Elsevier

Clinical Neurophysiology

Volume 124, Issue 11, November 2013, Pages 2079-2090
Clinical Neurophysiology

Review
Mechanisms of axonal dysfunction in diabetic and uraemic neuropathies

https://doi.org/10.1016/j.clinph.2013.04.012Get rights and content

Highlights

  • Nerve excitability techniques have provided important insights into the mechanisms underlying axonal dysfunction in diabetic and uraemic neuropathy.

  • Excitability studies in diabetes have suggested that axonal ion channel dysfunction may contribute to the development of neuropathic symptoms.

  • Membrane depolarization due to hyperkalemia may underlie the development of uraemic neuropathy.

Abstract

The global burden imposed by metabolic diseases and associated complications continue to escalate. Neurological complications, most commonly peripheral neuropathy, represent a significant cause of morbidity and disability in patients with diabetes and chronic kidney disease. Furthermore, health care costs are substantially increased by the presence of complications making investigation into treatment a matter of high priority. Over the last decade nerve excitability techniques have entered the clinical realm and enabled in vivo assessment of biophysical properties and function of peripheral nerves in health and disease. Studies of excitability in diabetic neuropathy have demonstrated alteration in biophysical properties, including changes in Na+ conductances and Na+/K+ pump function, which may contribute to the development of neuropathic symptoms. Interventional studies have demonstrated that these changes are responsive to pharmacological agents. Excitability studies in patients with chronic kidney disease have demonstrated prominent changes that may contribute to the development of uraemic neuropathy. In particular, these studies have demonstrated strong correlation between hyperkalaemia and the development of nerve dysfunction. These studies have provided a basis for future work assessing the benefits of potassium restriction as a therapeutic strategy in this condition.

Introduction

The prevalence and burden of metabolic disorders continues to escalate worldwide, largely due to the increasing rates of diabetes (Atkins and Zimmet, 2010). The global prevalence of diabetes is estimated to climb from 366 million in 2011 to 552 million in 2030 (Whiting et al., 2011). Furthermore, the presence of complications in diabetes has been demonstrated to result in a substantial increase to health costs and to have a marked impact on patient quality of life (Boulton et al., 2005, Gordois et al., 2003, Happich et al., 2008). In addition to causing macrovascular disease, diabetes is associated with significant microvascular complications, including neuropathy, nephropathy and retinopathy. From a neurological perspective, neuropathy remains the most prevalent long-term complication of diabetes, affecting >50% of patients with long standing diabetes (Dyck et al., 1993, Pirart, 1978). In addition to causing neuropathy, diabetes is now the leading cause of chronic kidney disease in both developed and developing nations (Atkins and Zimmet, 2010). The implications of this are significant from a neurological perspective, given that chronic kidney disease on its own causes peripheral neuropathy, also known as uraemic neuropathy, in >90% of patients with severe chronic kidney disease (Laaksonen et al., 2002, Tilki et al., 2009, Van den Neucker et al., 1998).

Metabolic polyneuropathies due to diabetes and chronic kidney disease share similar clinical features, typically manifesting as a progressive, symmetrical, length dependent neuropathy that commences in lower limb nerves. In its initial stages, patients present with sensory symptoms with motor involvement occurring in more severe cases (Krishnan et al., 2009b). Consequently, early symptoms include pain, paraesthesia and numbness in the distal lower limbs, and clinical examination reveals loss of distal sensory function and a reduction in ankle deep tendon reflexes. With disease progression distal motor involvement may develop resulting in muscle atrophy and weakness.

Neurophysiological techniques have provided important insights into the mechanisms that underlie nerve dysfunction in metabolic neuropathies. Early neurophysiological studies utilised nerve conduction methods to assess important nerve parameters, including action potential amplitude and latency. While these methods continue to provide important diagnostic information regarding neuropathy, they provide only limited information regarding pathophysiological mechanisms that may underlie the development of axonal dysfunction in these disorders.

