiliation(s)
- Sonja Ständer
- Kompetenzzentrum Chronischer Pruritus, Universitätsklinikum Münster, Münster, Deutschland
| | - Martin Schmelz
- Experimentelle Schmerzforschung, MCTN, Medizinische Fakultät Mannheim, Universität Heidelberg, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Deutschland.
| |
Collapse
|
40
|
Gylfadottir SS, Itani M, Krøigård T, Kristensen AG, Christensen DH, Nicolaisen SK, Karlsson P, Callaghan BC, Bennett DL, Andersen H, Tankisi H, Nielsen JS, Andersen NT, Jensen TS, Thomsen RW, Sindrup SH, Finnerup NB. Diagnosis and prevalence of diabetic polyneuropathy: a cross-sectional study of Danish patients with type 2 diabetes. Eur J Neurol 2020; 27:2575-2585. [PMID: 32909392 DOI: 10.1111/ene.14469] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/29/2020] [Indexed: 01/16/2023]
Abstract
BACKGROUND AND PURPOSE Diabetic polyneuropathy (DPN) is a common complication of diabetes. Using the Toronto criteria for diabetic polyneuropathy and the grading system for neuropathic pain, the performance of neuropathy scales and questionnaires were assessed by comparing them to a clinical gold standard diagnosis of DPN and painful DPN in a cohort of patients with recently diagnosed type 2 diabetes. METHODS A questionnaire on neuropathy and pain was sent to a cohort of 5514 Danish type 2 diabetes patients. A sample of 389 patients underwent a detailed clinical examination and completed neuropathy questionnaires and scales. RESULTS Of the 389 patients with a median diabetes duration of 5.9 years, 126 had definite DPN (including 53 with painful DPN), 88 had probable DPN and 53 had possible DPN. There were 49 patients with other causes of polyneuropathy, neuropathy symptoms or pain, 10 with subclinical DPN and 63 without DPN. The sensitivity of the Michigan Neuropathy Screening Instrument questionnaire to detect DPN was 25.7% and the specificity 84.6%. The sensitivity of the Toronto Clinical Neuropathy Scoring System, including questionnaire and clinical examination, was 62.9% and the specificity was 74.6%. CONCLUSIONS Diabetic polyneuropathy affects approximately one in five Danish patients with recently diagnosed type 2 diabetes but neuropathic pain is not as common as previously reported. Neuropathy scales with clinical examination perform better compared with questionnaires alone, but better scales are needed for future epidemiological studies.
Collapse
Affiliation(s)
- S S Gylfadottir
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - M Itani
- Department of Neurology, Odense University Hospital, Odense, Denmark
| | - T Krøigård
- Department of Neurology, Odense University Hospital, Odense, Denmark
| | - A G Kristensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Neurophysiology, Aarhus University Hospital, Aarhus, Denmark
| | - D H Christensen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - S K Nicolaisen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - P Karlsson
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Aarhus University, Aarhus, Denmark
| | - B C Callaghan
- Department of Neurology, University of Michigan, Ann Arbor,, MI, USA
| | - D L Bennett
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - H Andersen
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - H Tankisi
- Department of Clinical Neurophysiology, Aarhus University Hospital, Aarhus, Denmark
| | - J S Nielsen
- Danish Centre for Strategic Research in Type 2 Diabetes, Steno Diabetes Center, Odense, Denmark
| | - N T Andersen
- Biostatistics, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - T S Jensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - R W Thomsen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - S H Sindrup
- Department of Neurology, Odense University Hospital, Odense, Denmark
| | - N B Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
41
|
Sierra-Silvestre E, Somerville M, Bisset L, Coppieters MW. Altered pain processing in patients with type 1 and 2 diabetes: systematic review and meta-analysis of pain detection thresholds and pain modulation mechanisms. BMJ Open Diabetes Res Care 2020; 8:8/1/e001566. [PMID: 32868312 PMCID: PMC7462232 DOI: 10.1136/bmjdrc-2020-001566] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/24/2020] [Accepted: 07/06/2020] [Indexed: 12/15/2022] Open
