Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Pathogenesis, diagnosis and clinical management of diabetic sensorimotor peripheral neuropathy

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.

Key points

  • Diabetic sensorimotor peripheral neuropathy (DSPN) is a common complication of diabetes mellitus that is associated with increased mortality, neuropathic pain, foot ulceration and lower-limb amputation.

  • The mechanisms of DSPN are not fully understood but involve downstream injurious pathways associated with hyperglycaemia, dyslipidaemia and microvascular disease leading to neuronal inflammation, oxidative stress, mitochondrial dysfunction and cell death.

  • There are no universally accepted disease-modifying treatments for DSPN; management is focused on optimizing glycaemic control, the achievement of cardiometabolic targets, and the prevention and/or treatment of diabetic foot complications.

  • The reasons why some patients develop painless rather than painful DSPN are unknown; however, alterations in the peripheral and central nervous system have emerged as potential explanations.

  • New compounds for the treatment of painful DSPN are being developed and the concept of stratifying patients according to various pain characteristics to improve analgesic response is being explored.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: The pathways to foot ulceration and amputation.
Fig. 2: Peripheral and central nervous system structural and functional abnormalities in DSPN.
Fig. 3: Mechanisms of neuronal cell death in DSPN.
Fig. 4: Mechanisms of painful DSPN.
Fig. 5: Management of DSPN.

Similar content being viewed by others

References

  1. World Health Organization. Global Report on Diabetes. WHO https://www.who.int/publications/i/item/9789241565257 (2016).

  2. Saeedi, P. et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the International Diabetes Federation Diabetes Atlas, 9. Diabetes Res. Clin. Pract. 157, 107843 (2019).

    PubMed  Google Scholar 

  3. Atun, R. et al. Diabetes in sub-Saharan Africa: from clinical care to health policy. Lancet Diabetes Endocrinol. 5, 622–667 (2017).

    PubMed  Google Scholar 

  4. Paisey, R. B. et al. Diabetes-related major lower limb amputation incidence is strongly related to diabetic foot service provision and improves with enhancement of services: peer review of the South-West of England. Diabet. Med. 35, 53–62 (2018).

    CAS  PubMed  Google Scholar 

  5. Zhang, Y. et al. Global disability burdens of diabetes-related lower-extremity complications in 1990 and 2016. Diabetes Care 43, 964–974 (2020).

    PubMed  Google Scholar 

  6. Brownrigg, J. R. et al. Microvascular disease and risk of cardiovascular events among individuals with type 2 diabetes: a population-level cohort study. Lancet Diabetes Endocrinol. 4, 588–597 (2016).

    PubMed  Google Scholar 

  7. Vadiveloo, T. et al. Amputation-free survival in 17,353 people at high risk for foot ulceration in diabetes: a national observational study. Diabetologia 61, 2590–2597 (2018).

    PubMed  PubMed Central  Google Scholar 

  8. Rastogi, A. et al. Long term outcomes after incident diabetic foot ulcer: multicenter large cohort prospective study (EDI-FOCUS investigators) Epidemiology of Diabetic Foot Complications Study. Diabetes Res. Clin. Pract. 162, 108113 (2020).

    PubMed  Google Scholar 

  9. Kerr, M. Foot care in diabetes: the human and financial cost. Insight Health Economics http://www.londonscn.nhs.uk/wp-content/uploads/2017/04/dia-foot-care-mtg-kerr-27042017.pdf (2017).

  10. Feldman, E. L., Nave, K. A., Jensen, T. S. & Bennett, D. L. New horizons in diabetic neuropathy: mechanisms, bioenergetics, and pain. Neuron 93, 1296–1313 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Rosenberger, D. C., Blechschmidt, V., Timmerman, H., Wolff, A. & Treede, R. D. Challenges of neuropathic pain: focus on diabetic neuropathy. J. Neural Transm. 127, 589–624 (2020).

    PubMed  Google Scholar 

  12. Calcutt, N. A. Diabetic neuropathy and neuropathic pain: a (con)fusion of pathogenic mechanisms? Pain 161, S65–S86 (2020).

    PubMed  PubMed Central  Google Scholar 

  13. Selvarajah, D. et al. Diabetic peripheral neuropathy: advances in diagnosis and strategies for screening and early intervention. Lancet Diabetes Endocrinol. 7, 938–948 (2019).

    PubMed  Google Scholar 

  14. Bönhof, G. J. et al. Emerging biomarkers, tools, and treatments for diabetic polyneuropathy. Endocr. Rev. 40, 153–192 (2019).

    PubMed  Google Scholar 

  15. Finnerup, N. B. et al. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurol. 14, 162–173 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Boulton, A. J. M., et al. Diagnosis and management of diabetic foot complications. (American Diabetes Association, 2018).

  17. Pop-Busui, R. et al. Diabetic neuropathy: a position statement by the American Diabetes Association. Diabetes Care 40, 136–154 (2017).

    CAS  PubMed  Google Scholar 

  18. Tesfaye, S. et al. Diabetic neuropathies: update on definitions, diagnostic criteria, estimation of severity, and treatments. Diabetes Care 33, 2285–2293 (2010).

    PubMed  PubMed Central  Google Scholar 

  19. Ward, J. D. The diabetic leg. Diabetologia 22, 141–147 (1982).

    CAS  PubMed  Google Scholar 

  20. Sloan, G. et al. A new look at painful diabetic neuropathy. Diabetes Res. Clin. Pract. 144, 177–191 (2018).

    PubMed  Google Scholar 

  21. Tesfaye, S., Boulton, A. J. & Dickenson, A. H. Mechanisms and management of diabetic painful distal symmetrical polyneuropathy. Diabetes Care 36, 2456–2465 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Reiber, G. E. et al. Causal pathways for incident lower-extremity ulcers in patients with diabetes from two settings. Diabetes Care 22, 157–162 (1999).

    CAS  PubMed  Google Scholar 

  23. Rogers, L. C. et al. The Charcot foot in diabetes. Diabetes Care 34, 2123–2129 (2011).

    PubMed  PubMed Central  Google Scholar 

  24. Rayman, G. et al. The Ipswich Touch Test: a simple and novel method to identify inpatients with diabetes at risk of foot ulceration. Diabetes Care 34, 1517–1518 (2011).

    PubMed  PubMed Central  Google Scholar 

  25. Miller, J. D. et al. How to do a 3-minute diabetic foot exam. J. Fam. Pract. 63, 646–656 (2014).

    PubMed  Google Scholar 

  26. Feldman, E. L. et al. A practical two-step quantitative clinical and electrophysiological assessment for the diagnosis and staging of diabetic neuropathy. Diabetes Care 17, 1281–1289 (1994).

    CAS  PubMed  Google Scholar 

  27. Bril, V. & Perkins, B. A. Validation of the Toronto Clinical Scoring System for diabetic polyneuropathy. Diabetes Care 25, 2048–2052 (2002).

    PubMed  Google Scholar 

  28. Bril, V., Tomioka, S., Buchanan, R. A. & Perkins, B. A., mTCNS Study Group. Reliability and validity of the modified Toronto Clinical Neuropathy Score in diabetic sensorimotor polyneuropathy. Diabet. Med. 26, 240–246 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Young, M. J., Boulton, A. J., MacLeod, A. F., Williams, D. R. & Sonksen, P. H. A multicentre study of the prevalence of diabetic peripheral neuropathy in the United Kingdom hospital clinic population. Diabetologia 36, 150–154 (1993).

    CAS  PubMed  Google Scholar 

  30. Dyck, P. J. Detection, characterization, and staging of polyneuropathy: assessed in diabetics. Muscle Nerve 11, 21–32 (1988).

    CAS  PubMed  Google Scholar 

  31. Singleton, J. R. et al. The Utah Early Neuropathy Scale: a sensitive clinical scale for early sensory predominant neuropathy. J. Peripher. Nerv. Syst. 13, 218–227 (2008).

    PubMed  Google Scholar 

  32. Bastyr, E. J., Price, K. L., Bril, V. & Group, M. S. Development and validity testing of the neuropathy total symptom score-6: questionnaire for the study of sensory symptoms of diabetic peripheral neuropathy. Clin. Ther. 27, 1278–1294 (2005).

    PubMed  Google Scholar 

  33. Freynhagen, R., Baron, R., Gockel, U. & Tölle, T. R. painDETECT: a new screening questionnaire to identify neuropathic components in patients with back pain. Curr. Med. Res. Opin. 22, 1911–1920 (2006).

    PubMed  Google Scholar 

  34. Spallone, V. et al. Validation of DN4 as a screening tool for neuropathic pain in painful diabetic polyneuropathy. Diabet. Med. 29, 578–585 (2012).

    CAS  PubMed  Google Scholar 

  35. Bouhassira, D. et al. Development and validation of the neuropathic pain symptom inventory. Pain 108, 248–257 (2004).

    PubMed  Google Scholar 

  36. Lee, J. A. et al. Reliability and validity of a point-of-care sural nerve conduction device for identification of diabetic neuropathy. PLoS ONE 9, e86515 (2014).

    PubMed  PubMed Central  Google Scholar 

  37. Selvarajah, D. et al. SUDOSCAN: a simple, rapid, and objective method with potential for screening for diabetic peripheral neuropathy. PLoS ONE 10, e0138224 (2015).