Over the last decade, nerve excitability methods have been developed as clinical techniques for the investigation of the pathophysiological processes involved in the development of peripheral neuropathy. These methods enable the indirect assessment of axonal membrane potential and the activity of ion channels, energy dependent pumps and ion exchange processes involved in impulse conduction in peripheral nerve axons (Bostock et al., 1998, Krishnan et al., 2009a). In recent years these techniques have enabled the assessment of biophysical properties and function of peripheral nerves in health and disease (Krishnan et al., 2009a). While studies of excitability have been undertaken in inflammatory, toxic, metabolic and degenerative disorders of peripheral nerve (Cheah et al., 2012, Krishnan et al., 2006b, Park et al., 2009), it may be argued that the greatest clinical mileage has been gained in the investigation of metabolic neuropathies, particularly those relating to diabetes and chronic kidney disease. This review will focus on recent studies of nerve excitability that have been undertaken in diabetic and uraemic neuropathies. The Review will address the potential mechanisms of axonal dysfunction in these disorders and the clinical relevance of these findings in terms of the development of future diagnostic methods and novel treatments.

The concept of nerve excitability assessment as a method of investigating axonal function was pioneered by the work of Bergmans, who demonstrated that changes in the ‘threshold’ or current required to elicit a target response reflected physiological alterations of the axon (Bergmans, 1970). Bergmans discovered that by measuring changes in the threshold of axons induced by impulse activity or artificial polarisation, considerable information about axonal physiology could be inferred (Bergmans, 1970, Bostock et al., 1998). The recent development of automated excitability software has enabled the rapid acquisition of multiple excitability parameters in a short period of time (Kiernan et al., 2000). These developments have resulted in significant clinical translation, with studies of excitability having been conducted in a range of neurological disorders (Krishnan et al., 2009a).

Automated excitability protocols consist of four discrete testing paradigms; strength-duration time constant, threshold electrotonus, the current-threshold relationship and the recovery cycle.

Strength-duration behaviour is calculated from the relationship between the stimulus intensity and its duration (Fig. 1A). The strength duration properties of the nerve reflect the properties of the nodal membrane (Mogyoros et al., 1996). Specifically, strength-duration time constant provides an assessment of nodal persistent Na+ currents (Bostock and Rothwell, 1997). Though these currents only contribute to ∼2.5% of the total Na+ current, these conductances have an important role in repetitive and spontaneous activity, making this measurement a clinically relevant excitability parameter (Kiernan et al., 2000). However, the strength-duration time constant may also be influenced by passive membrane properties and membrane potential; as such measurements of latent addition may help separate persistent Na+ current from the passive nodal time constant (Nodera and Kaji, 2006). Latent addition uses subthreshold polarising currents of short duration to enable an in depth assessment of axonal strength duration properties (Bostock and Rothwell, 1997).

Threshold electrotonus (TE) is assessed by measuring the change in threshold at multiple time points during and after prolonged (100 ms) subthreshold, depolarizing (TEd) and hyperpolarizing (TEh) conditioning currents (Fig. 1B). Although these currents are not sufficient to produce an action potential, they enable a slow spread of current under the myelin sheath into the internode, consequently altering the membrane potential and activating a number of internodal accommodative conductances (Baker et al., 1987, Bostock et al., 1998, Burke et al., 2001).

Current/threshold relationship (I/V) assesses the effects of longer duration polarising currents by measuring the change in threshold following polarising currents of 200 ms duration, ranging from +50% (depolarising) to −100% (hyperpolarising) of the unconditioned threshold intensity (Fig 1C). This parameter is particularly useful for examining the rectifying properties of the axon (Kiernan et al., 2000). Strong depolarising currents result in outward rectification achieved by activation of fast and slow K+ channels (Kiernan and Bostock, 2000). Strong hyperpolarising currents result in activation of inwardly rectifying (IH) conductances (Kiernan et al., 2000, Pape, 1996).