Abstract
The first signs of diabetic neuropathy typically result from small-diameter nerve fiber dysfunction. This review synthesized the evidence for small-diameter nerve fiber neuropathy measured via quantitative sensory testing (QST) in patients with diabetes with and without painful and non-painful neuropathies. Electronic databases were searched to identify studies in patients with diabetes with at least one QST measure reflecting small-diameter nerve fiber function (thermal or electrical pain detection threshold, contact heat-evoked potentials, temporal summation or conditioned pain modulation). Four groups were compared: patients with diabetes (1) without neuropathy, (2) with non-painful diabetic neuropathy, (3) with painful diabetic neuropathy and (4) healthy individuals. Recommended methods were used for article identification, selection, risk of bias assessment, data extraction and analysis. For the meta-analyses, data were pooled using random-effect models. Twenty-seven studies with 2422 participants met selection criteria; 18 studies were included in the meta-analysis. Patients with diabetes without symptoms of neuropathy already showed loss of nerve function for heat (standardized mean difference (SMD): 0.52, p<0.001), cold (SMD: -0.71, p=0.01) and electrical pain thresholds (SMD: 1.26, p=0.01). Patients with non-painful neuropathy had greater loss of function in heat pain threshold (SMD: 0.75, p=0.01) and electrical stimuli (SMD: 0.55, p=0.03) compared with patients with diabetes without neuropathy. Patients with painful diabetic neuropathy exhibited a greater loss of function in heat pain threshold (SMD: 0.55, p=0.005) compared with patients with non-painful diabetic neuropathy. Small-diameter nerve fiber function deteriorates progressively in patients with diabetes. Because the dysfunction is already present before symptoms occur, early detection is possible, which may assist in prevention and effective management of diabetic neuropathy.
Collapse
Affiliation(s)
- Eva Sierra-Silvestre
- School of Allied Health Sciences, Griffith University, Gold Coast, Queensland, Australia
- Faculty of Human Movement Sciences, Free University Amsterdam, Amsterdam, The Netherlands
| | - Mari Somerville
- School of Allied Health Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Leanne Bisset
- School of Allied Health Sciences, Griffith University, Gold Coast, Queensland, Australia
- Menzies Health Institute Queensland, Griffith University, Nathan, Queensland, Australia
| | - Michel W Coppieters
- Menzies Health Institute Queensland, Griffith University, Nathan, Queensland, Australia
- Amsterdam Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
42
|
Abstract
Neuropathic pain caused by a lesion or disease of the somatosensory nervous system is a common chronic pain condition with major impact on quality of life. Examples include trigeminal neuralgia, painful polyneuropathy, postherpetic neuralgia, and central poststroke pain. Most patients complain of an ongoing or intermittent spontaneous pain of, for example, burning, pricking, squeezing quality, which may be accompanied by evoked pain, particular to light touch and cold. Ectopic activity in, for example, nerve-end neuroma, compressed nerves or nerve roots, dorsal root ganglia, and the thalamus may in different conditions underlie the spontaneous pain. Evoked pain may spread to neighboring areas, and the underlying pathophysiology involves peripheral and central sensitization. Maladaptive structural changes and a number of cell-cell interactions and molecular signaling underlie the sensitization of nociceptive pathways. These include alteration in ion channels, activation of immune cells, glial-derived mediators, and epigenetic regulation. The major classes of therapeutics include drugs acting on α2δ subunits of calcium channels, sodium channels, and descending modulatory inhibitory pathways.