    PubMed  PubMed Central  Google Scholar 

  38. Papanas, N. & Ziegler, D. Corneal confocal microscopy: recent progress in the evaluation of diabetic neuropathy. J. Diabetes Investig. 6, 381–389 (2015).

    PubMed  PubMed Central  Google Scholar 

  39. Binns-Hall, O. et al. One-stop microvascular screening service: an effective model for the early detection of diabetic peripheral neuropathy and the high-risk foot. Diabet. Med. 35, 887–894 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Didangelos, T. et al. Vitamin B12 supplementation in diabetic neuropathy: a 1-year, randomized, double-blind, placebo-controlled trial. Nutrients 13, 395 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. England, J. D. et al. Distal symmetric polyneuropathy: a definition for clinical research: report of the American Academy of Neurology, the American Association of Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. Neurology 64, 199–207 (2005).

    CAS  PubMed  Google Scholar 

  42. Dyck, P. J., Davies, J. L., Litchy, W. J. & O’Brien, P. C. Longitudinal assessment of diabetic polyneuropathy using a composite score in the Rochester Diabetic Neuropathy Study cohort. Neurology 49, 229–239 (1997).

    CAS  PubMed  Google Scholar 

  43. Cornblath, D. R. et al. Total neuropathy score: validation and reliability study. Neurology 53, 1660–1664 (1999).

    CAS  PubMed  Google Scholar 

  44. Vileikyte, L. et al. The development and validation of a neuropathy- and foot ulcer-specific quality of life instrument. Diabetes Care 26, 2549–2555 (2003).

    PubMed  Google Scholar 

  45. Vinik, E. J. et al. The development and validation of the Norfolk QOL-DN, a new measure of patients’ perception of the effects of diabetes and diabetic neuropathy. Diabetes Technol. Ther. 7, 497–508 (2005).

    PubMed  Google Scholar 

  46. Devigili, G. et al. Diagnostic criteria for small fibre neuropathy in clinical practice and research. Brain 142, 3728–3736 (2019).

    PubMed  PubMed Central  Google Scholar 

  47. Ziegler, D., Papanas, N., Vinik, A. I. & Shaw, J. E. Epidemiology of polyneuropathy in diabetes and prediabetes. Handb. Clin. Neurol. 126, 3–22 (2014).

    PubMed  Google Scholar 

  48. Mizokami-Stout, K. R. et al. The contemporary prevalence of diabetic neuropathy in type 1 diabetes: findings from the T1D exchange. Diabetes Care 43, 806–812 (2020).

    PubMed  PubMed Central  Google Scholar 

  49. Jeyam, A. et al. Diabetic neuropathy is a substantial burden in people with type 1 diabetes and is strongly associated with socioeconomic disadvantage: a population-representative study from Scotland. Diabetes Care 43, 734–742 (2020).

    CAS  PubMed  Google Scholar 

  50. Tesfaye, S. et al. Prevalence of diabetic peripheral neuropathy and its relation to glycaemic control and potential risk factors: the EURODIAB IDDM Complications Study. Diabetologia 39, 1377–1384 (1996).

    CAS  PubMed  Google Scholar 

  51. Braffett, B. H. et al. Risk factors for diabetic peripheral neuropathy and cardiovascular autonomic neuropathy in the diabetes control and complications trial/epidemiology of diabetes interventions and complications (DCCT/EDIC) Study. Diabetes 69, 1000–1010 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Dyck, P. J. et al. The prevalence by staged severity of various types of diabetic neuropathy, retinopathy, and nephropathy in a population-based cohort: the Rochester Diabetic Neuropathy Study. Neurology 43, 817–824 (1993).

    CAS  PubMed  Google Scholar 

  53. Ziegler, D. et al. Prevalence of polyneuropathy in pre-diabetes and diabetes is associated with abdominal obesity and macroangiopathy: the MONICA/KORA Augsburg Surveys S2 and S3. Diabetes Care 31, 464–469 (2008).

    CAS  PubMed  Google Scholar 

  54. Christensen, D. H. et al. 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 43, 1266–1275 (2020).

    PubMed  Google Scholar 

  55. Gylfadottir, S. S. et al. Diabetic polyneuropathy and pain, prevalence, and patient characteristics: a cross-sectional questionnaire study of 5,514 patients with recently diagnosed type 2 diabetes. Pain 161, 574–583 (2020).

    PubMed  Google Scholar 

  56. Pop-Busui, R., Lu, J., Lopes, N. & Jones, T. L., BARI 2D Investigators. Prevalence of diabetic peripheral neuropathy and relation to glycemic control therapies at baseline in the BARI 2D cohort. J. Peripher. Nerv. Syst. 14, 1–13 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Ponirakis, G. et al. Prevalence and management of diabetic neuropathy in secondary care in Qatar. Diabetes Metab. Res. Rev. 36, e3286 (2020).

    CAS  PubMed  Google Scholar 

  58. Partanen, J. et al. Natural history of peripheral neuropathy in patients with non-insulin-dependent diabetes mellitus. N. Engl. J. Med. 333, 89–94 (1995).

    CAS  PubMed  Google Scholar 

  59. Jaiswal, M. et al. Prevalence of and risk factors for diabetic peripheral neuropathy in youth with type 1 and type 2 diabetes: SEARCH for diabetes in youth study. Diabetes Care 40, 1226–1232 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Treede, R. D. et al. Neuropathic pain: redefinition and a grading system for clinical and research purposes. Neurology 70, 1630–1635 (2008).

    CAS  PubMed  Google Scholar 

  61. Abbott, C. A., Malik, R. A., van Ross, E. R., Kulkarni, J. & Boulton, A. J. Prevalence and characteristics of painful diabetic neuropathy in a large community-based diabetic population in the U.K. Diabetes Care 34, 2220–2224 (2011).

    PubMed  PubMed Central  Google Scholar 

  62. Truini, A. et al. A cross-sectional study investigating frequency and features of definitely diagnosed diabetic painful polyneuropathy. Pain 159, 2658–2666 (2018).

    PubMed  Google Scholar 

  63. Ziegler, D. et al. Neuropathic pain in diabetes, prediabetes and normal glucose tolerance: the MONICA/KORA Augsburg Surveys S2 and S3. Pain. Med. 10, 393–400 (2009).

    PubMed  Google Scholar 

  64. Ponirakis, G. et al. Prevalence and risk factors for painful diabetic neuropathy in secondary healthcare in Qatar. J. Diabetes Investig. 10, 1558–1564 (2019).

    PubMed  PubMed Central  Google Scholar 

  65. Tesfaye, S. et al. Vascular risk factors and diabetic neuropathy. N. Engl. J. Med. 352, 341–350 (2005).

    CAS  PubMed  Google Scholar 

  66. Ziegler, D. et al. High prevalence of diagnosed and undiagnosed polyneuropathy in subjects with and without diabetes participating in a nationwide educational initiative (PROTECT study). J. Diabetes Complications 29, 998–1002 (2015).

    PubMed  Google Scholar 

  67. Andersen, S. T. et al. Risk factors for incident diabetic polyneuropathy in a cohort with screen-detected type 2 diabetes followed for 13 years: ADDITION-denmark. Diabetes Care 41, 1068–1075 (2018).

    CAS  PubMed  Google Scholar 

  68. Wu, S. et al. Effects of common polymorphisms in the MTHFR and ACE genes on diabetic peripheral neuropathy progression: a meta-analysis. Mol. Neurobiol. 54, 2435–2444 (2017).

    CAS  PubMed  Google Scholar 

  69. Tang, Y. et al. A genetic locus on chromosome 2q24 predicting peripheral neuropathy risk in type 2 diabetes: results from the ACCORD and BARI 2D studies. Diabetes 68, 1649–1662 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Hébert, H. L., Veluchamy, A., Torrance, N. & Smith, B. H. Risk factors for neuropathic pain in diabetes mellitus. Pain 158, 560–568 (2017).

    PubMed  Google Scholar 

  71. Raputova, J. et al. Sensory phenotype and risk factors for painful diabetic neuropathy: a cross-sectional observational study. Pain 158, 2340–2353 (2017).

    PubMed  PubMed Central  Google Scholar 

  72. Themistocleous, A. C. et al. The Pain in Neuropathy Study (PiNS): a cross-sectional observational study determining the somatosensory phenotype of painful and painless diabetic neuropathy. Pain 157, 1132–1145 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Van Acker, K. et al. Prevalence and impact on quality of life of peripheral neuropathy with or without neuropathic pain in type 1 and type 2 diabetic patients attending hospital outpatients clinics. Diabetes Metab. 35, 206–213 (2009).

    PubMed  Google Scholar 

  74. Ziegler, D. et al. Painful and painless neuropathies are distinct and largely undiagnosed entities in subjects participating in an educational initiative (PROTECT study). Diabetes Res. Clin. Pract. 139, 147–154 (2018).

    PubMed  Google Scholar 

  75. Bartley, E. J. & Fillingim, R. B. Sex differences in pain: a brief review of clinical and experimental findings. Br. J. Anaesth. 111, 52–58 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Blesneac, I. et al. Rare Nav1.7 variants associated with painful diabetic peripheral neuropathy. Pain 159, 469–480 (2018).