Recovery cycle (RC) measures the changes in threshold that occur over 200 ms following a supramaximal stimulus (Fig. 1D). These parameters provide insights into the behaviour of nodal and juxtaparanodal Na+ and K+ channels. With short conditioning-test intervals, a supramaximal stimulus causes a brief period (0.5–1 ms) of absolute refractoriness, during which the axon is completely inexcitable (Hodgkin and Huxley, 1952). This period is followed by ∼3 ms of relative refractoriness while Na+ channels recover from inactivation. (Hodgkin and Huxley, 1952). During this time the axon is less excitable and an action potential can only be generated with greater than usual stimuli. Following the recovery of Na+ channel inactivation, a passive capacitive charge stored in the internodal membrane produces a depolarising afterpotential, effectively increasing membrane excitability (Barrett and Barrett, 1982, Kiernan et al., 1996). This is referred to as superexcitability and peaks between 5–7 ms following impulse conduction. Finally, there is a second period of reduced excitability due to activation of slow K+ channels at the node of Ranvier during depolarisation (Baker et al., 1987, Schwarz et al., 1995). These channels are slow to deactivate and produce hyperpolarisation as the final stage of the recovery cycle known as subexcitability.

Current excitability software enables the acquisition of these parameters from a single nerve in a ten minute period. This technological advance has enabled detailed assessment of axonal function in a clinical setting, thus providing a valuable tool for clinical translation.

Section snippets

Diabetic neuropathy

The most common neurological complication of diabetes (DM) is the development of a distal symmetric polyneuropathy affecting ∼50% of patients at long-term follow-up (Dyck et al., 1993, Pirart, 1978). Diabetic neuropathy has a profound negative impact on patient morbidity and quality of life (Kwai et al., 2013), with the presence of diabetic neuropathy accounting for 60–70% of diabetic foot ulceration and 50–75% of non-traumatic foot amputations (Gonzalez and Oley, 2000). Patients typically

Uraemic neuropathy

Chronic kidney disease is a significant global health concern with a prevalence of ∼15% in developed countries (Chadban et al., 2003, Couser et al., 2011). Chronic kidney disease encompasses a continuum of disease from mild kidney damage, to end-stage disease which requires renal replacement therapy in the form of dialysis or renal transplantation to sustain life. The aetiology of chronic kidney disease may be due to a primary renal disorder or as a complication of a multisystem disorder

Conclusion

The growing prevalence of diabetes and its associated complications present an ongoing global health concern. Metabolic neuropathies are a major cause of morbidity and disability in patients with diabetes and chronic kidney disease. As such, investigation into the pathophysiology and possible treatment options for these patients remains a matter of high priority. Sensitive neurophysiological techniques play a critical role in this process. Nerve excitability techniques have proven a valuable

References (120)

  • S. Hong et al.

    Early painful diabetic neuropathy is associated with differential changes in tetrodotoxin-sensitive and -resistant sodium channels in dorsal root ganglion neurons in the rat

    J Biol Chem

    (2004)
  • S. Hong et al.

    Altered expression and function of sodium channels in large DRG neurons and myelinated A-fibers in early diabetic neuropathy in the rat

    Biochem Biophys Res Commun

    (2006)
  • A.V. Krishnan et al.

    Axonal ion channels from bench to bedside: a translational neuroscience perspective

    Prog Neurobiol

    (2009)
  • A.V. Krishnan et al.

    Neuropathy, axonal Na+/K+ pump function and activity-dependent excitability changes in end-stage kidney disease

    Clin Neurophysiol

    (2006)
  • S. Kuwabara

    Shortened refractory periods in human diabetic neuropathy

    Clin Neurophysiol

    (2003)
  • S. Misawa et al.

    Axonal potassium conductance and glycemic control in human diabetic nerves

    Clin Neurophysiol

    (2005)
  • S. Misawa et al.

    Nodal persistent Na+ currents in human diabetic nerves estimated by the technique of latent addition

    Clin Neurophysiol

    (2006)
  • S. Misawa et al.