Collapse
Affiliation(s)
- Nanna Brix Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Rohini Kuner
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Troels Staehelin Jensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
43
|
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
|
44
|
Rosenberger DC, Blechschmidt V, Timmerman H, Wolff A, Treede RD. Challenges of neuropathic pain: focus on diabetic neuropathy. J Neural Transm (Vienna) 2020; 127:589-624. [PMID: 32036431 PMCID: PMC7148276 DOI: 10.1007/s00702-020-02145-7] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/19/2020] [Indexed: 02/07/2023]
Abstract
Neuropathic pain is a frequent condition caused by a lesion or disease of the central or peripheral somatosensory nervous system. A frequent cause of peripheral neuropathic pain is diabetic neuropathy. Its complex pathophysiology is not yet fully elucidated, which contributes to underassessment and undertreatment. A mechanism-based treatment of painful diabetic neuropathy is challenging but phenotype-based stratification might be a way to develop individualized therapeutic concepts. Our goal is to review current knowledge of the pathophysiology of peripheral neuropathic pain, particularly painful diabetic neuropathy. We discuss state-of-the-art clinical assessment, validity of diagnostic and screening tools, and recommendations for the management of diabetic neuropathic pain including approaches towards personalized pain management. We also propose a research agenda for translational research including patient stratification for clinical trials and improved preclinical models in relation to current knowledge of underlying mechanisms.
Collapse
Affiliation(s)
- Daniela C Rosenberger
- Department of Neurophysiology, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Vivian Blechschmidt
- Department of Neurophysiology, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Hans Timmerman
- Department of Anesthesiology, Pain Center, University Medical Center of Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - André Wolff
- Department of Anesthesiology, Pain Center, University Medical Center of Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Rolf-Detlef Treede
- Department of Neurophysiology, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
45
|
Yang H, Sloan G, Ye Y, Wang S, Duan B, Tesfaye S, Gao L. New Perspective in Diabetic Neuropathy: From the Periphery to the Brain, a Call for Early Detection, and Precision Medicine. Front Endocrinol (Lausanne) 2020; 10:929. [PMID: 32010062 PMCID: PMC6978915 DOI: 10.3389/fendo.2019.00929] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022] Open
Abstract
Diabetic peripheral neuropathy (DPN) is a common chronic complication of diabetes mellitus. It leads to distressing and expensive clinical sequelae such as foot ulceration, leg amputation, and neuropathic pain (painful-DPN). Unfortunately, DPN is often diagnosed late when irreversible nerve injury has occurred and its first presentation may be with a diabetic foot ulcer. Several novel diagnostic techniques are available which may supplement clinical assessment and aid the early detection of DPN. Moreover, treatments for DPN and painful-DPN are limited. Only tight glucose control in type 1 diabetes has robust evidence in reducing the risk of developing DPN. However, neither glucose control nor pathogenetic treatments are effective in painful-DPN and symptomatic treatments are often inadequate. It has recently been hypothesized that using various patient characteristics it may be possible to stratify individuals and assign them targeted therapies to produce better pain relief. We review the diagnostic techniques which may aid the early detection of DPN in the clinical and research environment, and recent advances in precision medicine techniques for the treatment of painful-DPN.
Collapse
Affiliation(s)
- Heng Yang
- Endocrinology Department, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gordon Sloan
- Diabetes Research Unit, Sheffield Teaching Hospitals, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Yingchun Ye
- Endocrinology Department, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shuo Wang
- Endocrinology Department, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bihan Duan
- Endocrinology Department, Renmin Hospital of Wuhan University, Wuhan, China
| | - Solomon Tesfaye
- Diabetes Research Unit, Sheffield Teaching Hospitals, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Ling Gao
- Endocrinology Department, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
46
|
Püttgen S, Bönhof GJ, Strom A, Müssig K, Szendroedi J, Roden M, Ziegler D. Augmented Corneal Nerve Fiber Branching in Painful Compared With Painless Diabetic Neuropathy. J Clin Endocrinol Metab 2019; 104:6220-6228. [PMID: 31390004 DOI: 10.1210/jc.2019-01072] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 08/01/2019] [Indexed: 02/13/2023]
Abstract
CONTEXT The factors that determine the development of diabetic sensorimotor polyneuropathy (DSPN) as a painful or painless entity are unknown. OBJECTIVE We hypothesized that corneal nerve pathology could be more pronounced in painful DSPN, indicating predominant small nerve fiber damage. DESIGN AND METHODS In this cross-sectional study, we assessed 53 patients with painful DSPN, 63 with painless DSPN, and 46 glucose-tolerant volunteers by corneal confocal microscopy (CCM), nerve conduction (NC), and quantitative sensory testing. DSPN was diagnosed according to modified Toronto Consensus criteria. A cutoff at 4 points on the 11-point rating scale was used to differentiate between painful and painless DSPN. RESULTS After adjustment for age, sex, body mass index, and smoking, corneal nerve fiber density, corneal nerve fiber length, and corneal nerve branch density (CNBD) were reduced in both DSPN types compared with the control group (P < 0.05). Only CNBD differed between the groups; it was greater in patients with painful DSPN compared with those with painless DSPN [55.8 (SD, 29.9) vs 43.8 (SD, 28.3) branches/mm2; P < 0.05]. Several CCM measures were associated with NC and cold perception threshold in patients with painless DSPN (P < 0.05) but not those with painful DSPN. CONCLUSION Despite a similarly pronounced peripheral nerve dysfunction and corneal nerve fiber loss in patients with painful and painless DSPN, corneal nerve branching was enhanced in those with painful DSPN, pointing to some susceptibility of corneal nerve fibers toward regeneration in this entity, albeit possibly not to a sufficient degree.