    CAS  PubMed  Google Scholar 

  77. Alsaloum, M. et al. A gain-of-function sodium channel β2-subunit mutation in painful diabetic neuropathy. Mol. Pain. 15, 1744806919849802 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Meng, W. et al. A genome-wide association study provides evidence of sex-specific involvement of Chr1p35.1 (ZSCAN20-TLR12P) and Chr8p23.1 (HMGB1P46) with diabetic neuropathic pain. EBioMedicine 2, 1386–1393 (2015).

    PubMed  PubMed Central  Google Scholar 

  79. Meng, W. et al. A genome-wide association study suggests an association of Chr8p21.3 (GFRA2) with diabetic neuropathic pain. Eur. J. Pain. 19, 392–399 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Yorek, M. A. Alternatives to the streptozotocin-diabetic rodent. Int. Rev. Neurobiol. 127, 89–112 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Feldman, E. L. et al. Diabetic neuropathy. Nat. Rev. Dis. Prim. 5, 41 (2019).

    PubMed  Google Scholar 

  82. Callaghan, B. C., Little, A. A., Feldman, E. L. & Hughes, R. A. Enhanced glucose control for preventing and treating diabetic neuropathy. Cochrane Database Syst. Rev. 6, CD007543 (2012).

    Google Scholar 

  83. Hur, J. et al. Transcriptional networks of murine diabetic peripheral neuropathy and nephropathy: common and distinct gene expression patterns. Diabetologia 59, 1297–1306 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. McGregor, B. A. et al. Conserved transcriptional signatures in human and murine diabetic peripheral neuropathy. Sci. Rep. 8, 17678 (2018).

    PubMed  PubMed Central  Google Scholar 

  85. Schamarek, I. et al. Adiponectin, markers of subclinical inflammation and nerve conduction in individuals with recently diagnosed type 1 and type 2 diabetes. Eur. J. Endocrinol. 174, 433–443 (2016).

    CAS  PubMed  Google Scholar 

  86. Løseth, S. et al. Small and large fiber neuropathy in those with type 1 and type 2 diabetes: a 5-year follow-up study. J. Peripher. Nerv. Syst. 21, 15–21 (2016).

    PubMed  Google Scholar 

  87. Borire, A. A. et al. Correlation between markers of peripheral nerve function and structure in type 1 diabetes. Diabetes Metab. Res. Rev. 34, e3028 (2018).

    PubMed  Google Scholar 

  88. Jende, J. M. E. et al. Diabetic neuropathy differs between type 1 and type 2 diabetes: insights from magnetic resonance neurography. Ann. Neurol. 83, 588–598 (2018).

    CAS  PubMed  Google Scholar 

  89. Oates, P. J. Aldose reductase, still a compelling target for diabetic neuropathy. Curr. Drug. Targets 9, 14–36 (2008).

    CAS  PubMed  Google Scholar 

  90. Brownlee, M. Biochemistry and molecular cell biology of diabetic complications. Nature 414, 813–820 (2001).

    CAS  PubMed  Google Scholar 

  91. Edwards, J. L., Vincent, A. M., Cheng, H. T. & Feldman, E. L. Diabetic neuropathy: mechanisms to management. Pharmacol. Ther. 120, 1–34 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Wada, R. & Yagihashi, S. Role of advanced glycation end products and their receptors in development of diabetic neuropathy. Ann. N. Y. Acad. Sci. 1043, 598–604 (2005).

    CAS  PubMed  Google Scholar 

  93. Vincent, A. M. et al. Receptor for advanced glycation end products activation injures primary sensory neurons via oxidative stress. Endocrinology 148, 548–558 (2007).

    CAS  PubMed  Google Scholar 

  94. Vincent, A. M., Russell, J. W., Low, P. & Feldman, E. L. Oxidative stress in the pathogenesis of diabetic neuropathy. Endocr. Rev. 25, 612–628 (2004).

    CAS  PubMed  Google Scholar 

  95. Pop-Busui, R., Sima, A. & Stevens, M. Diabetic neuropathy and oxidative stress. Diabetes Metab. Res. Rev. 22, 257–273 (2006).

    CAS  PubMed  Google Scholar 

  96. Ziegler, D., Buchholz, S., Sohr, C., Nourooz-Zadeh, J. & Roden, M. Oxidative stress predicts progression of peripheral and cardiac autonomic nerve dysfunction over 6 years in diabetic patients. Acta Diabetol. 52, 65–72 (2015).

    CAS  PubMed  Google Scholar 

  97. Herder, C. et al. Myeloperoxidase, superoxide dismutase-3, cardiometabolic risk factors, and distal sensorimotor polyneuropathy: The KORA F4/FF4 study. Diabetes Metab. Res. Rev. 34, e3000 (2018).

    PubMed  Google Scholar 

  98. Morales, J. et al. Review of poly (ADP-ribose) polymerase (PARP) mechanisms of action and rationale for targeting in cancer and other diseases. Crit. Rev. Eukaryot. Gene Expr. 24, 15–28 (2014).

    PubMed  PubMed Central  Google Scholar 

  99. Obrosova, I. G. et al. Oxidative-nitrosative stress and poly(ADP-ribose) polymerase (PARP) activation in experimental diabetic neuropathy: the relation is revisited. Diabetes 54, 3435–3441 (2005).

    CAS  PubMed  Google Scholar 

  100. Obrosova, I. G. et al. Role of poly(ADP-ribose) polymerase activation in diabetic neuropathy. Diabetes 53, 711–720 (2004).

    CAS  PubMed  Google Scholar 

  101. Vincent, A. M. et al. Mitochondrial biogenesis and fission in axons in cell culture and animal models of diabetic neuropathy. Acta Neuropathol. 120, 477–489 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Edwards, J. L. et al. Diabetes regulates mitochondrial biogenesis and fission in mouse neurons. Diabetologia 53, 160–169 (2010).

    CAS  PubMed  Google Scholar 

  103. Chowdhury, S. K., Smith, D. R. & Fernyhough, P. The role of aberrant mitochondrial bioenergetics in diabetic neuropathy. Neurobiol. Dis. 51, 56–65 (2013).

    CAS  PubMed  Google Scholar 

  104. Fernyhough, P. Mitochondrial dysfunction in diabetic neuropathy: a series of unfortunate metabolic events. Curr. Diab. Rep. 15, 89 (2015).

    PubMed  Google Scholar 

  105. Akude, E. et al. Diminished superoxide generation is associated with respiratory chain dysfunction and changes in the mitochondrial proteome of sensory neurons from diabetic rats. Diabetes 60, 288–297 (2011).

    CAS  PubMed  Google Scholar 

  106. Davidson, E. P., Coppey, L. J., Calcutt, N. A., Oltman, C. L. & Yorek, M. A. Diet-induced obesity in Sprague-Dawley rats causes microvascular and neural dysfunction. Diabetes Metab. Res. Rev. 26, 306–318 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Yorek, M. S. et al. Effect of diet-induced obesity or type 1 or type 2 diabetes on corneal nerves and peripheral neuropathy in C57Bl/6J mice. J. Peripher. Nerv. Syst. 20, 24–31 (2015).

    PubMed  PubMed Central  Google Scholar 

  108. Wiggin, T. D. et al. Elevated triglycerides correlate with progression of diabetic neuropathy. Diabetes 58, 1634–1640 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Rumora, A. E. et al. Dyslipidemia impairs mitochondrial trafficking and function in sensory neurons. FASEB J. 32, 195–207 (2018).

    CAS  PubMed  Google Scholar 

  110. Padilla, A., Descorbeth, M., Almeyda, A. L., Payne, K. & De Leon, M. Hyperglycemia magnifies Schwann cell dysfunction and cell death triggered by PA-induced lipotoxicity. Brain Res. 1370, 64–79 (2011).

    CAS  PubMed  Google Scholar 

  111. Nowicki, M. et al. Oxidized low-density lipoprotein (oxLDL)-induced cell death in dorsal root ganglion cell cultures depends not on the lectin-like oxLDL receptor-1 but on the toll-like receptor-4. J. Neurosci. Res. 88, 403–412 (2010).

    CAS  PubMed  Google Scholar 

  112. Honjo, T. et al. Essential role of NOXA1 in generation of reactive oxygen species induced by oxidized low-density lipoprotein in human vascular endothelial cells. Endothelium 15, 137–141 (2008).

    CAS  PubMed  Google Scholar 

  113. Vincent, A. M. et al. Dyslipidemia-induced neuropathy in mice: the role of oxLDL/LOX-1. Diabetes 58, 2376–2385 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Hur, J. et al. The identification of gene expression profiles associated with progression of human diabetic neuropathy. Brain 134, 3222–3235 (2011).

    PubMed  PubMed Central  Google Scholar 

  115. Pande, M. et al. Transcriptional profiling of diabetic neuropathy in the BKS db/db mouse: a model of type 2 diabetes. Diabetes 60, 1981–1989 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Hur, J. et al. The metabolic syndrome and microvascular complications in a murine model of type 2 diabetes. Diabetes 64, 3294–3304 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Doupis, J. et al. Microvascular reactivity and inflammatory cytokines in painful and painless peripheral diabetic neuropathy. J. Clin. Endocrinol. Metab. 94, 2157–2163 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Herder, C. et al. Proinflammatory cytokines predict the incidence and progression of distal sensorimotor polyneuropathy: KORA F4/FF4 study. Diabetes Care 40, 569–576 (2017).