    Strength-duration properties and glycemic control in human diabetic motor nerves

    Clin Neurophysiol

    (2005)
  • S. Misawa et al.

    Hyperglycemia alters refractory periods in human diabetic neuropathy

    Clin Neurophysiol

    (2004)
  • H. Nodera et al.

    Nerve excitability testing and its clinical application to neuromuscular diseases

    Clin Neurophysiol

    (2006)
  • G. Preswick et al.

    Subclinical polyneuropathy in renal insufficiency

    Lancet

    (1964)
  • K.E. Schemmel et al.

    Aldose reductase inhibitors in the treatment of diabetic peripheral neuropathy: a review

    J Diabetes Complications

    (2010)
  • A.A. Ackil et al.

    Late response and sural conduction studies. Usefulness in patients with chronic renal failure

    Arch Neurol

    (1981)
  • H. Angus-Leppan et al.

    The function of large and small nerve fibers in renal failure

    Muscle Nerve

    (1992)
  • R. Arnold et al.

    Axonal dysfunction prior to neuropathy onset in type 1 diabetes

    Diabetes Metab Res Rev

    (2013)
  • R. Arnold et al.

    Evidence for a causal relationship between nerve dysfucntion and hyperkalaemia in end-stage kidney disease

    Nephrol Dial Transplant

    (2012)
  • R. Arnold et al.

    Effects of hemodiafiltration and high flux hemodialysis on nerve excitability in end-stage kidney disease

    PLoS One

    (2013)
  • A.K. Asbury

    Recovery from uremic neuropathy

    N Engl J Med

    (1971)
  • A.L. Babb et al.

    Bi-directional permeability of the human peritoneum to middle molecules

    Proc Eur Dial Transplant Assoc.

    (1973)
  • M. Baker et al.

    Function and distribution of three types of rectifying channel in rat spinal root myelinated axons

    J Physiol

    (1987)
  • E.F. Barrett et al.

    Intracellular recording from vertebrate myelinated axons: mechanism of the depolarizing after potential

    J Physiol

    (1982)
  • C. Bazzi et al.

    Uremic polyneuropathy: a clinical and electrophysiological study in 135 short- and long-term hemodialyzed patients

    Clin Nephrol

    (1991)
  • F. Behse et al.

    Nerve biopsy and conduction studies in diabetic neuropathy

    J Neurol Neurosurg Psychiatry

    (1977)
  • J. Bergmans

    The physiology of single human nerve fibres

    (1970)
  • A. Bierhaus et al.

    Methylglyoxal modification of Nav1.8 facilitates nociceptive neuron firing and causes hyperalgesia in diabetic neuropathy

    Nat Med

    (2012)
  • E.E. Bittar

    Maia muscle fibre as a model for the study of uraemic toxicity

    Nature

    (1967)
  • H. Bostock et al.

    Threshold tracking techniques in the study of human peripheral nerve

    Muscle Nerve

    (1998)
  • H. Bostock et al.

    Latent addition in motor and sensory fibres of human peripheral nerve

    J Physiol

    (1997)
  • H. Bostock et al.

    Has potassium been prematurely discarded as a contributing factor to the development of uraemic neuropathy?

    Nephrol Dial Transplant

    (2004)
  • T. Brismar et al.

    Reversible and irreversible nodal dysfunction in diabetic neuropathy

    Ann Neurol

    (1987)
  • J. Carracedo et al.

    On-line hemodiafiltration reduces the proinflammatory CD14+CD16+ monocyte-derived dendritic cells: a prospective, crossover study

    J Am Soc Nephrol

    (2006)
  • S.J. Chadban et al.

    Prevalence of kidney damage in Australian adults: the AusDiab kidney study

    J Am Soc Nephrol

    (2003)
  • Chalk C, Benstead TJ, Moore F. Aldose reductase inhibitors for the treatment of diabetic polyneuropathy. Cochrane...
  • M.J. Craner et al.

    Changes of sodium channel expression in experimental painful diabetic neuropathy

    Ann Neurol

    (2002)
  • M.L. D’Amour et al.