Collapse
Affiliation(s)
- Sonja Püttgen
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at 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
| | - Alexander Strom
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Karsten Müssig
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Julia Szendroedi
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, 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, München-Neuherberg, 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, München-Neuherberg, Germany
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
47
|
Gylfadottir SS, Weeracharoenkul D, Andersen ST, Niruthisard S, Suwanwalaikorn S, Jensen TS. Painful and non-painful diabetic polyneuropathy: Clinical characteristics and diagnostic issues. J Diabetes Investig 2019; 10:1148-1157. [PMID: 31222961 PMCID: PMC6717899 DOI: 10.1111/jdi.13105] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/06/2019] [Accepted: 06/17/2019] [Indexed: 12/17/2022] Open
Abstract
Diabetic neuropathy (DN) is a common complication of diabetes and can be either painful or non-painful. It is challenging to diagnose this complication, as no biomarker or clear consensus on the clinical definition of either painful or non-painful DN exists. Hence, a hierarchical classification has been developed categorizing the probability of the diagnosis into: possible, probable or definite, based on the clinical presentation of symptoms and signs. Pain is a warning signal of tissue damage, and non-painful DN therefore represents a clinical and diagnostic challenge because it often goes unnoticed until irreversible nerve damage has occurred. Simple clinical tests seem to be the best for evaluation of DN in the general care for diabetes. Screening programs at regular intervals might be the most optimal strategy for early detection and interventions to possibly prevent further neuronal damage and to lower the economic burden of this complication.
Collapse
Affiliation(s)
| | - Danita Weeracharoenkul
- Pain Management Research UnitDepartment of AnesthesiologyFaculty of MedicineKing Chulalongkorn Memorial HospitalChulalongkorn UniversityBangkokThailand
| | - Signe Toft Andersen
- Danish Pain Research CenterAarhus UniversityAarhusDenmark
- Department of Public HealthAarhus UniversityAarhusDenmark
| | - Supranee Niruthisard
- Pain Management Research UnitDepartment of AnesthesiologyFaculty of MedicineKing Chulalongkorn Memorial HospitalChulalongkorn UniversityBangkokThailand
| | - Sompongse Suwanwalaikorn
- Department of MedicineFaculty of MedicineKing Chulalongkorn Memorial HospitalChulalongkorn UniversityBangkokThailand
| | - Troels Staehelin Jensen
- Danish Pain Research CenterAarhus UniversityAarhusDenmark
- Department of NeurologyAarhus University HospitalAarhusDenmark
| |
Collapse
|
48
|
Abstract
Sensory polyneuropathies, which are caused by dysfunction of peripheral sensory nerve fibers, are a heterogeneous group of disorders that range from the common diabetic neuropathy to the rare sensory neuronopathies. The presenting symptoms, acuity, time course, severity, and subsequent morbidity vary and depend on the type of fiber that is affected and the underlying cause. Damage to small thinly myelinated and unmyelinated nerve fibers results in neuropathic pain, whereas damage to large myelinated sensory afferents results in proprioceptive deficits and ataxia. The causes of these disorders are diverse and include metabolic, toxic, infectious, inflammatory, autoimmune, and genetic conditions. Idiopathic sensory polyneuropathies are common although they should be considered a diagnosis of exclusion. The diagnostic evaluation involves electrophysiologic testing including nerve conduction studies, histopathologic analysis of nerve tissue, serum studies, and sometimes autonomic testing and cerebrospinal fluid analysis. The treatment of these diseases depends on the underlying cause and may include immunotherapy, mitigation of risk factors, symptomatic treatment, and gene therapy, such as the recently developed RNA interference and antisense oligonucleotide therapies for transthyretin familial amyloid polyneuropathy. Many of these disorders have no directed treatment, in which case management remains symptomatic and supportive. More research is needed into the underlying pathophysiology of nerve damage in these polyneuropathies to guide advances in treatment.
Collapse
Affiliation(s)
- Kelly Graham Gwathmey
- Virginia Commonwealth University, Department of Neurology, 1101 E. Marshall Street, PO Box 980599, Richmond, VA 23298, USA
| | - Kathleen T Pearson
- Virginia Commonwealth University, Department of Neurology, 1101 E. Marshall Street, PO Box 980599, Richmond, VA 23298, USA
| |
Collapse
|
49
|
Shillo P, Sloan G, Greig M, Hunt L, Selvarajah D, Elliott J, Gandhi R, Wilkinson ID, Tesfaye S. Painful and Painless Diabetic Neuropathies: What Is the Difference? Curr Diab Rep 2019; 19:32. [PMID: 31065863 PMCID: PMC6505492 DOI: 10.1007/s11892-019-1150-5] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW The prevalence of diabetes mellitus and its chronic complications are increasing to epidemic proportions. This will unfortunately result in massive increases in diabetic distal symmetrical polyneuropathy (DPN) and its troublesome sequelae, including disabling neuropathic pain (painful-DPN), which affects around 25% of patients with diabetes. Why these patients develop neuropathic pain, while others with a similar degree of neuropathy do not, is not clearly understood. This review will look at recent advances that may shed some light on the differences between painful and painless-DPN. RECENT FINDINGS Gender, clinical pain phenotyping, serum biomarkers, brain imaging, genetics, and skin biopsy findings have been reported to differentiate painful- from painless-DPN. Painful-DPN seems to be associated with female gender and small fiber dysfunction. Moreover, recent brain imaging studies have found neuropathic pain signatures within the central nervous system; however, whether this is the cause or effect of the pain is yet to be determined. Further research is urgently required to develop our understanding of the pathogenesis of pain in DPN in order to develop new and effective mechanistic treatments for painful-DPN.
Collapse
Affiliation(s)
- Pallai Shillo
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Glossop Road, Sheffield, S10 2JF UK
| | - Gordon Sloan
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Glossop Road, Sheffield, S10 2JF UK
| | - Marni Greig
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Glossop Road, Sheffield, S10 2JF UK
| | - Leanne Hunt
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Glossop Road, Sheffield, S10 2JF UK
| | - Dinesh Selvarajah
- Department of Oncology and Human Metabolism, University of Sheffield, Sheffield, UK
| | - Jackie Elliott
- Department of Oncology and Human Metabolism, University of Sheffield, Sheffield, UK
| | - Rajiv Gandhi
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Glossop Road, Sheffield, S10 2JF UK
| | | | - Solomon Tesfaye
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Glossop Road, Sheffield, S10 2JF UK
- Department of Oncology and Human Metabolism, University of Sheffield, Sheffield, UK
| |
Collapse
|
50
|
Ciaramitaro P, Cruccu G, de Tommaso M, Devigili G, Fornasari D, Geppetti P, Lacerenza M, Lauria G, Mameli S, Marchettini P, Nolano M, Polati E, Provitera V, Romano M, Solaro C, Tamburin S, Tugnoli V, Valeriani M, Truini A. A Delphi consensus statement of the Neuropathic Pain Special Interest Group of the Italian Neurological Society on pharmacoresistant neuropathic pain. Neurol Sci 2019; 40:1425-1431. [PMID: 30941628 DOI: 10.1007/s10072-019-03870-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/23/2019] [Indexed: 12/25/2022]
Abstract
To improve patient care and help clinical research, the Neuropathic Pain Special Interest Group of the Italian Neurological Society appointed a task force to elaborate a consensus statement on pharmacoresistant neuropathic pain. The task force included 19 experts in neuropathic pain. These experts participated in a Delphi survey consisting of three consecutive rounds of questions and a face-to-face meeting, designed to achieve a consensus definition of pharmacoresistant neuropathic pain. In the three rounds of questions, the participants identified and described the main distinguishing features of pharmacoresistance. In the face-to-face meeting the participants discussed the clinical features determining pharmacoresistance. They finally agreed that neuropathic pain is pharmacoresistant when "the patient does not reach the 50% reduction of pain or an improvement of at least 2 points in the Patient Global Impression of Change, having used all drug classes indicated as first, second, or third line in the most recent and widely agreed international guidelines, for at least 1 month after titration to the highest tolerable dose." Our consensus statement might be useful for identifying eligible patients for invasive treatments, and selecting patients in pharmacological trials, thus improving patient care and helping clinical research.
Collapse
Affiliation(s)
- P Ciaramitaro
- Clinical Neurophysiology, Neuroscience Department, AOU Città della Salute e della Scienza di Torino, Torino, Italy
| | - G Cruccu
- Department of Human Neuroscience, University Sapienza, Viale Università 30, 00185, Rome, Italy
| | - M de Tommaso
- Applied Neurophysiology and Pain Unit, Basic Medical Science, Neuroscience and Sensory System-SMBNOS-Department, Aldo Moro University, Bari, Italy
| | - G Devigili
- Neurological Unit 1, Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, 20133, Milan, Italy
| | - D Fornasari
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - P Geppetti
- Department of Health Sciences, Section of Clinical Pharmacology and Headache Center, University of Florence, Florence, Italy
| | - M Lacerenza
- Neurology and Pain Medicine Center, Humanitas, San Pio X Clinic, Milan, Italy
| | - G Lauria
- Neuroalgology Unit, IRCCS Foundation, "Carlo Besta" Neurological Institute, Milan, Italy
- Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, Milan, Italy
| | - S Mameli
- Pain Therapy Unit, "A. Businco" Hospital, ASL 8, 09134, Cagliari, Italy
| | - P Marchettini
- Department of Health Sciences, Section of Clinical Pharmacology and Headache Center, University of Florence, Florence, Italy
- Pain Medicine Centre, Ospedale San Raffaele, Milan, Italy
- University of Applied Science of Southern Switzerland, Pain Pathophysiology and Therapy Programme, Manno, Switzerland
| | - M Nolano
- Neurology Department, Skin Biopsy Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri, 4, 27100, Pavia, Italy
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University Federico II of Naples, Naples, Italy
| | - E Polati
- Department of Surgery, Dentistry, Maternal and Infant Sciences, Pain Therapy Centre, Verona University Hospital, Verona, Italy
| | - V Provitera
- Istituti Clinici Scientifici Maugeri IRCCS, Institute of Telese Terme (BN), Telese Terme, Italy
| | - M Romano
- Neurology Unit, Azienda Ospedaliera Ospedali Riuniti Villa Sofia Cervello, Palermo, Italy
| | - C Solaro
- CRRF Mons L Novarese Moncrivello (VC), Moncrivello, Italy
| | - S Tamburin
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - V Tugnoli
- Neurology Unit, Azienda Ospedaliero-Universitaria, Ferrara, Italy
| | - M Valeriani
- Division of Neurology, Ospedale Pediatrico Bambino Gesú, IRCCS, Rome, Italy
- Center for Sensory-Motor Interaction, Aalborg University, Aalborg, Denmark
| | - Andrea Truini
- Department of Human Neuroscience, University Sapienza, Viale Università 30, 00185, Rome, Italy.
| |
Collapse
|