    CAS  PubMed  Google Scholar 

  119. Younger, D. S., Rosoklija, G., Hays, A. P., Trojaborg, W. & Latov, N. Diabetic peripheral neuropathy: a clinicopathologic and immunohistochemical analysis of sural nerve biopsies. Muscle Nerve 19, 722–727 (1996).

    CAS  PubMed  Google Scholar 

  120. Hotamisligil, G. S. Inflammation and metabolic disorders. Nature 444, 860–867 (2006).

    CAS  PubMed  Google Scholar 

  121. Shoelson, S. E., Lee, J. & Goldfine, A. B. Inflammation and insulin resistance. J. Clin. Invest. 116, 1793–1801 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Libby, P. Inflammation in atherosclerosis. Nature 420, 868–874 (2002).

    CAS  PubMed  Google Scholar 

  123. Pop-Busui, R., Ang, L., Holmes, C., Gallagher, K. & Feldman, E. L. Inflammation as a therapeutic target for diabetic neuropathies. Curr. Diab. Rep. 16, 29 (2016).

    PubMed  PubMed Central  Google Scholar 

  124. Leinninger, G. M., Vincent, A. M. & Feldman, E. L. The role of growth factors in diabetic peripheral neuropathy. J. Peripher. Nerv. Syst. 9, 26–53 (2004).

    CAS  PubMed  Google Scholar 

  125. Hellweg, R., Raivich, G., Hartung, H. D., Hock, C. & Kreutzberg, G. W. Axonal transport of endogenous nerve growth factor (NGF) and NGF receptor in experimental diabetic neuropathy. Exp. Neurol. 130, 24–30 (1994).

    CAS  PubMed  Google Scholar 

  126. Said, G., Slama, G. & Selva, J. Progressive centripetal degeneration of axons in small fibre diabetic polyneuropathy. Brain 106, 791–807 (1983).

    PubMed  Google Scholar 

  127. Gonçalves, N. P. et al. Schwann cell interactions with axons and microvessels in diabetic neuropathy. Nat. Rev. Neurol. 13, 135–147 (2017).

    PubMed  PubMed Central  Google Scholar 

  128. Viader, A. et al. Aberrant Schwann cell lipid metabolism linked to mitochondrial deficits leads to axon degeneration and neuropathy. Neuron 77, 886–898 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Malik, R. A. et al. Microangiopathy in human diabetic neuropathy: relationship between capillary abnormalities and the severity of neuropathy. Diabetologia 32, 92–102 (1989).

    CAS  PubMed  Google Scholar 

  130. Malik, R. A. et al. Sural nerve pathology in diabetic patients with minimal but progressive neuropathy. Diabetologia 48, 578–585 (2005).

    CAS  PubMed  Google Scholar 

  131. Tesfaye, S. et al. Impaired blood flow and arterio-venous shunting in human diabetic neuropathy: a novel technique of nerve photography and fluorescein angiography. Diabetologia 36, 1266–1274 (1993).

    CAS  PubMed  Google Scholar 

  132. Tesfaye, S., Harris, N. D., Wilson, R. M. & Ward, J. D. Exercise-induced conduction velocity increment: a marker of impaired peripheral nerve blood flow in diabetic neuropathy. Diabetologia 35, 155–159 (1992).

    CAS  PubMed  Google Scholar 

  133. Tabit, C. E., Chung, W. B., Hamburg, N. M. & Vita, J. A. Endothelial dysfunction in diabetes mellitus: molecular mechanisms and clinical implications. Rev. Endocr. Metab. Disord. 11, 61–74 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Sytze Van Dam, P., Cotter, M. A., Bravenboer, B. & Cameron, N. E. Pathogenesis of diabetic neuropathy: focus on neurovascular mechanisms. Eur. J. Pharmacol. 719, 180–186 (2013).

    PubMed  Google Scholar 

  135. Newrick, P. G., Wilson, A. J., Jakubowski, J., Boulton, A. J. & Ward, J. D. Sural nerve oxygen tension in diabetes. Br. Med. J. 293, 1053–1054 (1986).

    CAS  Google Scholar 

  136. Østergaard, L. et al. The effects of capillary dysfunction on oxygen and glucose extraction in diabetic neuropathy. Diabetologia 58, 666–677 (2015).

    PubMed  Google Scholar 

  137. Tesfaye, S. et al. Diabetic peripheral neuropathy may not be as its name suggests: evidence from magnetic resonance imaging. Pain 157 (Suppl. 1), S72–80 (2016).

    PubMed  Google Scholar 

  138. Reske-Nielsen, E. & Lundbaek, K. Pathological changes in the central and peripheral nervous system of young long-term diabetics. II. The spinal cord and peripheral nerves. Diabetologia 4, 34–43 (1968).

    CAS  PubMed  Google Scholar 

  139. Suzuki, C. et al. Peripheral and central conduction abnormalities in diabetes mellitus. Neurology 54, 1932–1937 (2000).

    CAS  PubMed  Google Scholar 

  140. Kucera, P., Goldenberg, Z., Varsik, P., Buranova, D. & Traubner, P. Spinal cord lesions in diabetes mellitus. Somatosensory and motor evoked potentials and spinal conduction time in diabetes mellitus. Neuro Endocrinol. Lett. 26, 143–147 (2005).

    PubMed  Google Scholar 

  141. Biessels, G. J. et al. Neurophysiological changes in the central and peripheral nervous system of streptozotocin-diabetic rats. Course of development and effects of insulin treatment. Brain 122 (Pt 4), 757–768 (1999).

    PubMed  Google Scholar 

  142. Selvarajah, D. et al. Early involvement of the spinal cord in diabetic peripheral neuropathy. Diabetes Care 29, 2664–2669 (2006).

    PubMed  Google Scholar 

  143. Eaton, S. E. et al. Spinal-cord involvement in diabetic peripheral neuropathy. Lancet 358, 35–36 (2001).

    CAS  PubMed  Google Scholar 

  144. Selvarajah, D. et al. Magnetic resonance neuroimaging study of brain structural differences in diabetic peripheral neuropathy. Diabetes Care 37, 1681–1688 (2014).

    PubMed  Google Scholar 

  145. Selvarajah, D. et al. Thalamic neuronal dysfunction and chronic sensorimotor distal symmetrical polyneuropathy in patients with type 1 diabetes mellitus. Diabetologia 51, 2088–2092 (2008).

    CAS  PubMed  Google Scholar 

  146. Hansen, T. M. et al. Brain spectroscopy reveals that N-acetylaspartate is associated to peripheral sensorimotor neuropathy in type 1 diabetes. J. Diabetes Complications 33, 323–328 (2019).

    PubMed  Google Scholar 

  147. Selvarajah, D., Wilkinson, I. D., Gandhi, R., Griffiths, P. D. & Tesfaye, S. Microvascular perfusion abnormalities of the Thalamus in painful but not painless diabetic polyneuropathy: a clue to the pathogenesis of pain in type 1 diabetes. Diabetes Care 34, 718–720 (2011).

    PubMed  PubMed Central  Google Scholar 

  148. Cauda, F. et al. Low-frequency BOLD fluctuations demonstrate altered thalamocortical connectivity in diabetic neuropathic pain. BMC Neurosci. 10, 138 (2009).

    PubMed  PubMed Central  Google Scholar 

  149. Selvarajah, D. et al. Structural and functional abnormalities of the primary somatosensory cortex in diabetic peripheral neuropathy: a multimodal MRI Study. Diabetes 68, 796–806 (2019).

    CAS  PubMed  Google Scholar 

  150. Colloca, L. et al. Neuropathic pain. Nat. Rev. Dis. Prim. 3, 17002 (2017).

    PubMed  Google Scholar 

  151. Shillo, P. et al. Painful and painless diabetic neuropathies: what is the difference? Curr. Diab. Rep. 19, 32 (2019).

    PubMed  PubMed Central  Google Scholar 

  152. Spallone, V. & Greco, C. Painful and painless diabetic neuropathy: one disease or two? Curr. Diab. Rep. 13, 533–549 (2013).

    CAS  PubMed  Google Scholar 

  153. Maier, C. et al. Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): somatosensory abnormalities in 1236 patients with different neuropathic pain syndromes. Pain 150, 439–450 (2010).

    CAS  PubMed  Google Scholar 

  154. Anand, P. & Bley, K. Topical capsaicin for pain management: therapeutic potential and mechanisms of action of the new high-concentration capsaicin 8% patch. Br. J. Anaesth. 107, 490–502 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Hoeijmakers, J. G., Faber, C. G., Lauria, G., Merkies, I. S. & Waxman, S. G. Small-fibre neuropathies–advances in diagnosis, pathophysiology and management. Nat. Rev. Neurol. 8, 369–379 (2012).

    CAS  PubMed  Google Scholar 

  156. Marshall, A. G. et al. Spinal disinhibition in experimental and clinical painful diabetic neuropathy. Diabetes 66, 1380–1390 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Quattrini, C. et al. Surrogate markers of small fiber damage in human diabetic neuropathy. Diabetes 56, 2148–2154 (2007).