    Sensory nerve conduction in chronic uremic patients during the first six months of hemodialysis

    Can J Neurol Sci

    (1984)
  • P.J. Dyck et al.

    Segmental demyelination secondary to axonal degeneration in uremic neuropathy

    Mayo Clin Proc

    (1971)
  • P.J. Dyck et al.

    The spatial distribution of fiber loss in diabetic polyneuropathy suggests ischemia

    Ann Neurol

    (1986)
  • P.J. Dyck 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

    (1993)
  • V. Filiopoulos et al.

    Inflammation and oxidative stress in patients on hemodiafiltration

    Am J Nephrol

    (2008)
  • M.J. Gillett

    International expert committee report on the role of the A1c assay in the diagnosis of diabetes

    Diabetes Care

    (2009)
  • Cited by (29)

    • Renal hemodynamic dysfunction and neuropathy in longstanding type 1 diabetes: Results from the Canadian study of longevity in type 1 diabetes

      2022, Journal of Diabetes and its Complications
      Citation Excerpt :

      Both, CKD and diabetes, are independent risk factors for peripheral neuropathy 38–40. Recent studies suggest that hyperkalemia may be a contributing factor to development of neuropathy in patients with kidney disease 41,42, with limited clinical trials suggesting that potassium restriction is neuroprotective 43. In most patients, however, overt hyperkalemia is less likely to occur until GFR is below 10 mL/min/1.73 m2 and thus, hyperkalemia was unlikely to contribute to our findings in participants with longstanding type 1 diabetes and an average GFR of 102.8 ± 17.1 mL/min/1.73 m2 with the DKD non-resistor group still having preserved kidney function.

    • Corneal nerve fiber loss in diabetes with chronic kidney disease

      2020, Ocular Surface
      Citation Excerpt :

      CKD and diabetes together can amplify the prevalence and severity of peripheral neuropathy, as both conditions are major and independent risk factors for the development of peripheral neuropathy [4,7,8]. Recent work has suggested that hyperkalemia may underlie the development of neuropathy in CKD [9,10] and a recent clinical trial has demonstrated that potassium restriction is neuroprotective [11] in people with both diabetic and non-diabetic CKD. Given these results, early detection of nerve injury is critical in patients with impaired renal function.

    • Haemodialysis alters peripheral nerve morphology in end-stage kidney disease

      2017, Clinical Neurophysiology
      Citation Excerpt :

      Investigation of contributing serum metabolites revealed a strong correlation between changes in nerve morphology and K+ concentration. Previous studies have demonstrated that axons of ESKD patients are in a depolarized state (Kiernan et al.; 2000, 2002; Arnold et al., 2013c) and a recent interventional study demonstrated that hyperkalaemia was likely to be the underlying cause of the altered nerve membrane potential (Arnold et al., 2014). It has been hypothesized that prolonged membrane depolarization may induce morphological changes (Baker, 2002) by causing swelling of the node of Ranvier and myelin sheath and expansion of the periaxonal space, reflecting the movement of water underneath and within the myelin sheath (Baker, 2002; Love and Cruz-Höfling, 1986; Love et al., 1986).

    • Nerve excitability in the rat forelimb: a technique to improve translational utility

      2017, Journal of Neuroscience Methods
      Citation Excerpt :

      Nerve excitability studies have advanced the technical utility of electrophysiology from providing only basic measures of amplitude, velocity and late responses, to enabling the indirect assessment of ion channels, pumps, exchangers and membrane potential (Krarup and Moldovan, 2009; Krishnan et al., 2009). These studies have demonstrated the potential of nerve excitability studies as a biomarker in several neurological diseases (Park et al., 2009; Sung et al., 2012; Arnold et al., 2013), however, important mechanistic and validation questions are yet to be clarified. Experimental studies in animal models of neurological diseases provide an opportunity to directly assess the validity of interpretations made using these indirect assessments.

    View all citing articles on Scopus
    View full text