    CAS  PubMed  Google Scholar 

  158. Kalteniece, A. et al. Corneal confocal microscopy detects small nerve fibre damage in patients with painful diabetic neuropathy. Sci. Rep. 10, 3371 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Bönhof, G. J. et al. Patterns of cutaneous nerve fibre loss and regeneration in type 2 diabetes with painful and painless polyneuropathy. Diabetologia 60, 2495–2503 (2017).

    PubMed  Google Scholar 

  160. Cheng, H. T. et al. Increased axonal regeneration and swellings in intraepidermal nerve fibers characterize painful phenotypes of diabetic neuropathy. J. Pain. 14, 941–947 (2013).

    PubMed  PubMed Central  Google Scholar 

  161. Scheytt, S., Riediger, N., Braunsdorf, S., Sommer, C. & Üçeyler, N. Increased gene expression of growth associated protein-43 in skin of patients with early-stage peripheral neuropathies. J. Neurol. Sci. 355, 131–137 (2015).

    CAS  PubMed  Google Scholar 

  162. Cheung, A., Podgorny, P., Martinez, J. A., Chan, C. & Toth, C. Epidermal axonal swellings in painful and painless diabetic peripheral neuropathy. Muscle Nerve 51, 505–513 (2015).

    PubMed  Google Scholar 

  163. Alam, U. et al. Vitamin D deficiency is associated with painful diabetic neuropathy. Diabetes Metab. Res. Rev. https://doi.org/10.1002/dmrr.3361 (2020).

    Article  PubMed  Google Scholar 

  164. Gandhi, R. A., Marques, J. L., Selvarajah, D., Emery, C. J. & Tesfaye, S. Painful diabetic neuropathy is associated with greater autonomic dysfunction than painless diabetic neuropathy. Diabetes Care 33, 1585–1590 (2010).

    PubMed  PubMed Central  Google Scholar 

  165. Spallone, V. et al. Clinical correlates of painful diabetic neuropathy and relationship of neuropathic pain with sensorimotor and autonomic nerve function. Eur. J. Pain. 15, 153–160 (2011).

    PubMed  Google Scholar 

  166. D’Amato, C. et al. A novel association between nondipping and painful diabetic polyneuropathy. Diabetes Care 37, 2640–2642 (2014).

    PubMed  Google Scholar 

  167. Krämer, H. H., Rolke, R., Bickel, A. & Birklein, F. Thermal thresholds predict painfulness of diabetic neuropathies. Diabetes Care 27, 2386–2391 (2004).

    PubMed  Google Scholar 

  168. Hansen, C. S. et al. The role of serum methylglyoxal on diabetic peripheral and cardiovascular autonomic neuropathy: the ADDITION Denmark study. Diabet. Med. 32, 778–785 (2015).

    CAS  PubMed  Google Scholar 

  169. Tesfaye, S. et al. Arterio-venous shunting and proliferating new vessels in acute painful neuropathy of rapid glycaemic control (insulin neuritis). Diabetologia 39, 329–335 (1996).

    CAS  PubMed  Google Scholar 

  170. Eaton, S. E. et al. Increased sural nerve epineurial blood flow in human subjects with painful diabetic neuropathy. Diabetologia 46, 934–939 (2003).

    CAS  PubMed  Google Scholar 

  171. Rayman, G., Baker, N. R. & Krishnan, S. T. Glyceryl trinitrate patches as an alternative to isosorbide dinitrate spray in the treatment of chronic painful diabetic neuropathy. Diabetes Care 26, 2697–2698 (2003).

    PubMed  Google Scholar 

  172. Yuen, K. C., Baker, N. R. & Rayman, G. Treatment of chronic painful diabetic neuropathy with isosorbide dinitrate spray: a double-blind placebo-controlled cross-over study. Diabetes Care 25, 1699–1703 (2002).

    CAS  PubMed  Google Scholar 

  173. Calvo, M., Dawes, J. M. & Bennett, D. L. The role of the immune system in the generation of neuropathic pain. Lancet Neurol. 11, 629–642 (2012).

    CAS  PubMed  Google Scholar 

  174. Purwata, T. E. High TNF-α plasma levels and macrophages iNOS and TNF-α expression as risk factors for painful diabetic neuropathy. J. Pain. Res. 4, 169–175 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  175. Herder, C. et al. Differential association between biomarkers of subclinical inflammation and painful polyneuropathy: results from the KORA F4 study. Diabetes Care 38, 91–96 (2015).

    CAS  PubMed  Google Scholar 

  176. Austin, P. J. & Moalem-Taylor, G. The neuro-immune balance in neuropathic pain: involvement of inflammatory immune cells, immune-like glial cells and cytokines. J. Neuroimmunol. 229, 26–50 (2010).

    CAS  PubMed  Google Scholar 

  177. Sun, J. S. et al. Minocycline attenuates pain by inhibiting spinal microglia activation in diabetic rats. Mol. Med. Rep. 12, 2677–2682 (2015).

    CAS  PubMed  Google Scholar 

  178. Bierhaus, A. et al. Methylglyoxal modification of Nav1.8 facilitates nociceptive neuron firing and causes hyperalgesia in diabetic neuropathy. Nat. Med. 18, 926–933 (2012).

    CAS  PubMed  Google Scholar 

  179. Baron, R., Binder, A. & Wasner, G. Neuropathic pain: diagnosis, pathophysiological mechanisms, and treatment. Lancet Neurol. 9, 807–819 (2010).

    PubMed  Google Scholar 

  180. Bennett, D. L., Clark, A. J., Huang, J. Y., Waxman, S. G. & Dib-Hajj, S. D. The role of voltage-gated sodium channels in pain signaling. Physiol. Rev. 99, 1079–1151 (2019).

    CAS  PubMed  Google Scholar 

  181. Sun, W. et al. Reduced conduction failure of the main axon of polymodal nociceptive C-fibres contributes to painful diabetic neuropathy in rats. Brain 135, 359–375 (2012).

    PubMed  Google Scholar 

  182. Djouhri, L., Malki, M. I., Zeidan, A., Nagi, K. & Smith, T. Activation of Kv7 channels with the anticonvulsant retigabine alleviates neuropathic pain behaviour in the streptozotocin rat model of diabetic neuropathy. J. Drug Target. 27, 1118–1126 (2019).

    CAS  PubMed  Google Scholar 

  183. Todorovic, S. M. & Jevtovic-Todorovic, V. T-type voltage-gated calcium channels as targets for the development of novel pain therapies. Brit. J. Pharmacol. 163, 484–495 (2011).

    CAS  Google Scholar 

  184. Bauer, C. S. et al. The Increased trafficking of the calcium channel subunit alpha(2)delta-1 to presynaptic terminals in neuropathic pain is inhibited by the alpha(2)delta Ligand Pregabalin. J. Neurosci. 29, 4076–4088 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Latham, J. R. et al. Selective T-type calcium channel blockade alleviates hyperalgesia in ob/ob mice. Diabetes 58, 2656–2665 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Latremoliere, A. & Woolf, C. J. Central sensitization: a generator of pain hypersensitivity by central neural plasticity. J. Pain. 10, 895–926 (2009).

    PubMed  PubMed Central  Google Scholar 

  187. Segerdahl, A. R., Themistocleous, A. C., Fido, D., Bennett, D. L. & Tracey, I. A brain-based pain facilitation mechanism contributes to painful diabetic polyneuropathy. Brain 141, 357–364 (2018).

    PubMed  PubMed Central  Google Scholar 

  188. Tracey, I. & Mantyh, P. W. The cerebral signature and its modulation for pain perception. Neuron 55, 377–391 (2007).

    CAS  PubMed  Google Scholar 

  189. Fischer, T. Z., Tan, A. M. & Waxman, S. G. Thalamic neuron hyperexcitability and enlarged receptive fields in the STZ model of diabetic pain. Brain Res. 1268, 154–161 (2009).

    CAS  PubMed  Google Scholar 

  190. Fischer, T. Z. & Waxman, S. G. Neuropathic pain in diabetes–evidence for a central mechanism. Nat. Rev. Neurol. 6, 462–466 (2010).

    PubMed  Google Scholar 

  191. Watanabe, K. et al. Altered cerebral blood flow in the anterior cingulate cortex is associated with neuropathic pain. J. Neurol. Neurosurg. Psychiatry 89, 1082–1087 (2018).

    PubMed  Google Scholar 

  192. Tseng, M. T., Chiang, M. C., Chao, C. C., Tseng, W. Y. & Hsieh, S. T. fMRI evidence of degeneration-induced neuropathic pain in diabetes: enhanced limbic and striatal activations. Hum. Brain Mapp. 34, 2733–2746 (2013).

    PubMed  Google Scholar 

  193. Wilkinson, I. D. et al. Determinants of treatment response in painful diabetic peripheral neuropathy. A combined deep sensory phenotyping and multi-modal brain magnetic resonance imaging study. Diabetes 69, 1804–1814 (2020).

    PubMed  Google Scholar 

  194. Dy, S. M., et al. Preventing Complications and Treating Symptoms of Diabetic Peripheral Neuropathy (Agency for Healthcare Research and Quality, 2017).

  195. No Authors Listed. Effect of intensive diabetes treatment on nerve conduction in the Diabetes Control and Complications Trial. Ann. Neurol. 38, 869–880 (1995).

    Google Scholar 

  196. Ziegler, D., Behler, M., Schroers-Teuber, M. & Roden, M. Near-normoglycaemia and development of neuropathy: a 24-year prospective study from diagnosis of type 1 diabetes. BMJ Open 5, e006559 (2015).

    PubMed  PubMed Central  Google Scholar 

  197. Kennedy, W. R., Navarro, X., Goetz, F. C., Sutherland, D. E. & Najarian, J. S. Effects of pancreatic transplantation on diabetic neuropathy. N. Engl. J. Med. 322, 1031–1037 (1990).

    CAS  PubMed  Google Scholar 

  198. Azmi, S. et al. Early nerve fibre regeneration in individuals with type 1 diabetes after simultaneous pancreas and kidney transplantation. Diabetologia 62, 1478–1487 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  199. Ismail-Beigi, F. et al. Effect of intensive treatment of hyperglycaemia on microvascular outcomes in type 2 diabetes: an analysis of the ACCORD randomised trial. Lancet 376, 419–430 (2010).

    PubMed  PubMed Central  Google Scholar 

  200. No Authors Listed. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study (UKPDS) Group. Lancet 352, 837–853 (1998).

    Google Scholar 

  201. Duckworth, W. et al. Glucose control and vascular complications in veterans with type 2 diabetes. N. Engl. J. Med. 360, 129–139 (2009).

    CAS  PubMed  Google Scholar 

  202. Ohkubo, Y. et al. Intensive insulin therapy prevents the progression of diabetic microvascular complications in Japanese patients with non-insulin-dependent diabetes mellitus: a randomized prospective 6-year study. Diabetes Res. Clin. Pract. 28, 103–117 (1995).

    CAS  PubMed  Google Scholar 

  203. Ishibashi, F., Taniguchi, M., Kosaka, A., Uetake, H. & Tavakoli, M. Improvement in neuropathy outcomes with normalizing HbA. Diabetes Care 42, 110–118 (2019).

    CAS  PubMed  Google Scholar 

  204. Ward, J. D., Barnes, C. G., Fisher, D. J., Jessop, J. D. & Baker, R. W. Improvement in nerve conduction following treatment in newly diagnosed diabetics. Lancet 1, 428–430 (1971).

    CAS  PubMed  Google Scholar 

  205. Pop-Busui, R. et al. Impact of glycemic control strategies on the progression of diabetic peripheral neuropathy in the Bypass Angioplasty Revascularization Investigation 2 Diabetes (BARI 2D) Cohort. Diabetes Care 36, 3208–3215 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  206. No Authors Listed. Tight blood pressure control and risk of macrovascular and microvascular complications in type 2 diabetes: UKPDS 38. UK Prospective Diabetes Study Group. BMJ 317, 703–713 (1998).

    Google Scholar 

  207. Gaede, P., Lund-Andersen, H., Parving, H. H. & Pedersen, O. Effect of a multifactorial intervention on mortality in type 2 diabetes. N. Engl. J. Med. 358, 580–591 (2008).

    CAS  PubMed  Google Scholar 

  208. Charles, M. et al. Prevalence of neuropathy and peripheral arterial disease and the impact of treatment in people with screen-detected type 2 diabetes: the ADDITION-Denmark study. Diabetes Care 34, 2244–2249 (2011).

    PubMed  PubMed Central  Google Scholar 

  209. Look AHEAD Research Group. Effects of a long-term lifestyle modification programme on peripheral neuropathy in overweight or obese adults with type 2 diabetes: the Look AHEAD study. Diabetologia 60, 980–988 (2017).

    Google Scholar 

  210. Balducci, S. et al. Exercise training can modify the natural history of diabetic peripheral neuropathy. J. Diabetes Complications 20, 216–223 (2006).

    PubMed  Google Scholar 

  211. Kluding, P. M. et al. The effect of exercise on neuropathic symptoms, nerve function, and cutaneous innervation in people with diabetic peripheral neuropathy. J. Diabetes Complications 26, 424–429 (2012).

    PubMed  PubMed Central  Google Scholar 

  212. Diabetes Prevention Program Research Group. Long-term effects of lifestyle intervention or metformin on diabetes development and microvascular complications over 15-year follow-up: the Diabetes Prevention Program Outcomes Study. Lancet Diabetes Endocrinol. 3, 866–875 (2015).

    PubMed Central  Google Scholar 

  213. Adams, T. D., Arterburn, D. E., Nathan, D. M. & Eckel, R. H. Clinical outcomes of metabolic surgery: microvascular and macrovascular complications. Diabetes Care 39, 912–923 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  214. Nielsen, S. F. & Nordestgaard, B. G. Statin use before diabetes diagnosis and risk of microvascular disease: a nationwide nested matched study. Lancet Diabetes Endocrinol. 2, 894–900 (2014).

    CAS  PubMed  Google Scholar 

  215. Kang, E. Y. et al. Association of statin therapy with prevention of vision-threatening diabetic retinopathy. JAMA Ophthalmol. 137, 363–371 (2019).

    PubMed  PubMed Central  Google Scholar 

  216. Davis, T. M., Yeap, B. B., Davis, W. A. & Bruce, D. G. Lipid-lowering therapy and peripheral sensory neuropathy in type 2 diabetes: the Fremantle Diabetes Study. Diabetologia 51, 562–566 (2008).

    CAS  PubMed  Google Scholar 

  217. Hernández-Ojeda, J. et al. Effect of rosuvastatin on diabetic polyneuropathy: a randomized, double-blind, placebo-controlled Phase IIa study. Diabetes Metab. Syndr. Obes. 7, 401–407 (2014).

    PubMed  PubMed Central  Google Scholar 

  218. Zangiabadi, N., Shafiee, K., Alavi, K. H., Assadi, A. R. & Damavandi, M. Atorvastatin treatment improves diabetic polyneuropathy electrophysiological changes in non-insulin dependent diabetic patients: a double blind, randomized clinical trial. Minerva Endocrinol. 37, 195–200 (2012).

    CAS  PubMed  Google Scholar 

  219. Rajamani, K. et al. Effect of fenofibrate on amputation events in people with type 2 diabetes mellitus (FIELD study): a prespecified analysis of a randomised controlled trial. Lancet 373, 1780–1788 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  220. Reja, A., Tesfaye, S., Harris, N. D. & Ward, J. D. Is ACE inhibition with lisinopril helpful in diabetic neuropathy? Diabet. Med. 12, 307–309 (1995).

    CAS  PubMed  Google Scholar 

  221. Malik, R. A. et al. Effect of angiotensin-converting-enzyme (ACE) inhibitor trandolapril on human diabetic neuropathy: randomised double-blind controlled trial. Lancet 352, 1978–1981 (1998).

    CAS  PubMed  Google Scholar 

  222. Ruggenenti, P. et al. Effects of manidipine and delapril in hypertensive patients with type 2 diabetes mellitus: the delapril and manidipine for nephroprotection in diabetes (DEMAND) randomized clinical trial. Hypertension 58, 776–783 (2011).

    CAS  PubMed  Google Scholar 

  223. Boulton, A. J., Kempler, P., Ametov, A. & Ziegler, D. Whither pathogenetic treatments for diabetic polyneuropathy? Diabetes Metab. Res. Rev. 29, 327–333 (2013).

    CAS  PubMed  Google Scholar 

  224. Low, P. A., Nickander, K. K. & Tritschler, H. J. The roles of oxidative stress and antioxidant treatment in experimental diabetic neuropathy. Diabetes 46 (Suppl. 2), S38–42 (1997).

    CAS  PubMed  Google Scholar 

  225. Ziegler, D. et al. Oral treatment with alpha-lipoic acid improves symptomatic diabetic polyneuropathy: the SYDNEY 2 trial. Diabetes Care 29, 2365–2370 (2006).

    CAS  PubMed  Google Scholar 

  226. Ziegler, D. et al. Treatment of symptomatic diabetic polyneuropathy with the antioxidant alpha-lipoic acid: a 7-month multicenter randomized controlled trial (ALADIN III Study). ALADIN III Study Group. Alpha-Lipoic Acid in Diabetic Neuropathy. Diabetes Care 22, 1296–1301 (1999).

    CAS  PubMed  Google Scholar 

  227. Çakici, N., Fakkel, T. M., van Neck, J. W., Verhagen, A. P. & Coert, J. H. Systematic review of treatments for diabetic peripheral neuropathy. Diabet. Med. 33, 1466–1476 (2016).

    PubMed  Google Scholar 

  228. Ziegler, D. et al. Efficacy and safety of antioxidant treatment with α-lipoic acid over 4 years in diabetic polyneuropathy: the NATHAN 1 trial. Diabetes Care 34, 2054–2060 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  229. Hammes, H. P. et al. Benfotiamine blocks three major pathways of hyperglycemic damage and prevents experimental diabetic retinopathy. Nat. Med. 9, 294–299 (2003).

    CAS  PubMed  Google Scholar 

  230. Stracke, H., Lindemann, A. & Federlin, K. A benfotiamine-vitamin B combination in treatment of diabetic polyneuropathy. Exp. Clin. Endocrinol. Diabetes 104, 311–316 (1996).

    CAS  PubMed  Google Scholar 

  231. Stracke, H., Gaus, W., Achenbach, U., Federlin, K. & Bretzel, R. G. Benfotiamine in diabetic polyneuropathy (BENDIP): results of a randomised, double blind, placebo-controlled clinical study. Exp. Clin. Endocrinol. Diabetes 116, 600–605 (2008).

    CAS  PubMed  Google Scholar 

  232. Fraser, D. A. et al. The effects of long-term oral benfotiamine supplementation on peripheral nerve function and inflammatory markers in patients with type 1 diabetes: a 24-month, double-blind, randomized, placebo-controlled trial. Diabetes Care 35, 1095–1097 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  233. Ekberg, K. et al. C-Peptide replacement therapy and sensory nerve function in type 1 diabetic neuropathy. Diabetes Care 30, 71–76 (2007).

    CAS  PubMed  Google Scholar 

  234. Wahren, J., Foyt, H., Daniels, M. & Arezzo, J. C. Long-acting C-peptide and neuropathy in type 1 diabetes: a 12-month clinical trial. Diabetes Care 39, 596–602 (2016).

    CAS  PubMed  Google Scholar 

  235. Chalk, C., Benstead, T. J. & Moore, F. Aldose reductase inhibitors for the treatment of diabetic polyneuropathy. Cochrane Database Syst. Rev. 4, CD004572 (2007).

    Google Scholar 

  236. Hotta, N. et al. Long-term clinical effects of epalrestat, an aldose reductase inhibitor, on diabetic peripheral neuropathy: the 3-year, multicenter, comparative Aldose Reductase Inhibitor-Diabetes Complications Trial. Diabetes Care 29, 1538–1544 (2006).

    CAS  PubMed  Google Scholar 

  237. Apfel, S. C. et al. Efficacy and safety of recombinant human nerve growth factor in patients with diabetic polyneuropathy: A randomized controlled trial. rhNGF Clinical Investigator Group. JAMA 284, 2215–2221 (2000).

    CAS  PubMed  Google Scholar 

  238. Bansal, D., Badhan, Y., Gudala, K. & Schifano, F. Ruboxistaurin for the treatment of diabetic peripheral neuropathy: a systematic review of randomized clinical trials. Diabetes Metab. J. 37, 375–384 (2013).

    PubMed  PubMed Central  Google Scholar 

  239. Akahori, H. et al. Prostaglandin E1 in lipid microspheres ameliorates diabetic peripheral neuropathy: clinical usefulness of Semmes-Weinstein monofilaments for evaluating diabetic sensory abnormality. Diabetes Res. Clin. Pract. 64, 153–159 (2004).

    CAS  PubMed  Google Scholar 

  240. Rolim, L. C., da Silva, E. M., Flumignan, R. L., Abreu, M. M. & Dib, S. A. Acetyl-L-carnitine for the treatment of diabetic peripheral neuropathy. Cochrane Database Syst. Rev. 6, CD011265 (2019).

    PubMed  Google Scholar 

  241. Ziegler, D. et al. Treatment of symptomatic polyneuropathy with actovegin in type 2 diabetic patients. Diabetes Care 32, 1479–1484 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  242. Gerstein, H. C. et al. Effects of intensive glucose lowering in type 2 diabetes. N. Engl. J. Med. 358, 2545–2559 (2008).

    CAS  PubMed  Google Scholar 

  243. Ziegler, D. & Luft, D. Clinical trials for drugs against diabetic neuropathy: can we combine scientific needs with clinical practicalities? Int. Rev. Neurobiol. 50, 431–463 (2002).

    CAS  PubMed  Google Scholar 

  244. Tesfaye, S. et al. Factors that impact symptomatic diabetic peripheral neuropathy in placebo-administered patients from two 1-year clinical trials. Diabetes Care 30, 2626–2632 (2007).

    PubMed  Google Scholar 

  245. Wiffen, P. J. et al. Gabapentin for chronic neuropathic pain in adults. Cochrane Database Syst. Rev. 6, CD007938 (2017).

    PubMed  Google Scholar 

  246. Alam, U., Sloan, G. & Tesfaye, S. Treating pain in diabetic neuropathy: current and developmental drugs. Drugs 80, 363–384 (2020).

    CAS  PubMed  Google Scholar 

  247. Rosenstock, J., Tuchman, M., LaMoreaux, L. & Sharma, U. Pregabalin for the treatment of painful diabetic peripheral neuropathy: a double-blind, placebo-controlled trial. Pain 110, 628–638 (2004).

    CAS  PubMed  Google Scholar 

  248. Derry, S. et al. Pregabalin for neuropathic pain in adults. Cochrane Database Syst. Rev. 1, CD007076 (2019).

    PubMed  Google Scholar 

  249. Tesfaye, S. et al. Painful diabetic peripheral neuropathy: consensus recommendations on diagnosis, assessment and management. Diabetes Metab. Res. Rev. 27, 629–638 (2011).

    CAS  PubMed  Google Scholar 

  250. Max, M. B. et al. Amitriptyline relieves diabetic neuropathy pain in patients with normal or depressed mood. Neurology 37, 589–596 (1987).

    CAS  PubMed  Google Scholar 

  251. Max, M. B. et al. Effects of desipramine, amitriptyline, and fluoxetine on pain in diabetic neuropathy. N. Engl. J. Med. 326, 1250–1256 (1992).

    CAS  PubMed  Google Scholar 

  252. Boyle, J. et al. Randomized, placebo-controlled comparison of amitriptyline, duloxetine, and pregabalin in patients with chronic diabetic peripheral neuropathic pain: impact on pain, polysomnographic sleep, daytime functioning, and quality of life. Diabetes Care 35, 2451–2458 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  253. Morello, C. M., Leckband, S. G., Stoner, C. P., Moorhouse, D. F. & Sahagian, G. A. Randomized double-blind study comparing the efficacy of gabapentin with amitriptyline on diabetic peripheral neuropathy pain. Arch. Intern. Med. 159, 1931–1937 (1999).

    CAS  PubMed  Google Scholar 

  254. Moore, R. A., Derry, S., Aldington, D., Cole, P. & Wiffen, P. J. Amitriptyline for neuropathic pain in adults. Cochrane Database Syst. Rev 7, CD008242 (2015).

    Google Scholar 

  255. Lunn, M. P., Hughes, R. A. & Wiffen, P. J. Duloxetine for treating painful neuropathy, chronic pain or fibromyalgia. Cochrane Database Syst. Rev. 1, CD007115 (2014).

    Google Scholar 

  256. Gallagher, H. C., Gallagher, R. M., Butler, M., Buggy, D. J. & Henman, M. C. Venlafaxine for neuropathic pain in adults.Cochrane Database Syst. Rev. 8, CD011091 (2015).

    Google Scholar 

  257. Rowbotham, M. C., Goli, V., Kunz, N. R. & Lei, D. Venlafaxine extended release in the treatment of painful diabetic neuropathy: a double-blind, placebo-controlled study. Pain 110, 697–706 (2004).

    CAS  PubMed  Google Scholar 

  258. Gilron, I. et al. Morphine, gabapentin, or their combination for neuropathic pain. N. Engl. J. Med. 352, 1324–1334 (2005).

    CAS  PubMed  Google Scholar 

  259. Tesfaye, S. et al. Duloxetine and pregabalin: high-dose monotherapy or their combination? The “COMBO-DN study”–a multinational, randomized, double-blind, parallel-group study in patients with diabetic peripheral neuropathic pain. Pain 154, 2616–2625 (2013).

    CAS  PubMed  Google Scholar 

  260. Vowles, K. E. et al. Rates of opioid misuse, abuse, and addiction in chronic pain: a systematic review and data synthesis. Pain 156, 569–576 (2015).

    PubMed  Google Scholar 

  261. Bohnert, A. S. B. et al. Association between opioid prescribing patterns and opioid overdose-related deaths. JAMA 305, 1315–1321 (2011).

    CAS  PubMed  Google Scholar 

  262. Finnerup, N. B. Nonnarcotic methods of pain management. N. Engl. J. Med. 380, 2440–2448 (2019).

    CAS  PubMed  Google Scholar 

  263. Duehmke, R. M. et al. Tramadol for neuropathic pain in adults. Cochrane Database Syst. Rev. 6, CD003726 (2017).

    PubMed  Google Scholar 

  264. Gaskell, H., Derry, S., Stannard, C. & Moore, R. A. Oxycodone for neuropathic pain in adults. Cochrane Database Syst. Rev. 7, CD010692 (2016).

    PubMed  Google Scholar 

  265. Vadivelu, N. et al. Tapentadol extended release in the management of peripheral diabetic neuropathic pain. Ther. Clin. Risk Manag. 11, 95–105 (2015).

    PubMed  PubMed Central  Google Scholar 

  266. Noble, M. et al. Long-term opioid management for chronic noncancer pain. Cochrane Database Syst. Rev. 1, CD006605 (2010).

    Google Scholar 

  267. Balhara, Y. P. S., Singh, S. & Kalra, S. Pragmatic opioid use in painful diabetic neuropathy. Eur. Endocrinol. 16, 21–24 (2020).

    PubMed  PubMed Central  Google Scholar 

  268. van Nooten, F., Treur, M., Pantiri, K., Stoker, M. & Charokopou, M. Capsaicin 8% patch versus oral neuropathic pain medications for the treatment of painful diabetic peripheral neuropathy: a systematic literature review and network meta-analysis. Clin. Ther. 39, 787–803.e18 (2017).

    PubMed  Google Scholar 

  269. Viola, V., Newnham, H. H. & Simpson, R. W. Treatment of intractable painful diabetic neuropathy with intravenous lignocaine. J. Diabetes Complications 20, 34–39 (2006).

    PubMed  Google Scholar 

  270. Moulin, D. E., Morley-Forster, P. K., Pirani, Z., Rohfritsch, C. & Stitt, L. Intravenous lidocaine in the management of chronic peripheral neuropathic pain: a randomized-controlled trial. Can. J. Anaesth. 66, 820–827 (2019).

    CAS  PubMed  Google Scholar 

  271. Tesfaye, S. et al. Electrical spinal-cord stimulation for painful diabetic peripheral neuropathy. Lancet 348, 1698–1701 (1996).

    CAS  PubMed  Google Scholar 

  272. Pluijms, W. A. et al. Electrical spinal cord stimulation in painful diabetic polyneuropathy, a systematic review on treatment efficacy and safety. Eur. J. Pain. 15, 783–788 (2011).

    PubMed  Google Scholar 

  273. Petersen, E. A. et al. Effect of high-frequency (10-kHz) spinal cord stimulation in patients with painful diabetic neuropathy: a randomized clinical trial. JAMA Neurol. https://doi.org/10.1001/jamaneurol.2021.0538 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  274. Selvarajah, D. et al. Multicentre, double-blind, crossover trial to identify the Optimal Pathway for TreatIng neurOpathic paiN in Diabetes Mellitus (OPTION-DM): study protocol for a randomised controlled trial. Trials 19, 578 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  275. Javed, S., Alam, U. & Malik, R. A. Mirogabalin and emerging therapies for diabetic neuropathy. J. Pain. Res. 11, 1559–1566 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  276. Baba, M., Matsui, N., Kuroha, M., Wasaki, Y. & Ohwada, S. Mirogabalin for the treatment of diabetic peripheral neuropathic pain: A randomized, double-blind, placebo-controlled phase III study in Asian patients. J. Diabetes Investig. 10, 1299–1306 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  277. Emery, E. C., Luiz, A. P. & Wood, J. N. Nav1.7 and other voltage-gated sodium channels as drug targets for pain relief. Expert Opin. Ther. Targets 20, 975–983 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  278. Kingwell, K. Nav1.7 withholds its pain potential. Nat. Rev. Drug Discov. 18, 321–323 (2019).

    Google Scholar 

  279. Calandre, E. P., Morillas-Arques, P., Molina-Barea, R., Rodriguez-Lopez, C. M. & Rico-Villademoros, F. Trazodone plus pregabalin combination in the treatment of fibromyalgia: a two-phase, 24-week, open-label uncontrolled study. BMC Musculoskelet. Disord. 12, 95 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  280. Tesfaye, S. PL37: a new hope in the treatment of painful diabetic neuropathy? Pain. Manag. 6, 129–132 (2016).

    PubMed  Google Scholar 

  281. Thibault, K. et al. Antinociceptive and anti-allodynic effects of oral PL37, a complete inhibitor of enkephalin-catabolizing enzymes, in a rat model of peripheral neuropathic pain induced by vincristine. Eur. J. Pharmacol. 600, 71–77 (2008).

    CAS  PubMed  Google Scholar 

  282. Bernier, L. P., Ase, A. R. & Séguéla, P. P2X receptor channels in chronic pain pathways. Br. J. Pharmacol. 175, 2219–2230 (2018).

    CAS  PubMed  Google Scholar 

  283. Brines, M. et al. ARA 290, a nonerythropoietic peptide engineered from erythropoietin, improves metabolic control and neuropathic symptoms in patients with type 2 diabetes. Mol. Med. 20, 658–666 (2015).

    PubMed  PubMed Central  Google Scholar 

  284. Demant, D. T. et al. The effect of oxcarbazepine in peripheral neuropathic pain depends on pain phenotype: a randomised, double-blind, placebo-controlled phenotype-stratified study. Pain 155, 2263–2273 (2014).

    CAS  PubMed  Google Scholar 

  285. Bouhassira, D. et al. Neuropathic pain phenotyping as a predictor of treatment response in painful diabetic neuropathy: data from the randomized, double-blind, COMBO-DN study. Pain 155, 2171–2179 (2014).

    PubMed  Google Scholar 

  286. Yarnitsky, D., Granot, M., Nahman-Averbuch, H., Khamaisi, M. & Granovsky, Y. Conditioned pain modulation predicts duloxetine efficacy in painful diabetic neuropathy. Pain 153, 1193–1198 (2012).

    CAS  PubMed  Google Scholar 

  287. Baron, R. et al. Peripheral neuropathic pain: a mechanism-related organizing principle based on sensory profiles. Pain 158, 261–272 (2017).

    PubMed  Google Scholar 

  288. Johnson-Lynn, S. E., McCaskie, A. W., Coll, A. P. & Robinson, A. H. N. Neuroarthropathy in diabetes: pathogenesis of Charcot arthropathy. Bone Jt. Res. 7, 373–378 (2018).

    CAS  Google Scholar 

  289. Jeffcoate, W. J., Game, F. & Cavanagh, P. R. The role of proinflammatory cytokines in the cause of neuropathic osteoarthropathy (acute Charcot foot) in diabetes. Lancet 366, 2058–2061 (2005).

    CAS  PubMed  Google Scholar 

  290. Cameron, N. E., Eaton, S. E., Cotter, M. A. & Tesfaye, S. Vascular factors and metabolic interactions in the pathogenesis of diabetic neuropathy. Diabetologia 44, 1973–1988 (2001).

    CAS  PubMed  Google Scholar 

  291. Shillo, P. et al. Reduced vitamin D levels in painful diabetic peripheral neuropathy. Diabet. Med. 36, 44–51 (2019).

    CAS  PubMed  Google Scholar 

  292. Nathan, D. M. et al. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. N. Engl. J. Med. 329, 977–986 (1993).

    CAS  PubMed  Google Scholar 

  293. Spallone, V. Update on the impact, diagnosis and management of cardiovascular autonomic neuropathy in diabetes: what is defined, what is new, and what is unmet. Diabetes Metab. J. 43, 3–30 (2019).

    PubMed  Google Scholar 

  294. Freeman, R. Clinical practice. Neurogenic orthostatic hypotension. N. Engl. J. Med. 358, 615–624 (2008).

    CAS  PubMed  Google Scholar 

  295. Camilleri, M. et al. Gastroparesis. Nat. Rev. Dis. Prim. 4, 41 (2018).

    PubMed  Google Scholar 

  296. Vinik, A. I., Maser, R. E., Mitchell, B. D. & Freeman, R. Diabetic autonomic neuropathy. Diabetes Care 26, 1553–1579 (2003).

    PubMed  Google Scholar 

  297. Brown, J. S. et al. Urologic complications of diabetes. Diabetes Care 28, 177–185 (2005).

    PubMed  Google Scholar 

  298. Maiorino, M. I., Bellastella, G. & Esposito, K. Diabetes and sexual dysfunction: current perspectives. Diabetes Metab. Syndr. Obes. 7, 95–105 (2014).

    PubMed  PubMed Central  Google Scholar 

  299. Kempler, P. et al. Management strategies for gastrointestinal, erectile, bladder, and sudomotor dysfunction in patients with diabetes. Diabetes Metab. Res. Rev. 27, 665–677 (2011).

    CAS  PubMed  Google Scholar 

  300. Greco, D., Gambina, F., Pisciotta, M., Abrignani, M. & Maggio, F. Clinical characteristics and associated comorbidities in diabetic patients with cranial nerve palsies. J. Endocrinol. Invest. 35, 146–149 (2012).

    CAS  PubMed  Google Scholar 

  301. Laughlin, R. S. & Dyck, P. J. Diabetic radiculoplexus neuropathies. Handb. Clin. Neurol. 126, 45–52 (2014).

    PubMed  Google Scholar 

  302. Selvarajah D., Sloan G. & Tesfaye S. in The Foot In Diabetes 5th Edn 89–104 (Wiley, 2020).

  303. Rota, E. & Morelli, N. Entrapment neuropathies in diabetes mellitus. World J. Diabetes 7, 342–353 (2016).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank M. Yorek (University of Iowa, USA) for his advice on the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to Solomon Tesfaye.

Ethics declarations

Competing interests

S.T. has received honoraria (speaker fees) from Abbott, Astellas Pharma, AstraZeneca, Eva Pharma, Grunenthal, Hikma, Impeto Medical, Merk, Novo Nordisk and Pfizer. S.T. is on, or has been on, the Advisory Board of Angelini, Bayer, Mitsubushi Tanabe Pharma, Nervo, Trigocare International and Worwag Pharma. G.S. and D.S. declare no competing interests.

Additional information

Peer review information

Nature Reviews Endocrinology thanks D. Armstrong, who co-reviewed with L. Shin; R. Malik; A. Veves; and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sloan, G., Selvarajah, D. & Tesfaye, S. Pathogenesis, diagnosis and clinical management of diabetic sensorimotor peripheral neuropathy. Nat Rev Endocrinol 17, 400–420 (2021). https://doi.org/10.1038/s41574-021-00496-z

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41574-021-00496-z

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing