Skip to main content
Erschienen in: Current Diabetes Reports 6/2019

Open Access 01.06.2019 | Microvascular Complications—Neuropathy (R Pop-Busui, Section Editor)

Painful and Painless Diabetic Neuropathies: What Is the Difference?

verfasst von: Pallai Shillo, Gordon Sloan, Marni Greig, Leanne Hunt, Dinesh Selvarajah, Jackie Elliott, Rajiv Gandhi, Iain D. Wilkinson, Solomon Tesfaye

Erschienen in: Current Diabetes Reports | Ausgabe 6/2019

Abstract

Purpose of Review

The prevalence of diabetes mellitus and its chronic complications are increasing to epidemic proportions. This will unfortunately result in massive increases in diabetic distal symmetrical polyneuropathy (DPN) and its troublesome sequelae, including disabling neuropathic pain (painful-DPN), which affects around 25% of patients with diabetes. Why these patients develop neuropathic pain, while others with a similar degree of neuropathy do not, is not clearly understood. This review will look at recent advances that may shed some light on the differences between painful and painless-DPN.

Recent Findings

Gender, clinical pain phenotyping, serum biomarkers, brain imaging, genetics, and skin biopsy findings have been reported to differentiate painful- from painless-DPN.

Summary

Painful-DPN seems to be associated with female gender and small fiber dysfunction. Moreover, recent brain imaging studies have found neuropathic pain signatures within the central nervous system; however, whether this is the cause or effect of the pain is yet to be determined. Further research is urgently required to develop our understanding of the pathogenesis of pain in DPN in order to develop new and effective mechanistic treatments for painful-DPN.
Hinweise
Pallai Shillo and Gordon Sloan are joint first authors
This article is part of the Topical Collection on Microvascular Complications—Neuropathy

Publisher’s Note

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

Introduction

The worldwide prevalence of diabetes mellitus (DM) has reached epidemic proportions, and is set to increase to 629 million by 2045 [1]. Rising population growth, aging, urbanization, and an increased prevalence of obesity and physical inactivity are amongst the major contributing factors. Diabetic neuropathies are one of the most common chronic complications of DM [2], and distal symmetrical polyneuropathy (DPN) is the most prevalent form of diabetic neuropathy, which may affect up to 50% of patients [2, 3•, 4•]. The Toronto Expert Group has defined DPN as “a symmetrical, length dependent sensorimotor polyneuropathy attributable to metabolic and micro-vessel alterations as a result of chronic hyperglycaemia exposure and cardiovascular risk covariates” [5•]. A more recent definition of DPN in the American Diabetes Association Position Statement is “the presence of symptoms and/or signs of peripheral nerve dysfunction in people with diabetes after the exclusion of other causes” [3•]. The rising numbers of patients diagnosed with neuropathic disorders related to DM will have an immense impact on health and social care provision [6].
DPN is a major risk factor for diabetic foot ulceration, which remains a major cause of morbidity and is the leading cause of non-traumatic amputations [7]. Although a large number of patients with DPN may be entirely asymptomatic, approximately 15–25% of people with DM present with neuropathic pain (painful-DPN) [811, 12•, 13]. The neuropathic pain is of varying degree of intensity [14] DPN and painful-DPN has different clinical syndromes with the most common of which is a mixed large and small fiber neuropathy. Small nerve-fibers (SF) are small-caliber sensory fibers, which are primarily responsible for peripheral nociception [15]. Pure SF neuropathy may occur in DM and the clinical features include symptoms of painful peripheral neuropathy with signs of SF impairment (e.g., pinprick or thermal hypoalgesia or allodynia) in a peripheral neuropathy distribution in the absence of large fiber impairment (e.g., impaired light touch, vibration, proprioception or motor signs).
Painful-DPN often results in insomnia, mood disorders, and a poor quality of life [12•]. The currently available therapies for the pain associated with DPN remain inadequate, given relatively modest pain relief and often troublesome side effects [3•, 16, 17]. There is thus an urgent need to have a better understanding of the pathogenesis of pain in DPN and this has been the subject of a recent review (Fig. 1) [17]. Central to this understanding will be to develop new insights as to why some patients develop disabling neuropathic symptoms while others with a similar degree of neuropathy do not. This review will discuss the differences in risk factors, clinical features, serum biomarkers, vascular alterations, quantitative sensory testing (QST), skin biopsy parameters, genetics, and brain imaging studies between painful- and painless-DPN.

Risk Factors

Several risk factors for DPN in general have been described and confirmed in cohorts of type 1 and type 2 diabetes. The EURODIAB Prospective Complications Study screened 3250 type 1 DM patients at baseline and followed 1172 patients without DPN looking for risk factors that predicted the development of DPN [4•]. The study found that in addition to glycemic control, traditional vascular risk factors such as hypertension, raised triglycerides, obesity, and cigarette smoking were independent risk factors for the development of new onset DPN. Similar vascular risk factors were also found in T2DM [18•, 19, 20]. However, the risk factors for neuropathic pain in DM are less well known. This is partly because of the wide variation in the diagnostic and population selection methods employed by the epidemiological studies for painful-DPN [21••, 22]. The reported risk factors include increasing age [9, 10], elevated HbA1c [23••, 24], duration of DM [9], and obesity [10, 25]. A high alcohol intake, type of diabetes, macro and microvascular disease, and ethnicity have also been implicated [21••]. Recent large studies have also suggested nephropathy and female gender as risk factors for painful-DPN [26, 27••, 28••]. Indeed, female gender was the only risk factor identified in a large cross-sectional study (n = 816) performed by Truini et al. which diagnosed painful-DPN using widely agreed criteria [28••]. Thirteen percent were diagnosed with painful-DPN and the only distinguishing risk factor from painless-DPN was female gender. Gender differences are well recognized in chronic pain conditions and neuropathic pain intensity has previously been reported to be more severe in females [29, 30].
Recent advances in gene sequencing technology have led to several studies examining genetic variants associated with DPN and painful-DPN [3134, 35•, 36]. Two recent studies by Meng et al. conducted genome-wide association studies in Tayside, Scotland [32, 33]. Chr8p21.3, Chr1p35.1, and Chr8p21.3 polymorphisms were associated with neuropathic pain. However, the study did not use validated diagnostic criteria for painful-DPN. Recently also, there has been great interest in the role of voltage-gated sodium channels and their role in neuropathic pain. The Nav 1.7 sodium channel is well recognized to be involved in pain signaling and “gain of function” mutations of its encoding gene, SCN9A, cause rare pain disorders. Additionally, studies have identified Nav 1.7 mutations in idiopathic small fiber neuropathy [36] and painful-DPN [34]. Blesneac et al. looked at the relationship between Nav 1.7 variants and painful-DPN and found that none of the participants with painless-DPN (n = 78) were found to have a genetic variant [35•]. However, a total of 12 rare Nav 1.7 variants were identified in 10 out of 111 patients with painful-DPN. The subjects with these variants were found to have a shorter duration of diabetes yet more severe burning pain. Painful-DPN is a heterogeneous condition and subjects with rare sodium channel gene variants may represent a subgroup that may respond to a particular treatment.
In summary, while the risk factors for DPN are well recognized, those for painful-DPN are less certain. This might indicate the complexity of painful-DPN as many factors including genetics, cultural, psycho-social, and gender may be involved.

Clinical Features

Neuropathic pain in diabetes has distinct presentations as burning, sharp, aching, electric, and evoked pains [37]. However, patients may also describe symptoms of numbness, tingling, and pins and needles, irrespective of the presence of pain. Neuropathic pain may also induce various degrees of physical disability, depression, anxiety, insomnia, and a poorer quality of life than patients with painless-DPN, particularly with moderate to severe neuropathic pain [23••, 27••, 38]. Despite these profound differences in a patient’s clinical presentation, there are few distinct differences in the neurological examination between painless- and painful-DPN. The majority of patients with painful-DPN demonstrate sensory loss on clinical examination but a small proportion of patients with painful-DPN have evidence of “gain of function” signs such as allodynia and hyperalgesia [23••]. There is controversy regarding whether the severity of neuropathic impairment is greater in painful-DPN. Several studies have reported a correlation between neuropathy severity and the presence and/or severity of neuropathic pain in DPN [8, 11, 15, 23••, 3941] whereas other studies have not [18•, 28••]. Although the weight of evidence seems to suggest that an increasing severity of DPN may increase the risk of developing painful neuropathic symptoms, severe DPN and pain are not mutually exclusive, and there may have been a selection bias in recruiting painful-DPN patients from tertiary referral centers.

Cardiovascular Autonomic Neuropathy

Both autonomic neuropathy and painful-DPN involve small fibers, and a potential relationship was therefore investigated. In a small study, we demonstrated greater changes in heart rate variability studies, as measures of cardiovascular autonomic neuropathy (CAN) in subjects with painful- compared with painless-DPN [42], while other small studies reported that painful DPN was more likely to be associated with the absence of a nocturnal fall in blood pressure (“non-dipping”) [43], or with reduced Valsalva ratio [40]. However, these are in contrast with other studies that have not found any differences in measures of CAN between painful and painless-DPN [39, 44, 45].

Diagnostic Methods of Painful-DPN

Conventional neurophysiological testing methods, which measure large fiber function, such as nerve conduction studies (NCS), cannot detect pure small fiber neuropathy (SFN) [46]. However, QST and more recent advances in diagnostic techniques, e.g., skin biopsy with intraepidermal nerve fiber density (IENFD) quantitation, corneal confocal microscopy (CCM), and laser Doppler imaging flare (LDI Flare) have allowed the reliable diagnosis of SFN [5•, 46]. Because of their role in physiological nociception, studies have explored whether damage or alterations in SF may relate to neuropathic pain in DPN.

Skin Biopsy

Immunostaining of skin biopsy samples with protein gene product 9.5 and quantitation of IENFD is a reliable means of diagnosing SFN [46]. However, IENFD is unable to distinguish between individuals with or without neuropathic pain [23••, 27••, 28••, 47••, 48, 49]. Other studies have been performed to determine whether morphological and functional markers of the epidermal innervation revealed differentiating features. Intraepidermal nerve fiber (IENF) regeneration, by measuring the ratio of growth associated protein-43 (GAP-43) to nerve fibers, has been shown to be enhanced in painful-compared with painless-DPN [49, 50•, 51]. However, Scheytt et al. found no relationship between pain and GAP-43 reactivity in subjects with peripheral neuropathies of varying etiologies [52]. There are contradictory findings in studies investigating other IENF markers to differentiate painful- from painless-DPN including IENF length [50•, 53] and axonal swellings, which are measures of axonal degeneration [49, 54]. Levels within the skin of the neurotrophin nerve growth factor (NGF) were increased in patients with DPN and sensory symptoms, including pain, compared to painless-DPN [55]. NGF has recently been shown to sensitize nociceptors in human skin and it has been hypothesized that the remaining IENF in painful-DPN may be exposed to excessive levels of NGF (“over-trophing”) resulting in hypersensitivity and neuropathic pain [19, 5558].

Corneal Confocal Microscopy

Confocal corneal microscopy (CCM) can rapidly, non-invasively, and accurately image corneal nerves and is a recently developed diagnostic test for DPN [5961]. Studies of CCM have explored the role of corneal innervation and neuropathic pain in DM [53, 62•, 63]. Quattrini et al. reported reduced corneal nerve fiber length with unaltered other CCM measures [53], whereas Marshall et al. found unaltered corneal nerve fiber length but reduced corneal nerve fiber density [62•]. Recently, Kalteniece et al. [63] described significantly lower corneal inferior whorl length, and average and total nerve length in painful- compared to painless-DPN. Changes within this region have been suggested to be indicative of early neuropathic damage. However, there were confounding factors, which could account for these group differences. Therefore, the association of CCM abnormalities to neuropathic pain in DPN is thus far inconclusive.

Evoked Responses

Non-invasive tests have been developed to investigate the peripheral function of SF to diagnose SFN. Such tests can measure evoked potentials in response to stimuli that activate the nociceptive pathway, for example contact heat-evoked potentials (CHEPS) [64]. CHEPs correlates with other measures of SFN including IENFD and leg skin flare responses [65, 66]. One small study found a relationship between enhanced brain CHEP amplitudes in subjects with painful-DPN; this result was most marked in those with thermal hyperalgesia and mechanical allodynia [67].

Quantitative Sensory Testing

QST is a psychophysical measure of the perception of different external stimuli of controlled intensity to assess a range of sensory modalities [68, 69]. Some studies with a relatively small sample size suggested that conventional QST measures of SF function may be statistically different between painful- and painless-DPN [41, 44, 70, 71]. More recent studies have employed the German Research Network on Neuropathic Pain (DFNS) QST protocol to quantify sensory loss, for small and large fiber function, and sensory gain abnormalities [72, 73••]. Three recent large cross-sectional cohort studies have applied this protocol to patients with painful- and painless-polyneuropathies with different etiologies [48] and painful- and painless-DPN [23••, 27••]. In two studies of painful-DPN, DFNS QST revealed more severe loss of function in those with neuropathic pain, particularly patients with moderate/severe pain [23••, 27••]. Thermal hyposensitivity was more severe in painful-DPN whereas mechanical stimuli showed fewer differences compared with painless-DPN. Gain of function abnormalities and preserved SF function with hyperalgesia were both rare. However, Üçeyler et al. studied patients with painful- and painless-polyneuropathies of different etiologies and found that patients with neuropathic pain demonstrated elevated mechanical pain and detection threshold, and lower mechanical pain sensitivity with no difference in SF deficits [48]. This perhaps indicates there may be a unique somatosensory phenotype associated with painful-DPN characterized by more severe SF dysfunction with thermal hyposensitivity [23••, 27••]. However, SF changes are common and can occur in early DPN without pain [7476]; therefore, these findings alone are unable to completely explain why some patients develop neuropathic pain and others do not. Perhaps, other investigations into small fiber function and structure, such as skin biopsy studies, may shed further light onto this paradox.

Pathogenesis of Painful-DPN

Microvascular Blood Flow

Consistent with vascular risk factors increasing the risk of DPN [4•], both structural and functional microvascular abnormalities of the vasa-nervorum have been shown to be involved in the pathogenesis of DPN [7779]. Patients with treatment induced neuropathy of diabetes who had extremely severe neuropathic pain have proliferating blood vessels on the epineurial surface bearing striking similarities to those found in proliferative diabetic retinopathy [80]. It is well recognized that very rapid improvement in glucose control can cause proliferative retinopathy mediated by retinal ischemia and a similar process appears to take place in the peripheral nerve. Furthermore, several studies have shown that regulation of peripheral blood flow is altered in patients with painful- compared with painless-DPN [8184]. Our group demonstrated elevated sural nerve epineurial oxygen saturation and faster blood flow in patients with painful- compared to painless-DPN, perhaps secondary to arteriovenous shunting [82]. Other studies have examined the role of skin microvascular vasodilator and vasoconstrictor responses in subjects with DPN, with contradictory findings [71, 8587].
Studies measuring serum markers of angiogenesis (vascular endothelial growth factor, VEGF) and endothelial dysfunction (soluble intercellular adhesion molecule – 1, sICAM-1) have found them to be elevated in painful-DPN [86, 88•] and symptomatic DPN respectively [89]. Furthermore, punch skin biopsy studies have also indicated that skin microcirculation may be involved in the pathogenesis of painful-DPN. One study demonstrated evidence of hypoxia, by immunostaining with hypoxia inducible factor 1α (HIF-1 α), to be related to pain intensity in subjects with DPN [90]. Recently, our group has also found dermal von Willebrand factor (vWF) immunoreactivity, as a blood vessel marker, to be significantly elevated in subjects with painful-DPN, in comparison to subjects with painless-DPN, patients with DM without DPN and healthy volunteers [56]. Moreover, small studies have demonstrated that pain improves with topical application of vasodilator treatments [91, 92], perhaps indicating that local blood flow dysregulation could be a viable target for the management of pain in DPN.

Hyperglycemia and Downstream Effects

Hyperglycemia mediated metabolic pathways have long been associated in the pathogenesis of DPN, but their role in those with neuropathic pain is less clearly defined. Studies using DM rodent models have found neuropathic pain behaviors to be related to numerous metabolic pathways including the polyol pathway, protein kinase C activity, and increased advanced glycation end-products (AGEs) [93]. However, there is limited evidence to support glycemic control or lifestyle modifications in improving painful neuropathic symptoms [3•]. Moreover, the evidence to support pathogenic treatments for neuropathic pain in DPN has generally been disappointing and only a few pharmacotherapeutic agents are available in select countries [50•].
Methylglyoxal is a highly reactive dicarbonyl compound and is a precursor to the formation of (AGEs). The formation of AGEs has downstream deleterious effects on peripheral nerves and Schwann cells including inflammation and oxidative stress [94]. Methylglyoxal has been suggested to be an important factor in the development of DM and incident DPN [20•, 95]. In rodent models of painful-DPN, methylglyoxal has been shown to induce hyperalgesia via activation of the voltage-gated sodium channel Nav 1.8 and transient receptor potential channel ankyrin-1 [96, 97]. Similarly, in a small number of patients with DM (n = 30), serum methylglyoxal levels were found to be elevated in painful-DPN [96]. In contrast to these findings, a larger study (n = 882) reported methylglyoxal levels to be unrelated to painful-DPN [98]. Although the role of hyperglycemia mediated pathways in generating neuropathic pain is uncertain, pathogenically oriented treatments, particularly anti-oxidants, have been demonstrated to improve pain in some pre-clinical and clinical trials [99, 100].

Vitamin D

Although vitamin D is most commonly recognized for its role in calcium metabolism and bone health, vitamin D is involved in many disparate physiological processes [101]. Deficiency of vitamin D has been shown to be predictive of numerous chronic diseases including DM, DPN, and chronic pain [101104]. Pre-clinical studies indicate that vitamin D appears to play a critical role in nerve function in health and may play a role in neuropathic pain syndromes [105109]. Our group recently found vitamin D levels to be significantly lower in patients with painful- compared to painless-DPN, with a significant correlation between serum 25-hydroxyvitamin D level and pain scores on the Doleur Neuropathique 4 neuropathic pain screening tool [110•]. The study was cross-sectional, and therefore cannot establish a causal relationship, but it does suggest a possible mechanistic link between vitamin D and painful-DPN. Indeed, three non-randomized clinical trials have demonstrated an improvement in painful neuropathic symptoms with vitamin D therapy but, further, larger, adequately powered RCTs are necessary to investigate this further [111113].

Inflammation

Inflammation has been postulated to play a major part in DM and DPN [114]. Low grade inflammation has been suggested as a link between obesity and T2DM, via inflammation induced insulin resistance [114]. Inflammatory chemokine and cytokine production has been reported to be induced by several metabolic pathways implicated in the pathogenesis of DPN [115, 116•]. Multiple studies have demonstrated higher systemic acute-phase proteins, cytokines, and chemokines in DPN [88•, 117•], recently reviewed by Bönhof et al. [118•]. Furthermore, rodent models of neuropathic pain associated with the metabolic syndrome and T2DM demonstrate elevated pro-inflammatory mediator expression in the serum [119] and the dorsal root ganglia [119, 120]. The outcomes of studies examining the association of inflammatory biomarkers and painful-DPN have been variable. Numerous inflammatory markers have been associated with painful-DPN: C-reactive protein (CRP) [86], tumor necrosis factor-α (TNF-α) [121], inducible nitric oxide synthase [121], and interleukin 6 [117•]. Additionally, inflammatory mediators have been shown to differentiate between painful- from painless-neuropathies of various etiologies, including elevated serum IL-2, TNF-α, and reduced anti-inflammatory IL-10 [122]; IL-6 and IL-10 sural nerve biopsy expression [123]; and TNF-α in human Schwann cells [124].

The Central Nervous System

Technological advances in imaging modalities have enabled detailed in vivo investigation of the nervous system in DM. Key differences have been identified within the CNS in painful-DPN, using a variety of different techniques, especially advanced MR imaging modalities.

Spinal Cord Changes in Painful-DPN

We have identified a reduction in the cross-sectional area of the spinal cord in subjects with DPN in comparison to patient with DM without DPN, healthy controls, and disease control subjects with hereditary sensory motor neuropathy type 1A [125]. However, structural differences in the spinal cord area have not been found between subjects with painless- and painful-DPN [125]. Recent studies have indicated that spinal disinhibition, measured using the rate dependent depression (RDD) of the Hoffman reflex (H-reflex), may be a potential biomarker of spinally mediated pain to differentiate painful- from painless-DPN [126]. The RDD has been demonstrated to assess γ-aminobutyric acid (GABA) type A receptor-mediated spinal inhibitory function in neuropathic pain models of DM rats [127]. Impaired RDD was found in DM rat models of T1DM and T2DM with neuropathic pain phenotypes [62•]. Also, the RDD in groups of healthy controls and T1DM subjects with painful- and painless-DPN was evaluated and it was significantly impaired in those with painful-DPN. Patients with greater RDD attenuation had higher pain scores but no difference in measures of large or small fiber dysfunction, perhaps suggesting spinal inhibitory dysfunction may occur independent of PNS alterations in painful-DPN.

Advanced MRI Studies of the Brain

Functional MRI (fMRI) measures the activity of brain regions by detecting changes in the oxygenation of hemoglobin, the blood oxygen level dependent signal (BOLD). The neurological signature of physical pain has been identified by fMRI and includes activation of the venterolateral thalamus, dorsal posterior insula, and somatosensory cortex, as well as brain regions related to emotional pain processing, including the anterior insula and anterior cingulate cortex (ACC) [128130].

The Thalamus

The thalamus receives somatosensory signals from the spinal cord where they are processed, modulated, and transmitted to higher brain centers. A variety of brain alterations have been demonstrated in DPN and painful-DPN using advanced imaging techniques that examine brain neurochemistry, microvascular blood flow, and functional changes. MR spectroscopy (MRS) enables the measurement of selected metabolites within the brain [131]. Our group has used MRS to show neuronal dysfunction within the thalamus, by reduced N-acetyl aspartate (NAA) to choline ratio as a neuronal marker, in subjects with painless-DPN [132]. Furthermore, DM animal models of painful-DPN have shown that the thalamus may be responsible for central amplification of somatosensory signals [133, 134]. Similarly, thalamic dysfunction appears to play a key role in human painful-DPN. We have recently shown preserved thalamic NAA and the GABA levels within the thalamus in patients with painful-DPN, whereas these levels were reduced in patients with painless-DPN [135, 136]. These findings suggest that neurochemical measures of the thalamic neuronal function and neurotransmitters may be essential for pain signal transmission and/or amplification in painful-DPN.
Recently, we performed a study administering exogenous perfusion contrast to subjects with painful- and painless-DSPN to compare the thalamic microvascular perfusion at rest [137•]. Subjects with DPN both demonstrated delayed bolus arrival time to the thalamus, but subjects with neuropathic pain had a significantly taller peak concentration, a higher mean cerebral blood volume, and the longest blood transit time compared to painless-DPN. Therefore, microvascular vasodilation within the thalamus may induce hyperperfusion which could be related to elevated thalamic neuronal activity. Finally, fMRI study of the brain has indicated there may be disruption in thalamocortical connectivity in painful-DPN. Cauda et al. measured resting state fMRI to determine temporal correlations of brain activity in a small number of subjects with painful-DPN and healthy control patients [138]. Compared with the control group, there was reduced synchrony between the somatosensory cortex and thalamic nuclei in painful-DPN patients.

Descending Inhibition

It is well recognized that the midbrain and medullary brain regions can exert bidirectional control over nociception [139]. The periaqueductal gray (PAG) and rostroventromedial medulla (RVM) are key sites for the control of descending pain modulation, disruption of which in rodent models of painful-DPN has been shown to lead to enhancement of descending pain facilitation [140, 141]. In human studies, our group performed MR-dynamic susceptibility contrast imaging at rest and under experimental pain, by applying heat pain to the lateral thigh where participants did not experience neuropathy [142]. During experimental pain, the time to peak concentration of contrast reduced in healthy volunteers but significantly increased in subjects with painful-DPN in the bilateral sensory cortices and thalami, perhaps indicating an underlying impairment in descending inhibition. Segerdahl et al. interrogated the ventrolateral PAG (vlPAG) using resting state fMRI and arterial spin labelling to determine cerebral blood flow at rest and during heat stimulation to the foot [143•]. The painful-DPN group demonstrated altered vlPAG functional connectivity, which correlated to their pain intensity and the cerebral blood flow changes induced by experimental thermal stimulation. These studies indicate that abnormalities within the descending pain modulatory system may result not only in reduced inhibition of pain but increased amplification of pain signals in painful-DPN.

Higher Brain Centers

The higher brain centers are involved in the localization of pain (e.g., somatosensory cortex) as well as the behavioral, cognitive, and emotional response to painful stimuli (e.g., ACC, amygdala, insular cortex). Using a technique known as voxel-based morphometry, we calculated the brain volumes in subjects with DPN and identified total brain volume reduction which was localized to the somatosensory regions [144, 145]. Furthermore, our group has performed the largest cohort study of brain volume changes in DPN and painful-DPN to date [146]. In painful-DPN, cortical atrophy is localized within the somatomotor cortex and insula. We have also demonstrated abnormal cortical interactions within the somatomotor network at rest which correlated with measures of pain and behavior in subjects with painful-DPN [147]. A recent study performed single-photon emission computed tomography to assess cerebral blood flow (CBF) in 24 subjects with painful- and 20 painless-DPN [148]. The painful-DPN group demonstrated increased CBF within the right ACC and left nucleus accuumbens. However, the painless-DPN group demonstrated more severe neurophysiological neuropathic impairment which may be a potential confounding factor. Furthermore, application of thermal heat stimuli resulted in altered BOLD fMRI responses in painful- compared with painless-DPN, seen in two studies [149, 150]. A pilot study within our group found greater BOLD response within the primary somatosensory cortex, lateral frontal, and cerebellar regions [149]. Whereas Tseng et al. demonstrated augmented responses in multiple limbic and striatal structures (i.e., ACC, superior frontal gyrus, medial thalamus, anterior insular cortex, lentiform nucleus, and premotor area) with the BOLD signal in the ACC and lentiform nucleus correlating with pain rating to thermal stimulation [150]. It is currently unknown whether the CNS changes described in these studies are a response to peripheral nervous system afferent inputs or a primary mechanism responsible for the maintenance of neuropathic pain.

Conclusions

Painful-DPN is a major cause of morbidity in patients with DM. Unfortunately, our understanding of why patients with DPN develop neuropathic pain remains inadequate (Fig. 1.). We have summarized the current evidence of the differences between painful- and painless-DPN (see Table 1.). However, there are limitations in many of the studies including small sample sizes, inappropriate definition of neuropathic pain and DPN, and measurement of multiple variables, leading to a risk of false positives. More recently, large, well-characterized cross-sectional cohort studies have given valid insights into the risk factors and somatosensory profiles of painful-DPN [23••, 27••, 28••]. Unfortunately, longitudinal studies which prospectively identify definitive differences in painful-DPN have not yet been performed, and would be logistically challenging and costly to perform. These limitations notwithstanding, painful-DPN seems to be associated with female gender, increased small fiber injury and/or function, and peripheral/central vascular alterations. The role of autonomic dysfunction, vitamin D, inflammatory mediators, genetic factors, and methylglyoxal needs further clarification. Studies of the CNS demonstrate clear differences in painful- compared with painless-DPN. The spinal, somatomotor, limbic, thalamic, and ascending and descending modulatory systems demonstrate alterations using numerous testing techniques. However, what remains unclear is the causal relationship between painful-DPN and CNS changes. Further studies are necessary to determine whether these findings are the primary cause of neuropathic pain or adaptive to neuropathic afferent impulses. Irrespective of this, advanced MR imaging modalities have the potential for acting as biomarkers for monitoring therapeutic responses to treatments [151••].
Table 1
Recently reported differences between painful- and painless-diabetic peripheral neuropathy
Contributing factor
Difference associated with painful-DPN
References
Risk factors
Female gender
[21••, 26, 27••, 28••]
 
Nephropathy
[26, 27••]
 
Nav 1.7 mutations
[35•]
Small nerve fiber alterations
Hyposensitivity phenotype
[23••, 27••]
 
Epidermal nerve fiber regeneration
[49, 50•, 51]
Microvascular alterations
Elevated immunostaining for blood vessels
[56]
Vitamin D
Reduced 25-hydroxyvitamin D levels
[110•]
Inflammatory biomarkers
C-reactive protein, tumor necrosis factor-α, inducible nitric oxide synthase and interleukin 6.
[86, 117•, 121]
Central nervous system
Spinal cord
Impaired spinal inhibitory function
[62•]
Thalamus
Preserved thalamic NAA and GABA neurochemistry
[135, 136]
 
Thalamic hyperperfusion
[137•]
 
Altered somatosensory cortex and thalamic functional connectivity
[138]
Descending modulatory pain centers
Descending pain facilitation
[142, 143•]
Higher brain centers
Somatomotor cortex and insula cortical atrophy
[146]
 
Abnormal cerebral blood flow at rest and in response to heat pain
[142, 148]
 
Altered functional connectivity in higher brain centers at rest and experimental pain conditions
[147, 149, 150]
DPN diabetic distal symmetrical polyneuropathy, NAA N-acetyl aspartate, GABA γ-aminobutyric acid, BOLD

Compliance with Ethical Standards

Conflict of Interest

Pallai Shillo, Gordon Sloan, Marni Greig, Leanne Hunt, Dinesh Selvarajah, Jackie Elliott, Rajiv Gandhi, and Iain D. Wilkinson declare that they have no conflict of interest.
Solomon Tesfaye reports grants from Impeto Medical; personal fees from Neurometrix, Pfizer, Miro, Worwag Pharma, Mundipharma, Merck, and Mitsubishi Pharma; and personal fees and other from Novo Nordisk.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Publisher’s Note

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Dent – Das Online-Abo der Zahnmedizin

Online-Abonnement

Mit e.Dent erhalten Sie Zugang zu allen zahnmedizinischen Fortbildungen und unseren zahnmedizinischen und ausgesuchten medizinischen Zeitschriften.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Weitere Produktempfehlungen anzeigen
Literatur
2.
Zurück zum Zitat Dyck PJ, Kratz KM, Karnes JL, Litchy WJ, Klein R, Pach JM, 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;43:817–24.PubMed Dyck PJ, Kratz KM, Karnes JL, Litchy WJ, Klein R, Pach JM, 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;43:817–24.PubMed
3.
Zurück zum Zitat • Pop-Busui R, Boulton AJM, Feldman EJ, Bril V, Freeman R, Malik RA, et al. Diabetic neuropathy: a position statement by the American Diabetes Association. Diabetes Care. 2017;40:136–54 Recent position statement of Diabetic Neuropathy by the American Diabetes Association. • Pop-Busui R, Boulton AJM, Feldman EJ, Bril V, Freeman R, Malik RA, et al. Diabetic neuropathy: a position statement by the American Diabetes Association. Diabetes Care. 2017;40:136–54 Recent position statement of Diabetic Neuropathy by the American Diabetes Association.
4.
Zurück zum Zitat • Tesfaye S, Chaturvedi N, Eaton SE, Ward JD, Manes C, Ionescu-Tirgoviste C, et al. Vascular risk factors and diabetic neuropathy. N Engl J Med. 2005;352:341–50 Prospective epidemiological study showing that, apart from glycemic control, incident neuropathy is associated with modifiable cardiovascular risk factors. PubMed • Tesfaye S, Chaturvedi N, Eaton SE, Ward JD, Manes C, Ionescu-Tirgoviste C, et al. Vascular risk factors and diabetic neuropathy. N Engl J Med. 2005;352:341–50 Prospective epidemiological study showing that, apart from glycemic control, incident neuropathy is associated with modifiable cardiovascular risk factors. PubMed
5.
Zurück zum Zitat • Tesfaye S, Boulton AJ, Dyck PJ, Freeman R, Horowitz M, Kempler P, et al. Diabetic neuropathies: update on definitions, diagnostic criteria, estimation of severity, and treatments. Diabetes Care. 2010;33:2285–93 Review by international panel of experts making recommendations in the clinical practice and research for DPN. PubMedPubMedCentral • Tesfaye S, Boulton AJ, Dyck PJ, Freeman R, Horowitz M, Kempler P, et al. Diabetic neuropathies: update on definitions, diagnostic criteria, estimation of severity, and treatments. Diabetes Care. 2010;33:2285–93 Review by international panel of experts making recommendations in the clinical practice and research for DPN. PubMedPubMedCentral
6.
Zurück zum Zitat Sadosky A, Mardekian J, Parsons B, Hopps M, Bienen EJ, Markman J. Healthcare utilization and costs in diabetes relative to the clinical spectrum of painful diabetic peripheral neuropathy. J Diabetes Complicat. 2015;29:212–7.PubMed Sadosky A, Mardekian J, Parsons B, Hopps M, Bienen EJ, Markman J. Healthcare utilization and costs in diabetes relative to the clinical spectrum of painful diabetic peripheral neuropathy. J Diabetes Complicat. 2015;29:212–7.PubMed
8.
Zurück zum Zitat Daousi C, MacFarlane IA, Woodward A, Nurmikko TJ, Bundred PE, Benbow SJ. Chronic painful peripheral neuropathy in an urban community: a controlled comparison of people with and without diabetes. Diabet Med. 2004;21:976–82.PubMed Daousi C, MacFarlane IA, Woodward A, Nurmikko TJ, Bundred PE, Benbow SJ. Chronic painful peripheral neuropathy in an urban community: a controlled comparison of people with and without diabetes. Diabet Med. 2004;21:976–82.PubMed
9.
Zurück zum Zitat Van Acker K, Bouhassira D, De Bacquer D, Weiss S, Matthys K, Raemen H, 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. 2009;35:206–13.PubMed Van Acker K, Bouhassira D, De Bacquer D, Weiss S, Matthys K, Raemen H, 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. 2009;35:206–13.PubMed
10.
Zurück zum Zitat Ziegler D, Rathmann W, Dickhaus T, Meisinger C, Mielck A, KORA Study Group. Neuropathic pain in diabetes, prediabetes and normal glucose tolerance: the MONICA/KORA Augsburg surveys S2 and S3. Pain Med. 2009;10:393–400.PubMed Ziegler D, Rathmann W, Dickhaus T, Meisinger C, Mielck A, KORA Study Group. Neuropathic pain in diabetes, prediabetes and normal glucose tolerance: the MONICA/KORA Augsburg surveys S2 and S3. Pain Med. 2009;10:393–400.PubMed
11.
Zurück zum Zitat Abbott CA, Malik RA, van Ross ER, Kulkarni J, Boulton AJ. Prevalence and characteristics of painful diabetic neuropathy in a large community-based diabetic population in the U.K. Diabetes Care. 2011;34:2220–4.PubMedPubMedCentral Abbott CA, Malik RA, van Ross ER, Kulkarni J, Boulton AJ. Prevalence and characteristics of painful diabetic neuropathy in a large community-based diabetic population in the U.K. Diabetes Care. 2011;34:2220–4.PubMedPubMedCentral
12.
Zurück zum Zitat • Alleman CJ, Westerhout KY, Hensen M, Chambers C, Stoker M, Long S, et al. Humanistic and economic burden of painful diabetic peripheral neuropathy in Europe: a review of the literature. Diabetes Res Clin Pract. 2015;109:215–25 Review article of the epidemiology, humanistic burden and increasing healthcare costs associated with painful-DPN. PubMed • Alleman CJ, Westerhout KY, Hensen M, Chambers C, Stoker M, Long S, et al. Humanistic and economic burden of painful diabetic peripheral neuropathy in Europe: a review of the literature. Diabetes Res Clin Pract. 2015;109:215–25 Review article of the epidemiology, humanistic burden and increasing healthcare costs associated with painful-DPN. PubMed
13.
Zurück zum Zitat Binns-Hall O, Selvarajah D, Sanger D, Walker J, Scott A, Tesfaye S. One-stop microvascular screening service: an effective model for the early detection of diabetic peripheral neuropathy and the high-risk foot. Diabet Med. 2018;35:887–94.PubMedPubMedCentral Binns-Hall O, Selvarajah D, Sanger D, Walker J, Scott A, Tesfaye S. One-stop microvascular screening service: an effective model for the early detection of diabetic peripheral neuropathy and the high-risk foot. Diabet Med. 2018;35:887–94.PubMedPubMedCentral
14.
Zurück zum Zitat Taylor-Stokes G, Pike J, Sadosky A, Chandran A, Toelle T. Association of patient-rated severity with other outcomes in patients with painful diabetic peripheral neuropathy. Diabetes Metab Syndr Obes. 2011;4:401–8.PubMedPubMedCentral Taylor-Stokes G, Pike J, Sadosky A, Chandran A, Toelle T. Association of patient-rated severity with other outcomes in patients with painful diabetic peripheral neuropathy. Diabetes Metab Syndr Obes. 2011;4:401–8.PubMedPubMedCentral
15.
Zurück zum Zitat Lauria G, Lombardi R. Small fiber neuropathy: is skin biopsy the holy grail? Curr Diab Rep. 2012;12:384–92.16.PubMed Lauria G, Lombardi R. Small fiber neuropathy: is skin biopsy the holy grail? Curr Diab Rep. 2012;12:384–92.16.PubMed
16.
Zurück zum Zitat Jensen TS, Backonja MM, Hernández Jiménez S, Tesfaye S, Valensi P, Ziegler D. New perspectives on the management of diabetic peripheral neuropathic pain. Diab Vasc Dis Res. 2006;3:108–19.PubMed Jensen TS, Backonja MM, Hernández Jiménez S, Tesfaye S, Valensi P, Ziegler D. New perspectives on the management of diabetic peripheral neuropathic pain. Diab Vasc Dis Res. 2006;3:108–19.PubMed
17.
Zurück zum Zitat Sloan G, Shillo P, Selvarajah D, Wu J, Wilkinson ID, Tracey I, et al. A new look at painful diabetic neuropathy. Diabetes Res Clin Pract. 2018;144:177–91.PubMed Sloan G, Shillo P, Selvarajah D, Wu J, Wilkinson ID, Tracey I, et al. A new look at painful diabetic neuropathy. Diabetes Res Clin Pract. 2018;144:177–91.PubMed
18.
Zurück zum Zitat • Andersen ST, Witte DR, Dalsgaard EM, Andersen H, Nawroth P, Fleming 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. 2018;41:1068–75 Recent prospective study finding that cardiovascular risk factors are associated with incident DPN in T2DM. PubMed • Andersen ST, Witte DR, Dalsgaard EM, Andersen H, Nawroth P, Fleming 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. 2018;41:1068–75 Recent prospective study finding that cardiovascular risk factors are associated with incident DPN in T2DM. PubMed
19.
Zurück zum Zitat Pop-Busui R, Lu J, Brooks MM, Albert S, Althouse AD, Escobedo J, 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. 2013;36:3208–15.PubMedPubMedCentral Pop-Busui R, Lu J, Brooks MM, Albert S, Althouse AD, Escobedo J, 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. 2013;36:3208–15.PubMedPubMedCentral
20.
Zurück zum Zitat Callaghan BC, Gao LL, Li Y, Zhou X, Reynolds E, Banerjee M, et al. Diabetes and obesity are the main metabolic drives of peripheral neuropathy. Ann Clin Transl Neurol. 2018;5:397–40521.PubMedPubMedCentral Callaghan BC, Gao LL, Li Y, Zhou X, Reynolds E, Banerjee M, et al. Diabetes and obesity are the main metabolic drives of peripheral neuropathy. Ann Clin Transl Neurol. 2018;5:397–40521.PubMedPubMedCentral
21.
Zurück zum Zitat •• Hébert HL, Veluchamy A, Torrance N, Smith BH. Risk factors for neuropathic pain in diabetes mellitus. Pain. 2017;158:560–8 Recent review of the risk factors for painful-DPN. PubMed •• Hébert HL, Veluchamy A, Torrance N, Smith BH. Risk factors for neuropathic pain in diabetes mellitus. Pain. 2017;158:560–8 Recent review of the risk factors for painful-DPN. PubMed
22.
Zurück zum Zitat Elliott J. Female sex: an independent risk factor for the development of painful neuropathy? Diabetes. 2006;55(Supp 1) Abstract, number 794-P Elliott J. Female sex: an independent risk factor for the development of painful neuropathy? Diabetes. 2006;55(Supp 1) Abstract, number 794-P
23.
Zurück zum Zitat •• Themistocleous AC, Ramirez JD, Shillo PR, Lees JG, Selvarajah D, Orengo 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. 2016;157:1132–45 Detailed cross-sectional study showing that the hyposensitivity sensory phenotype and increased severity of neuropathy are associated with painful-DPN. PubMedPubMedCentral •• Themistocleous AC, Ramirez JD, Shillo PR, Lees JG, Selvarajah D, Orengo 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. 2016;157:1132–45 Detailed cross-sectional study showing that the hyposensitivity sensory phenotype and increased severity of neuropathy are associated with painful-DPN. PubMedPubMedCentral
24.
Zurück zum Zitat Algeffari MA. Painful diabetic peripheral neuropathy among Saudi diabetic patients is common but under-recognized: multicenter cross-sectional study at primary health care setting. J Family Community Med. 2018;25:43–7.PubMedPubMedCentral Algeffari MA. Painful diabetic peripheral neuropathy among Saudi diabetic patients is common but under-recognized: multicenter cross-sectional study at primary health care setting. J Family Community Med. 2018;25:43–7.PubMedPubMedCentral
25.
Zurück zum Zitat Ziegler D, Landgraf R, Lobmann R, Reiners K, Rett K, Schnell O, 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. 2018;139:147–54.PubMed Ziegler D, Landgraf R, Lobmann R, Reiners K, Rett K, Schnell O, 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. 2018;139:147–54.PubMed
26.
Zurück zum Zitat Pai YW, Lin CH, Lee IT, Chang MH, et al. Prevalence and biochemical risk factors of diabetic peripheral neuropathy with or without neuropathic pain in Taiwanese adults with type 2 diabetes mellitus. Diabetol Metab Syndr. 2018;12:11–6. Pai YW, Lin CH, Lee IT, Chang MH, et al. Prevalence and biochemical risk factors of diabetic peripheral neuropathy with or without neuropathic pain in Taiwanese adults with type 2 diabetes mellitus. Diabetol Metab Syndr. 2018;12:11–6.
27.
Zurück zum Zitat •• Raputova J, Srotova I, Vlckova E, Sommer C, Üçeyler N, Birklein F, et al. Sensory phenotype and risk factors for painful diabetic neuropathy: a cross-sectional observational study. Pain. 2017;158:2340–53 Another detailed cross-sectional study finding painful-DPN is associated with severity of neuropathy and thermal hyposensitivity. PubMedPubMedCentral •• Raputova J, Srotova I, Vlckova E, Sommer C, Üçeyler N, Birklein F, et al. Sensory phenotype and risk factors for painful diabetic neuropathy: a cross-sectional observational study. Pain. 2017;158:2340–53 Another detailed cross-sectional study finding painful-DPN is associated with severity of neuropathy and thermal hyposensitivity. PubMedPubMedCentral
28.
Zurück zum Zitat •• Truini A, Spallone V, Morganti R, Tamburin S, Zenette G, Schenone A, et al. A cross-sectional study investigating frequency and features of definitely diagnosed diabetic painful polyneuropathy. Pain. 2018;159:2658–66 Large cross-sectional epidemiological study identifying female gender as the only risk factor associated with painful-DPN. PubMed •• Truini A, Spallone V, Morganti R, Tamburin S, Zenette G, Schenone A, et al. A cross-sectional study investigating frequency and features of definitely diagnosed diabetic painful polyneuropathy. Pain. 2018;159:2658–66 Large cross-sectional epidemiological study identifying female gender as the only risk factor associated with painful-DPN. PubMed
29.
Zurück zum Zitat Abraham A, Barnett C, Katzberg HD, Lovblom LE, Perkins BA, Bril V. Sex differences in neuropathic pain intensity in diabetes. J Neurol Sci. 2018;388:103–6.PubMed Abraham A, Barnett C, Katzberg HD, Lovblom LE, Perkins BA, Bril V. Sex differences in neuropathic pain intensity in diabetes. J Neurol Sci. 2018;388:103–6.PubMed
30.
Zurück zum Zitat Sorge RE, Strath LJ. Sex differences in pain responses. Curr Opin Physiol. 2018;6:75–81. Sorge RE, Strath LJ. Sex differences in pain responses. Curr Opin Physiol. 2018;6:75–81.
32.
Zurück zum Zitat Meng W, Deshmukh HA, Donnelly LA, Torrance N, Colhoun HM, Palmer CN, 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. 2015;2:1386–93.PubMedPubMedCentral Meng W, Deshmukh HA, Donnelly LA, Torrance N, Colhoun HM, Palmer CN, 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. 2015;2:1386–93.PubMedPubMedCentral
33.
Zurück zum Zitat Meng W, Deshmukh HA, van Zuydam NR, Liu Y, Donnelly LA, Zhou K, et al. A genome-wide association study suggests an association of Chr8p21.3 (GFRA2) with diabetic neuropathic pain. Eur J Pain. 2015;19:392–9.PubMedPubMedCentral Meng W, Deshmukh HA, van Zuydam NR, Liu Y, Donnelly LA, Zhou K, et al. A genome-wide association study suggests an association of Chr8p21.3 (GFRA2) with diabetic neuropathic pain. Eur J Pain. 2015;19:392–9.PubMedPubMedCentral
34.
Zurück zum Zitat Li QS, Cheng P, Favis R, Wickenden A, Romano G, Wang H. SCN9A variants may be implicated in neuropathic pain associated with diabetic peripheral neuropathy and pain severity. Clin J Pain. 2015;31:976–82.PubMedPubMedCentral Li QS, Cheng P, Favis R, Wickenden A, Romano G, Wang H. SCN9A variants may be implicated in neuropathic pain associated with diabetic peripheral neuropathy and pain severity. Clin J Pain. 2015;31:976–82.PubMedPubMedCentral
35.
Zurück zum Zitat • Blesneac I, Themistocleous AC, Fratter C, Conrad LJ, Ramirez JD, Cox JJ, et al. Rare Nav1.7 variants associated with painful diabetic peripheral neuropathy. Pain. 2017;159:469–80 Study showing that rare sodium channel genetic variants contribute to neuropathic pain in some patients with DPN. PubMedCentral • Blesneac I, Themistocleous AC, Fratter C, Conrad LJ, Ramirez JD, Cox JJ, et al. Rare Nav1.7 variants associated with painful diabetic peripheral neuropathy. Pain. 2017;159:469–80 Study showing that rare sodium channel genetic variants contribute to neuropathic pain in some patients with DPN. PubMedCentral
36.
Zurück zum Zitat Faber CG, Hoeijmakers JG, Ahn HS, Cheng X, Han C, Choi JS, et al. Gain of function Naν1.7 mutations in idiopathic small fiber neuropathy. Ann Neurol. 2012;71:26–39.PubMed Faber CG, Hoeijmakers JG, Ahn HS, Cheng X, Han C, Choi JS, et al. Gain of function Naν1.7 mutations in idiopathic small fiber neuropathy. Ann Neurol. 2012;71:26–39.PubMed
37.
Zurück zum Zitat Galer BS, Gianas A, Jensen MP. Painful diabetic polyneuropathy: epidemiology, pain description, and quality of life. Diabetes Res Clin Pract. 2000;47:123–8.PubMed Galer BS, Gianas A, Jensen MP. Painful diabetic polyneuropathy: epidemiology, pain description, and quality of life. Diabetes Res Clin Pract. 2000;47:123–8.PubMed
38.
Zurück zum Zitat Gore M, Brandenburg NA, Dukes E, Hoffman DL, Tai KS, Stacey B. Pain severity in diabetic peripheral neuropathy is associated with patient functioning, symptom levels of anxiety and depression, and sleep. J Pain Symptom Manag. 2005;30:374–85. Gore M, Brandenburg NA, Dukes E, Hoffman DL, Tai KS, Stacey B. Pain severity in diabetic peripheral neuropathy is associated with patient functioning, symptom levels of anxiety and depression, and sleep. J Pain Symptom Manag. 2005;30:374–85.
39.
Zurück zum Zitat Veves A, Young MJ, Manes C, Boulton AJ. Differences in peripheral and autonomic nerve function measurements in painful and painless neuropathy. A clinical study. Diabetes Care. 1994;17:1200–2.PubMed Veves A, Young MJ, Manes C, Boulton AJ. Differences in peripheral and autonomic nerve function measurements in painful and painless neuropathy. A clinical study. Diabetes Care. 1994;17:1200–2.PubMed
40.
Zurück zum Zitat Spallone V, Morganti R, D’Amato C, Cacciotti L, Fedele T, Maiello MR, et al. Clinical correlates of painful diabetic neuropathy and relationship of neuropathic pain with sensorimotor and autonomic nerve function. Eur J Pain. 2011;15:153–60.PubMed Spallone V, Morganti R, D’Amato C, Cacciotti L, Fedele T, Maiello MR, et al. Clinical correlates of painful diabetic neuropathy and relationship of neuropathic pain with sensorimotor and autonomic nerve function. Eur J Pain. 2011;15:153–60.PubMed
41.
Zurück zum Zitat Sorensen L, Molyneaux L, Yue DK. The level of small nerve fiber dysfunction dose not predict pain in diabetic neuropathy: a study using quantitative sensory testing. Clin J Pain. 2006;22:261–5.PubMed Sorensen L, Molyneaux L, Yue DK. The level of small nerve fiber dysfunction dose not predict pain in diabetic neuropathy: a study using quantitative sensory testing. Clin J Pain. 2006;22:261–5.PubMed
42.
Zurück zum Zitat Gandhi RA, Marques JL, Selvarajah D, Emery CJ, Tesfaye S. Painful diabetic neuropathy is associated with greater autonomic dysfunction than painless diabetic neuropathy. Diabetes Care. 2010;33:1585–90.PubMedPubMedCentral Gandhi RA, Marques JL, Selvarajah D, Emery CJ, Tesfaye S. Painful diabetic neuropathy is associated with greater autonomic dysfunction than painless diabetic neuropathy. Diabetes Care. 2010;33:1585–90.PubMedPubMedCentral
43.
Zurück zum Zitat D’Amato C, Morganti R, Di Gennaro F, Marfia GA, Spallone V. A novel association between nondipping and painful diabetic polyneuropathy. Diabetes Care. 2014;37:2640–2.PubMed D’Amato C, Morganti R, Di Gennaro F, Marfia GA, Spallone V. A novel association between nondipping and painful diabetic polyneuropathy. Diabetes Care. 2014;37:2640–2.PubMed
44.
Zurück zum Zitat Young RJ, Zhou TQ, Rodriguez E, Prescott RJ, Ewing DJ, Clarke BF. Variable relationship between peripheral somatic and autonomic neuropathy in patients with different syndromes of diabetic polyneuropathy. Diabetes. 1986;35:192–7.45.PubMed Young RJ, Zhou TQ, Rodriguez E, Prescott RJ, Ewing DJ, Clarke BF. Variable relationship between peripheral somatic and autonomic neuropathy in patients with different syndromes of diabetic polyneuropathy. Diabetes. 1986;35:192–7.45.PubMed
45.
Zurück zum Zitat Krämer HH, Rolke R, Bickel A, Birklein F. Thermal thresholds predict painfulness of diabetic neuropathies. Diabetes Care. 2004;27:2386–91.PubMed Krämer HH, Rolke R, Bickel A, Birklein F. Thermal thresholds predict painfulness of diabetic neuropathies. Diabetes Care. 2004;27:2386–91.PubMed
46.
Zurück zum Zitat Terkelsen AJ, Karlsson P, Lauria G, Freeman R, Finnerup NB, Jensen TS. The diagnostic challenge of small fibre neuropathy: clinical presentations, evaluations, and causes. Lancet Neurol. 2017;16:934–44.PubMed Terkelsen AJ, Karlsson P, Lauria G, Freeman R, Finnerup NB, Jensen TS. The diagnostic challenge of small fibre neuropathy: clinical presentations, evaluations, and causes. Lancet Neurol. 2017;16:934–44.PubMed
47.
Zurück zum Zitat •• Smith SM, Dworkin RH, Turk DC, Baron R, Polydefkis M, Tracey I, et al. The potential role of sensory testing, skin biopsy, and functional brain imaging as biomarkers in chronic pain clinical trials: IMMPACT Considerations. J Pain. 2017;18:757–77 Review of the role of sensory testing, skin biopsy and brain imaging as diagnostic, prognostic, predictive and pharmacodynamic biomarkers in analgesic treatment trials. PubMedPubMedCentral •• Smith SM, Dworkin RH, Turk DC, Baron R, Polydefkis M, Tracey I, et al. The potential role of sensory testing, skin biopsy, and functional brain imaging as biomarkers in chronic pain clinical trials: IMMPACT Considerations. J Pain. 2017;18:757–77 Review of the role of sensory testing, skin biopsy and brain imaging as diagnostic, prognostic, predictive and pharmacodynamic biomarkers in analgesic treatment trials. PubMedPubMedCentral
48.
Zurück zum Zitat Üçeyler N, Vollert J, Broll B, Riediger N, Langjahr M, Saffer N, et al. Sensory profiles and skin innervation of patients with painful and painless neuropathies. Pain. 2018;159:1867–76.PubMed Üçeyler N, Vollert J, Broll B, Riediger N, Langjahr M, Saffer N, et al. Sensory profiles and skin innervation of patients with painful and painless neuropathies. Pain. 2018;159:1867–76.PubMed
49.
Zurück zum Zitat Cheng HT, Dauch JR, Porzio MT, Yanik BM, Hsieh W, Smith AG, et al. Increased axonal regeneration and swellings in intraepidermal nerve fibers characterize painful phenotypes of diabetic neuropathy. J Pain. 2013;14:941–7.PubMedPubMedCentral Cheng HT, Dauch JR, Porzio MT, Yanik BM, Hsieh W, Smith AG, et al. Increased axonal regeneration and swellings in intraepidermal nerve fibers characterize painful phenotypes of diabetic neuropathy. J Pain. 2013;14:941–7.PubMedPubMedCentral
50.
Zurück zum Zitat • Bönhof GJ, Strom A, Püttgen S, Ringel B, Brüggemann J, Bódis K, et al. Patterns of cutaneous nerve fibre loss and regeneration in type 2 diabetes with painful and painless polyneuropathy. Diabetologia. 2017;60:2495–503 Study demonstrating measures of cutaneous nerve fibre regeneration are enhanced in painful-DPN. PubMed • Bönhof GJ, Strom A, Püttgen S, Ringel B, Brüggemann J, Bódis K, et al. Patterns of cutaneous nerve fibre loss and regeneration in type 2 diabetes with painful and painless polyneuropathy. Diabetologia. 2017;60:2495–503 Study demonstrating measures of cutaneous nerve fibre regeneration are enhanced in painful-DPN. PubMed
51.
Zurück zum Zitat Galosi E, La Cesa S, Di Stefano G, Karlsson P, Fasolino A, Leone C, et al. A pain in the skin. Regenerating nerve sprouts are distinctly associated with ongoing burning pain in patients with diabetes. Eur J Pain. 2018;22:1727–34.PubMed Galosi E, La Cesa S, Di Stefano G, Karlsson P, Fasolino A, Leone C, et al. A pain in the skin. Regenerating nerve sprouts are distinctly associated with ongoing burning pain in patients with diabetes. Eur J Pain. 2018;22:1727–34.PubMed
52.
Zurück zum Zitat 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. 2015;355:131–7.PubMed 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. 2015;355:131–7.PubMed
53.
Zurück zum Zitat Quattrini C, Tavakoli M, Jeziorska M, Kallinikos P, Tesfaye S, Finnigan J, et al. Surrogate markers of small fiber damage in human diabetic neuropathy. Diabetes. 2007;56:2148–54.PubMed Quattrini C, Tavakoli M, Jeziorska M, Kallinikos P, Tesfaye S, Finnigan J, et al. Surrogate markers of small fiber damage in human diabetic neuropathy. Diabetes. 2007;56:2148–54.PubMed
54.
Zurück zum Zitat Cheung A, Podgomy P, Martinez JA, Chan C, Toth C. Epidermal axonal swellings in painful and painless diabetic peripheral neuropathy. Muscle Nerve. 2015;51:505–13.PubMed Cheung A, Podgomy P, Martinez JA, Chan C, Toth C. Epidermal axonal swellings in painful and painless diabetic peripheral neuropathy. Muscle Nerve. 2015;51:505–13.PubMed
55.
Zurück zum Zitat Anand P, Terenghi G, Warner G, Kopelman P, Williams-Chestnut RE, Sinicropi DV. The role of endogenous nerve growth factor in human diabetic neuropathy. Nat Med. 1996;2:703–7.PubMed Anand P, Terenghi G, Warner G, Kopelman P, Williams-Chestnut RE, Sinicropi DV. The role of endogenous nerve growth factor in human diabetic neuropathy. Nat Med. 1996;2:703–7.PubMed
56.
Zurück zum Zitat Shillo P, Selvarajah D, Greig M, Wilkinson I, Yiangou Y, Donatien P, et al. Nerve and vascular biomarkers in skin biopsies differentiate painful from painless advanced diabetic peripheral neuropathy. Diabet Med. 2017;34(Supp 1):31–3. Shillo P, Selvarajah D, Greig M, Wilkinson I, Yiangou Y, Donatien P, et al. Nerve and vascular biomarkers in skin biopsies differentiate painful from painless advanced diabetic peripheral neuropathy. Diabet Med. 2017;34(Supp 1):31–3.
57.
Zurück zum Zitat Anand P, Bley K. Topical capsaicin for pain management: therapeutic potential and mechanism of action of the new high-concentration capsaicin 8% patch. Br J Anaesth. 2011;107:490–502.PubMedPubMedCentral Anand P, Bley K. Topical capsaicin for pain management: therapeutic potential and mechanism of action of the new high-concentration capsaicin 8% patch. Br J Anaesth. 2011;107:490–502.PubMedPubMedCentral
58.
Zurück zum Zitat Obreja O, Rukwied R, Nagler L, Schmidt M, Schmelz M, Namer B. Nerve growth factor locally sensitizes nociceptors in human skin. Pain. 2018;159:416–26.PubMed Obreja O, Rukwied R, Nagler L, Schmidt M, Schmelz M, Namer B. Nerve growth factor locally sensitizes nociceptors in human skin. Pain. 2018;159:416–26.PubMed
60.
Zurück zum Zitat Malik RA, Kallinikos P, Abbott CA, van Schie CH, Morgan P, Efron N, et al. Corneal confocal microscopy: a non-invasive surrogate of nerve fibre damage and repair in diabetic patients. Diabetologia. 2003;46:683–8.PubMed Malik RA, Kallinikos P, Abbott CA, van Schie CH, Morgan P, Efron N, et al. Corneal confocal microscopy: a non-invasive surrogate of nerve fibre damage and repair in diabetic patients. Diabetologia. 2003;46:683–8.PubMed
61.
Zurück zum Zitat Jiang MS, Yuan Y, Gu ZX, Zhuang SL. Corneal confocal microscopy for assessment of diabetic peripheral neuropathy: a meta-analysis. Br J Opthalmol. 2016;100:9–14. Jiang MS, Yuan Y, Gu ZX, Zhuang SL. Corneal confocal microscopy for assessment of diabetic peripheral neuropathy: a meta-analysis. Br J Opthalmol. 2016;100:9–14.
62.
Zurück zum Zitat • Marshall AG, Lee-Kubli C, Azmi S, Zhang M, Ferdousi M, Mixcoatl-Zecuatl T, et al. Spinal disinhibition in experimental and clinical painful diabetic neuropathy. Diabetes. 2017;66:1380–90 Experimental and clinical study showing spinal inhibitory dysfunction may contribute to the pathogenesis of painful-DPN. PubMedPubMedCentral • Marshall AG, Lee-Kubli C, Azmi S, Zhang M, Ferdousi M, Mixcoatl-Zecuatl T, et al. Spinal disinhibition in experimental and clinical painful diabetic neuropathy. Diabetes. 2017;66:1380–90 Experimental and clinical study showing spinal inhibitory dysfunction may contribute to the pathogenesis of painful-DPN. PubMedPubMedCentral
64.
Zurück zum Zitat Gasparotti R, Padua L, Briani C, Lauria G. New technologies for the assessment of neuropathies. Nat Rev Neurol. 2017;13:203–16.PubMed Gasparotti R, Padua L, Briani C, Lauria G. New technologies for the assessment of neuropathies. Nat Rev Neurol. 2017;13:203–16.PubMed
66.
Zurück zum Zitat Casanova-Molla J, Grau-Junyent JM, Morales M, Valls-Solé J. On the relationship between nociceptive evoked potentials and intraepidermal nerve fiber density in painful sensory polyneuropathies. Pain. 2011;152:410–8.PubMed Casanova-Molla J, Grau-Junyent JM, Morales M, Valls-Solé J. On the relationship between nociceptive evoked potentials and intraepidermal nerve fiber density in painful sensory polyneuropathies. Pain. 2011;152:410–8.PubMed
67.
Zurück zum Zitat Chao CC, Tseng MT, Lin YJ, Yang WS, Hsieh SC, Lin YH, et al. Pathophysiology of neuropathic pain in type 2 diabetes: skin denervation and contact heat-evoked potentials. Hum Brain Mapp. 2013;34:2733–46.PubMed Chao CC, Tseng MT, Lin YJ, Yang WS, Hsieh SC, Lin YH, et al. Pathophysiology of neuropathic pain in type 2 diabetes: skin denervation and contact heat-evoked potentials. Hum Brain Mapp. 2013;34:2733–46.PubMed
68.
Zurück zum Zitat Hansson P, Backonja M, Bouhassira D. Usefulness and limitations of quantitative sensory testing: clinical and research application in neuropathic pain states. Pain. 2007;129:256–9.PubMed Hansson P, Backonja M, Bouhassira D. Usefulness and limitations of quantitative sensory testing: clinical and research application in neuropathic pain states. Pain. 2007;129:256–9.PubMed
69.
Zurück zum Zitat Cruccu G, Sommer C, Anand P, Attal N, Baron R, Garcia-Larrea L, et al. EFNS guidelines on neuropathic pain assessment: revised 2009. Eur J Neurol. 2010;17:1010–8.PubMed Cruccu G, Sommer C, Anand P, Attal N, Baron R, Garcia-Larrea L, et al. EFNS guidelines on neuropathic pain assessment: revised 2009. Eur J Neurol. 2010;17:1010–8.PubMed
70.
Zurück zum Zitat Tsigos C, White A, Young RJ. Discrimination between painful and painless diabetic neuropathy based on testing of large somatic nerve and sympathetic nerve function. Diabet Med. 1992;9:359–65.PubMed Tsigos C, White A, Young RJ. Discrimination between painful and painless diabetic neuropathy based on testing of large somatic nerve and sympathetic nerve function. Diabet Med. 1992;9:359–65.PubMed
71.
Zurück zum Zitat Krishnan ST, Quattrini C, Jeziorska M, Malik RA, Rayman G. Abnormal LDIflare but normal quantitative sensory testing and dermal nerve fiber density in patients with painful diabetic neuropathy. Diabetes Care. 2009;32:451–5.PubMedPubMedCentral Krishnan ST, Quattrini C, Jeziorska M, Malik RA, Rayman G. Abnormal LDIflare but normal quantitative sensory testing and dermal nerve fiber density in patients with painful diabetic neuropathy. Diabetes Care. 2009;32:451–5.PubMedPubMedCentral
72.
Zurück zum Zitat Rolke R, Magerl W, Campbell KA, Schalber C, Caspari S, Birklein F, et al. Quantitative sensory testing: a comprehensive protocol for clinical trials. Eur J Pain. 2006;10:77–88.PubMed Rolke R, Magerl W, Campbell KA, Schalber C, Caspari S, Birklein F, et al. Quantitative sensory testing: a comprehensive protocol for clinical trials. Eur J Pain. 2006;10:77–88.PubMed
73.
Zurück zum Zitat •• Rolke R, Baron R, Maier C, Tölle TR, Treede RD, Beyer A, et al. Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): standardized protocol and reference values. Pain. 2006;123:231–43 QST protocol for the characterization the somatosensory phenotype of patients with neuropathic pain. PubMed •• Rolke R, Baron R, Maier C, Tölle TR, Treede RD, Beyer A, et al. Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): standardized protocol and reference values. Pain. 2006;123:231–43 QST protocol for the characterization the somatosensory phenotype of patients with neuropathic pain. PubMed
74.
Zurück zum Zitat Umapathi T, Tan WL, Loke SC, Soon PC, Tavintharan S, Chan YH. Intraepidermal nerve fiber density as a marker of early diabetic neuropathy. Muscle Nerve. 2007;35:591–8.PubMed Umapathi T, Tan WL, Loke SC, Soon PC, Tavintharan S, Chan YH. Intraepidermal nerve fiber density as a marker of early diabetic neuropathy. Muscle Nerve. 2007;35:591–8.PubMed
75.
Zurück zum Zitat Løseth S, Stålberg E, Jorde R, Mellgren SI. Early diabetic neuropathy: thermal thresholds and intraepidermal nerve fibre density in patients with normal nerve conduction studies. J Neurol. 2008;255:1197–202.PubMed Løseth S, Stålberg E, Jorde R, Mellgren SI. Early diabetic neuropathy: thermal thresholds and intraepidermal nerve fibre density in patients with normal nerve conduction studies. J Neurol. 2008;255:1197–202.PubMed
76.
Zurück zum Zitat Ragé M, Van Acker N, Knaapen MW, Timmers M, Streffer J, Hermans MP, et al. Asymptomatic small fiber neuropathy in diabetes mellitus: investigations with intraepidermal nerve fiber density, quantitative sensory testing and laser-evoked potentials. J Neurol. 2011;258:1852–64.PubMed Ragé M, Van Acker N, Knaapen MW, Timmers M, Streffer J, Hermans MP, et al. Asymptomatic small fiber neuropathy in diabetes mellitus: investigations with intraepidermal nerve fiber density, quantitative sensory testing and laser-evoked potentials. J Neurol. 2011;258:1852–64.PubMed
77.
Zurück zum Zitat Cameron NE, Eaton SE, Cotter MA, Tesfaye S. Vascular factors and metabolic interactions in the pathogenesis of diabetic neuropathy. Diabetologia. 2001;44:1973–88.PubMed Cameron NE, Eaton SE, Cotter MA, Tesfaye S. Vascular factors and metabolic interactions in the pathogenesis of diabetic neuropathy. Diabetologia. 2001;44:1973–88.PubMed
78.
Zurück zum Zitat Tesfaye S, Harris ND, Wilson RM, Ward JD. Exercise-induced conduction velocity increment: a marker of impaired peripheral nerve blood flow in diabetic neuropathy. Diabetologia. 1992;35:155–9.PubMed Tesfaye S, Harris ND, Wilson RM, Ward JD. Exercise-induced conduction velocity increment: a marker of impaired peripheral nerve blood flow in diabetic neuropathy. Diabetologia. 1992;35:155–9.PubMed
79.
Zurück zum Zitat Tesfaye S, Harris N, Jakubowski JJ, Mody C, Wilson RM, Rennie IG, et al. Impaired blood flow and arterio-venous shunting in human diabetic neuropathy: a novel technique of nerve photography and fluorescein angiography. Diabetologia. 1993;36:1266–74.PubMed Tesfaye S, Harris N, Jakubowski JJ, Mody C, Wilson RM, Rennie IG, et al. Impaired blood flow and arterio-venous shunting in human diabetic neuropathy: a novel technique of nerve photography and fluorescein angiography. Diabetologia. 1993;36:1266–74.PubMed
80.
Zurück zum Zitat Tesfaye S, Malik R, Harris N, Jakubowski JJ, Mody C, Rennie IG, et al. Arterio-venous shunting and proliferating new vessels in acute painful neuropathy of rapid glycaemic control (insulin neuritis). Diabetologia. 1996;39:329–35.PubMed Tesfaye S, Malik R, Harris N, Jakubowski JJ, Mody C, Rennie IG, et al. Arterio-venous shunting and proliferating new vessels in acute painful neuropathy of rapid glycaemic control (insulin neuritis). Diabetologia. 1996;39:329–35.PubMed
81.
Zurück zum Zitat Archer AG, Roberts VC, Watkins PJ. Blood flow patterns in painful diabetic neuropathy. Diabetologia. 1984;27:563–7.PubMed Archer AG, Roberts VC, Watkins PJ. Blood flow patterns in painful diabetic neuropathy. Diabetologia. 1984;27:563–7.PubMed
82.
Zurück zum Zitat Eaton SE, Harris ND, Ibrahim S, Patel KA, Selmi F, Radatz M, et al. Increased sural nerve epineurial blood flow in human subjects with painful diabetic neuropathy. Diabetologia. 2003;46:934–9.PubMed Eaton SE, Harris ND, Ibrahim S, Patel KA, Selmi F, Radatz M, et al. Increased sural nerve epineurial blood flow in human subjects with painful diabetic neuropathy. Diabetologia. 2003;46:934–9.PubMed
83.
Zurück zum Zitat Tsigos C, Reed P, Weinkove C, White A, Young RJ. Plasma norepinephrine in sensory diabetic polyneuropathy. Diabetes Care. 1993;16:722–7.PubMed Tsigos C, Reed P, Weinkove C, White A, Young RJ. Plasma norepinephrine in sensory diabetic polyneuropathy. Diabetes Care. 1993;16:722–7.PubMed
84.
Zurück zum Zitat Tack CJ, van Gurp PJ, Holmes C, Goldstein DS. Local sympathetic denervation in painful diabetic neuropathy. Diabetes. 2002;51:3545–53.PubMed Tack CJ, van Gurp PJ, Holmes C, Goldstein DS. Local sympathetic denervation in painful diabetic neuropathy. Diabetes. 2002;51:3545–53.PubMed
85.
Zurück zum Zitat Quattrini C, Harris ND, Malik RA, Tesfaye S. Impaired skin microvascular reactivity in painful diabetic neuropathy. Diabetes Care. 2007;30:655–9.PubMed Quattrini C, Harris ND, Malik RA, Tesfaye S. Impaired skin microvascular reactivity in painful diabetic neuropathy. Diabetes Care. 2007;30:655–9.PubMed
86.
Zurück zum Zitat Doupis J, Lyons TE, Wu S, Gnardellis C, Dinh T, Veves A. Microvascular reactivity and inflammatory cytokines in painful and painless peripheral diabetic neuropathy. J Clin Endocrinol Metab. 2009;94:2157–63.PubMedPubMedCentral Doupis J, Lyons TE, Wu S, Gnardellis C, Dinh T, Veves A. Microvascular reactivity and inflammatory cytokines in painful and painless peripheral diabetic neuropathy. J Clin Endocrinol Metab. 2009;94:2157–63.PubMedPubMedCentral
87.
Zurück zum Zitat Vas PR, Sharma S, Rayman G. Distal sensorimotor neuropathy: improvements in diagnosis. Rev Diabet Stud. 2015;12:29–47.PubMedPubMedCentral Vas PR, Sharma S, Rayman G. Distal sensorimotor neuropathy: improvements in diagnosis. Rev Diabet Stud. 2015;12:29–47.PubMedPubMedCentral
88.
Zurück zum Zitat • Herder C, Kannenberg JM, Huth C, Carstensen-Kirbgerg M, Rathmann W, Koening W, et al. Proinflammatory cytokines predict the incidence and progression of distal sensorimotor polyneuropathy: KORA F4/FF4 Study. Diabetes Care. 2017;40:569–76 Prospective study finding IL-6 and TNF -α are associated with incident DPN. PubMed • Herder C, Kannenberg JM, Huth C, Carstensen-Kirbgerg M, Rathmann W, Koening W, et al. Proinflammatory cytokines predict the incidence and progression of distal sensorimotor polyneuropathy: KORA F4/FF4 Study. Diabetes Care. 2017;40:569–76 Prospective study finding IL-6 and TNF -α are associated with incident DPN. PubMed
89.
Zurück zum Zitat Deguchi T, Hashiguchi T, Horinouchi S, Uto T, Oku H, Kimura K, et al. Serum VEGF increases in diabetic polyneuropathy, particularly in the neurologically active symptomatic stage. Diabet Med. 2009;26:247–52.PubMed Deguchi T, Hashiguchi T, Horinouchi S, Uto T, Oku H, Kimura K, et al. Serum VEGF increases in diabetic polyneuropathy, particularly in the neurologically active symptomatic stage. Diabet Med. 2009;26:247–52.PubMed
90.
Zurück zum Zitat Quattrini C, Jeziorska M, Boulton AJ, Malik RA. Reduced vascular endothelial growth factor expression and intra-epidermal nerve fiber loss in human diabetic neuropathy. Diabetes Care. 2008;31:140–5.PubMed Quattrini C, Jeziorska M, Boulton AJ, Malik RA. Reduced vascular endothelial growth factor expression and intra-epidermal nerve fiber loss in human diabetic neuropathy. Diabetes Care. 2008;31:140–5.PubMed
91.
Zurück zum Zitat Yuen KC, Baker NR, Rayman G. Treatment of chronic painful diabetic neuropathy with isosorbide dinitrate spray: a double-blind placebo-controlled cross-over study. Diabetes Care. 2002;25:1699–703.PubMed Yuen KC, Baker NR, Rayman G. Treatment of chronic painful diabetic neuropathy with isosorbide dinitrate spray: a double-blind placebo-controlled cross-over study. Diabetes Care. 2002;25:1699–703.PubMed
92.
Zurück zum Zitat Rayman G, Baker NR, Krishnan ST. Glyceryl trinitrate patches as an alternative to isosorbide dinitrate spray in the treatment of chronic painful diabetic neuropathy. Diabetes Care. 2003;26:2697–8.PubMed Rayman G, Baker NR, Krishnan ST. Glyceryl trinitrate patches as an alternative to isosorbide dinitrate spray in the treatment of chronic painful diabetic neuropathy. Diabetes Care. 2003;26:2697–8.PubMed
93.
Zurück zum Zitat Obrosova IG. Diabetic painful and insensate neuropathy: pathogenesis and potential treatments. Neurotherapeutics. 2009;6(4):638–47.PubMedPubMedCentral Obrosova IG. Diabetic painful and insensate neuropathy: pathogenesis and potential treatments. Neurotherapeutics. 2009;6(4):638–47.PubMedPubMedCentral
94.
Zurück zum Zitat Dewanjee S, Das S, Das AK, Bhattacharjee N, Dihingia A, Dua TK. Molecular mechanism of diabetic neuropathy and its pharmacotherapeutic targets. Eur J Pharmacol. 2018;833:472–523.PubMed Dewanjee S, Das S, Das AK, Bhattacharjee N, Dihingia A, Dua TK. Molecular mechanism of diabetic neuropathy and its pharmacotherapeutic targets. Eur J Pharmacol. 2018;833:472–523.PubMed
95.
Zurück zum Zitat Shamsaldeen YA, Mackenzie LS, Lione LA, Benham CD. Methylglyoxal, a metabolite increased in diabetes is associated with insulin resistance, vascular dysfunction and neuropathies. Curr Drug Metab. 2016;17:359–67.PubMed Shamsaldeen YA, Mackenzie LS, Lione LA, Benham CD. Methylglyoxal, a metabolite increased in diabetes is associated with insulin resistance, vascular dysfunction and neuropathies. Curr Drug Metab. 2016;17:359–67.PubMed
96.
Zurück zum Zitat Huang Q, Chen Y, Gong N, Wang YX. Methylglyoxal mediates streptozotocin-induced diabetic neuropathic pain via activation of the peripheral TRPA1 and Nav1.8 channels. Metabolism. 2016;65:463–74.PubMed Huang Q, Chen Y, Gong N, Wang YX. Methylglyoxal mediates streptozotocin-induced diabetic neuropathic pain via activation of the peripheral TRPA1 and Nav1.8 channels. Metabolism. 2016;65:463–74.PubMed
97.
Zurück zum Zitat Bierhaus A, Fleming T, Stoyanov S, Leffler A, Babes A, Neacsu C, et al. Methylglyoxal modification of Nav1.8 facilitates nociceptive neuron firing and causes hyperalgesia in diabetic neuropathy. Nat Med. 2012;18:926–33.PubMed Bierhaus A, Fleming T, Stoyanov S, Leffler A, Babes A, Neacsu C, et al. Methylglyoxal modification of Nav1.8 facilitates nociceptive neuron firing and causes hyperalgesia in diabetic neuropathy. Nat Med. 2012;18:926–33.PubMed
98.
Zurück zum Zitat Hansen CS, Jensen TM, Jensen JS, Nawroth P, Fleming T, Witte DR, et al. The role of serum methylglyoxal on diabetic peripheral and cardiovascular autonomic neuropathy: the ADDITION Denmark study. Diabetes Metab. 2015;32:778–85. Hansen CS, Jensen TM, Jensen JS, Nawroth P, Fleming T, Witte DR, et al. The role of serum methylglyoxal on diabetic peripheral and cardiovascular autonomic neuropathy: the ADDITION Denmark study. Diabetes Metab. 2015;32:778–85.
99.
Zurück zum Zitat Zhou J, Zhou S. Inflammation: therapeutic targets for diabetic neuropathy. Mol Neurobiol. 2014;49:536–46.PubMed Zhou J, Zhou S. Inflammation: therapeutic targets for diabetic neuropathy. Mol Neurobiol. 2014;49:536–46.PubMed
100.
Zurück zum Zitat Çakici N, Fakkel TM, van Neck JW, Verhagen AP, Coert JH. Systematic review of treatments for diabetic peripheral neuropathy. Diabet Med. 2016;33:1466–76.PubMed Çakici N, Fakkel TM, van Neck JW, Verhagen AP, Coert JH. Systematic review of treatments for diabetic peripheral neuropathy. Diabet Med. 2016;33:1466–76.PubMed
101.
Zurück zum Zitat Holick MF, Vitamin D. Deficiency. N Engl J Med. 2007;357:266–81.PubMed Holick MF, Vitamin D. Deficiency. N Engl J Med. 2007;357:266–81.PubMed
102.
Zurück zum Zitat Powanda MC. Is there a role for vitamin D in the treatment of chronic pain? Inflammopharmacology. 2014;22:327–32.PubMed Powanda MC. Is there a role for vitamin D in the treatment of chronic pain? Inflammopharmacology. 2014;22:327–32.PubMed
103.
Zurück zum Zitat Danescu LG, Levy S, Levy J. Vitamin D and diabetes mellitus. Endocrine. 2009;35:11–7.PubMed Danescu LG, Levy S, Levy J. Vitamin D and diabetes mellitus. Endocrine. 2009;35:11–7.PubMed
104.
Zurück zum Zitat Mathieu C. Vitamin D and diabetes: where do we stand? Diabetes Res Clin Pract. 2015;108:201–9.PubMed Mathieu C. Vitamin D and diabetes: where do we stand? Diabetes Res Clin Pract. 2015;108:201–9.PubMed
105.
Zurück zum Zitat Fukuoka M, Sakurai K, Ohta T, Kiyoki M, Katayama I. Tacalcitol, an active vitamin D3, induces nerve growth factor production in human epidermal keratinocytes. Skin Pharmacol Appl Ski Physiol. 2001;14:226–33. Fukuoka M, Sakurai K, Ohta T, Kiyoki M, Katayama I. Tacalcitol, an active vitamin D3, induces nerve growth factor production in human epidermal keratinocytes. Skin Pharmacol Appl Ski Physiol. 2001;14:226–33.
106.
Zurück zum Zitat Riaz S, Malcangio M, Miller M, Tomlinson DR. A vitamin D(3) derivative (CB1093) induces nerve growth factor and prevents neurotrophic deficits in streptozotocin-diabetic rats. Diabetologia. 1999;42:1308–13.PubMed Riaz S, Malcangio M, Miller M, Tomlinson DR. A vitamin D(3) derivative (CB1093) induces nerve growth factor and prevents neurotrophic deficits in streptozotocin-diabetic rats. Diabetologia. 1999;42:1308–13.PubMed
107.
Zurück zum Zitat Tague SE, Smith PG. Vitamin D receptor and enzyme expression in dorsal root ganglia of adult female rats: modulation by ovarian hormones. J Chem Neuroanat. 2011;41:1–12.PubMed Tague SE, Smith PG. Vitamin D receptor and enzyme expression in dorsal root ganglia of adult female rats: modulation by ovarian hormones. J Chem Neuroanat. 2011;41:1–12.PubMed
108.
Zurück zum Zitat Tague SE, Clarke GL, Winter MK, McCarson KE, Wright DE, Smith PG. Vitamin D deficiency promotes skeletal muscle hypersensitivity and sensory hyperinnervation. J Neurosci. 2011;31:13728–38.PubMedPubMedCentral Tague SE, Clarke GL, Winter MK, McCarson KE, Wright DE, Smith PG. Vitamin D deficiency promotes skeletal muscle hypersensitivity and sensory hyperinnervation. J Neurosci. 2011;31:13728–38.PubMedPubMedCentral
109.
Zurück zum Zitat Esteghamati A, Fotouhi A, Faghihi-Kashani S, Hafezi-Nejad N, Heidari B, Sheikhbahaei S, et al. Non-linear contribution of serum vitamin D to symptomatic diabetic neuropathy: a case-control study. Diabetes Res Clin Pract. 2016;111:44–50.PubMed Esteghamati A, Fotouhi A, Faghihi-Kashani S, Hafezi-Nejad N, Heidari B, Sheikhbahaei S, et al. Non-linear contribution of serum vitamin D to symptomatic diabetic neuropathy: a case-control study. Diabetes Res Clin Pract. 2016;111:44–50.PubMed
110.
Zurück zum Zitat • Shillo P, Selvarajah D, Greig M, Gandhi R, Rao G, Wilkinson ID, et al. Reduced vitamin D levels in painful diabetic peripheral neuropathy. Diabet Med. 2018;36:44–51. https://doi.org/10.1111/dme.13798. Cross-sectional study demonstrating that reduced serum 25-hydroxyvitamin D levels are associated with painful-DPN. CrossRefPubMed • Shillo P, Selvarajah D, Greig M, Gandhi R, Rao G, Wilkinson ID, et al. Reduced vitamin D levels in painful diabetic peripheral neuropathy. Diabet Med. 2018;36:44–51. https://​doi.​org/​10.​1111/​dme.​13798. Cross-sectional study demonstrating that reduced serum 25-hydroxyvitamin D levels are associated with painful-DPN. CrossRefPubMed
111.
Zurück zum Zitat Lee P, Chen R. Vitamin D as an analgesic for patients with type 2 diabetes and neuropathic pain. Arch Intern Med. 2008;168:771–2.PubMed Lee P, Chen R. Vitamin D as an analgesic for patients with type 2 diabetes and neuropathic pain. Arch Intern Med. 2008;168:771–2.PubMed
112.
Zurück zum Zitat Shehab D, Al-Jarallah K, Abdella N, Mojiminiyi OA, Al Mohamedy H. Prospective evaluation of the effect of short-term oral vitamin d supplementation on peripheral neuropathy in type 2 diabetes mellitus. Med Princ Pract. 2015;24:250–6.PubMedPubMedCentral Shehab D, Al-Jarallah K, Abdella N, Mojiminiyi OA, Al Mohamedy H. Prospective evaluation of the effect of short-term oral vitamin d supplementation on peripheral neuropathy in type 2 diabetes mellitus. Med Princ Pract. 2015;24:250–6.PubMedPubMedCentral
115.
Zurück zum Zitat Jin HY, Park TS. Role of inflammatory biomarkers in diabetic peripheral neuropathy. J Diabetes Investig. 2018;9:1016–8.PubMedPubMedCentral Jin HY, Park TS. Role of inflammatory biomarkers in diabetic peripheral neuropathy. J Diabetes Investig. 2018;9:1016–8.PubMedPubMedCentral
116.
Zurück zum Zitat • Feldman EL, Nave KA, Jensen TS, Bennett DL. New horizons in diabetic neuropathy: mechanisms, bioenergetics, and pain. Neuron. 2017;93:1296–313 Review of the recent discoveries into the pathogenesis of DPN and painful-DPN. PubMedPubMedCentral • Feldman EL, Nave KA, Jensen TS, Bennett DL. New horizons in diabetic neuropathy: mechanisms, bioenergetics, and pain. Neuron. 2017;93:1296–313 Review of the recent discoveries into the pathogenesis of DPN and painful-DPN. PubMedPubMedCentral
117.
Zurück zum Zitat • Herder C, Bongaerts BW, Rathmann W, Heier M, Kowall B, Koenig W, et al. Differential association between biomarkers of subclinical inflammation and painful polyneuropathy: results from the KORA F4 study. Diabetes Care. 2015;38:91–6 Study showing that biomarkers of subclinical and vascular inflammation are associated with painful-DPN. PubMed • Herder C, Bongaerts BW, Rathmann W, Heier M, Kowall B, Koenig W, et al. Differential association between biomarkers of subclinical inflammation and painful polyneuropathy: results from the KORA F4 study. Diabetes Care. 2015;38:91–6 Study showing that biomarkers of subclinical and vascular inflammation are associated with painful-DPN. PubMed
121.
Zurück zum Zitat Purwata TE. High TNF-alpha plasma levels and macrophages iNOS and TNF-alpha expression as risk factors for painful diabetic neuropathy. J Pain Res. 2011;4:169–75.PubMedPubMedCentral Purwata TE. High TNF-alpha plasma levels and macrophages iNOS and TNF-alpha expression as risk factors for painful diabetic neuropathy. J Pain Res. 2011;4:169–75.PubMedPubMedCentral
122.
Zurück zum Zitat Uçeyler N, Rogausch JP, Toyka KV, Sommer C. Differential expression of cytokines in painful and painless neuropathies. Neurology. 2007;69:42–9.PubMed Uçeyler N, Rogausch JP, Toyka KV, Sommer C. Differential expression of cytokines in painful and painless neuropathies. Neurology. 2007;69:42–9.PubMed
123.
Zurück zum Zitat Üçeyler N, Riediger N, Kafke W, Sommer C. Differential gene expression of cytokines and neurotrophic factors in nerve and skin of patients with peripheral neuropathies. J Neurol. 2015;262:203–12.PubMed Üçeyler N, Riediger N, Kafke W, Sommer C. Differential gene expression of cytokines and neurotrophic factors in nerve and skin of patients with peripheral neuropathies. J Neurol. 2015;262:203–12.PubMed
124.
Zurück zum Zitat Empl M, Renaud S, Erne B, Fuhr P, Straube A, Schaeren-Wiemers N, et al. TNF-alpha expression in painful and nonpainful neuropathies. Neurology. 2001;56:1371–7.PubMed Empl M, Renaud S, Erne B, Fuhr P, Straube A, Schaeren-Wiemers N, et al. TNF-alpha expression in painful and nonpainful neuropathies. Neurology. 2001;56:1371–7.PubMed
125.
Zurück zum Zitat Selvarajah D, Wilkinson ID, Emery CJ, Harris ND, Shaw PJ, Witte DR, et al. Early involvement of the spinal cord in diabetic peripheral neuropathy. Diabetes Care. 2006;29:2664–9.PubMed Selvarajah D, Wilkinson ID, Emery CJ, Harris ND, Shaw PJ, Witte DR, et al. Early involvement of the spinal cord in diabetic peripheral neuropathy. Diabetes Care. 2006;29:2664–9.PubMed
127.
Zurück zum Zitat Lee-Kubli CA, Calcutt NA. Altered rate-dependent depression of the spinal H-reflex as an indicator of spinal disinhibition in models of neuropathic pain. Pain. 2014;155:250–60.PubMed Lee-Kubli CA, Calcutt NA. Altered rate-dependent depression of the spinal H-reflex as an indicator of spinal disinhibition in models of neuropathic pain. Pain. 2014;155:250–60.PubMed
128.
Zurück zum Zitat Fomberstein K, Qadri S, Ramani R. Functional MRI and pain. Curr Opin Anaesthesiol. 2013;26:588–93.PubMed Fomberstein K, Qadri S, Ramani R. Functional MRI and pain. Curr Opin Anaesthesiol. 2013;26:588–93.PubMed
129.
130.
Zurück zum Zitat Wager T, Atlas L, Lindquist M, Roy M, Woo C, Kross E. An fMRI-based neurologic signature of physical pain. N Engl J Med. 2013;368:1388–97.PubMedPubMedCentral Wager T, Atlas L, Lindquist M, Roy M, Woo C, Kross E. An fMRI-based neurologic signature of physical pain. N Engl J Med. 2013;368:1388–97.PubMedPubMedCentral
131.
Zurück zum Zitat Buonocore MH, Maddock RJ. Magnetic resonance spectroscopy of the brain: a review of physical principles and technical methods. Rev Neurosci. 2015;26:609–32.PubMed Buonocore MH, Maddock RJ. Magnetic resonance spectroscopy of the brain: a review of physical principles and technical methods. Rev Neurosci. 2015;26:609–32.PubMed
132.
Zurück zum Zitat Selvarajah D, Wilkinson ID, Emery CJ, Shaw PJ, Griffiths PD, Gandhi R, et al. Thalamic neuronal dysfunction and chronic sensorimotor distal symmetrical polyneuropathy in patients with type 1 diabetes mellitus. Diabetologia. 2008;51:2088–92.PubMed Selvarajah D, Wilkinson ID, Emery CJ, Shaw PJ, Griffiths PD, Gandhi R, et al. Thalamic neuronal dysfunction and chronic sensorimotor distal symmetrical polyneuropathy in patients with type 1 diabetes mellitus. Diabetologia. 2008;51:2088–92.PubMed
133.
Zurück zum Zitat Fischer TZ, Tan AM, Waxman SG. Thalamic neuron hyperexcitability and enlarged receptive fields in the STZ model of diabetic pain. Brain Res. 2009;1268:154–61.PubMed Fischer TZ, Tan AM, Waxman SG. Thalamic neuron hyperexcitability and enlarged receptive fields in the STZ model of diabetic pain. Brain Res. 2009;1268:154–61.PubMed
134.
Zurück zum Zitat Freeman OJ, Evans MH, Cooper GJ, Petersen RS, Gardiner NJ. Thalamic amplification of sensory input in experimental diabetes. Eur J Neurosci. 2016;44:1779–86.PubMedPubMedCentral Freeman OJ, Evans MH, Cooper GJ, Petersen RS, Gardiner NJ. Thalamic amplification of sensory input in experimental diabetes. Eur J Neurosci. 2016;44:1779–86.PubMedPubMedCentral
135.
Zurück zum Zitat Gandhi R, Selvarajah D, Wilkinson I, Emery C, Shaw PJ, Griffiths P, et al. Preservation of thalamic neuronal function may be a prerequisite for pain perception in diabetic neuropathy. Diabetologia. 2006;49(Supp 1):2088–92. Gandhi R, Selvarajah D, Wilkinson I, Emery C, Shaw PJ, Griffiths P, et al. Preservation of thalamic neuronal function may be a prerequisite for pain perception in diabetic neuropathy. Diabetologia. 2006;49(Supp 1):2088–92.
136.
Zurück zum Zitat Shillo P, Selvarajah D, Greig M, Rao D, Edden R, Wilkinson I, et al. Painless diabetic peripheral neuropathy is characterised by reduced thalamic gamma-aminobutyric acid (GABA). Diabet Med. 2016;33(Supp 1):162. Shillo P, Selvarajah D, Greig M, Rao D, Edden R, Wilkinson I, et al. Painless diabetic peripheral neuropathy is characterised by reduced thalamic gamma-aminobutyric acid (GABA). Diabet Med. 2016;33(Supp 1):162.
137.
Zurück zum Zitat • Selvarajah D, Wilkinson ID, Gandhi R, Griffiths PD, 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. 2011;34:718–20 Study demonstrating increased microvascular perfusion within the thalamus is associated with painful-DPN. PubMedPubMedCentral • Selvarajah D, Wilkinson ID, Gandhi R, Griffiths PD, 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. 2011;34:718–20 Study demonstrating increased microvascular perfusion within the thalamus is associated with painful-DPN. PubMedPubMedCentral
139.
Zurück zum Zitat Ossipov MH, Morimura K, Porreca F. Descending pain modulation and chronification of pain. Curr Opin Support Palliat Care. 2014;8:143–51.PubMedPubMedCentral Ossipov MH, Morimura K, Porreca F. Descending pain modulation and chronification of pain. Curr Opin Support Palliat Care. 2014;8:143–51.PubMedPubMedCentral
140.
Zurück zum Zitat Silva M, Amorim D, Almeida A, Tavares I, Pinto-Ribeiro F, Morgado C. Pronociceptive changes in the activity of rostroventromedial medulla (RVM) pain modulatory cells in the streptozotocin-diabetic rat. Brain Res Bull. 2013;96:39–44.PubMed Silva M, Amorim D, Almeida A, Tavares I, Pinto-Ribeiro F, Morgado C. Pronociceptive changes in the activity of rostroventromedial medulla (RVM) pain modulatory cells in the streptozotocin-diabetic rat. Brain Res Bull. 2013;96:39–44.PubMed
141.
Zurück zum Zitat Morgado C, Terra PP, Tavares I. Neuronal hyperactivity at the spinal cord and periaqueductal grey during painful diabetic neuropathy: effects of gabapentin. Eur J Pain. 2010;14:693–9.PubMed Morgado C, Terra PP, Tavares I. Neuronal hyperactivity at the spinal cord and periaqueductal grey during painful diabetic neuropathy: effects of gabapentin. Eur J Pain. 2010;14:693–9.PubMed
143.
Zurück zum Zitat • Segerdahl AR, Themistocleous AC, Fido D, Bennett DL, Tracey I. A brain-based pain facilitation mechanism contributes to painful diabetic polyneuropathy. Brain. 2018;141:357–64 Study finding vlPAG altered function may contribute to neuropathic pain in painful-DPN. PubMedPubMedCentral • Segerdahl AR, Themistocleous AC, Fido D, Bennett DL, Tracey I. A brain-based pain facilitation mechanism contributes to painful diabetic polyneuropathy. Brain. 2018;141:357–64 Study finding vlPAG altered function may contribute to neuropathic pain in painful-DPN. PubMedPubMedCentral
144.
Zurück zum Zitat Ashburner J, Friston KJ. Voxel-based morphometry—the methods. Neuroimage. 2000;11:805–21.PubMed Ashburner J, Friston KJ. Voxel-based morphometry—the methods. Neuroimage. 2000;11:805–21.PubMed
145.
Zurück zum Zitat Selvarajah D, Wilkinson ID, Maxwell M, Davies J, Sankar A, Boland E, et al. Magnetic resonance neuroimaging study of brain structural differences in diabetic peripheral neuropathy. Diabetes Care. 2014;37:1681–8.PubMed Selvarajah D, Wilkinson ID, Maxwell M, Davies J, Sankar A, Boland E, et al. Magnetic resonance neuroimaging study of brain structural differences in diabetic peripheral neuropathy. Diabetes Care. 2014;37:1681–8.PubMed
147.
Zurück zum Zitat Selvarajah D, Awadh M, Gandhi R, Wilkinson ID, Tesfaye S. Alterations in somatomotor network functional connectivity in painful diabetic neuropathy—a resting state functional magnetic resonance imaging study. Diabetes. 2018;67(Supp 1):61–OR. https://doi.org/10.2337/db18-61-OR.CrossRef Selvarajah D, Awadh M, Gandhi R, Wilkinson ID, Tesfaye S. Alterations in somatomotor network functional connectivity in painful diabetic neuropathy—a resting state functional magnetic resonance imaging study. Diabetes. 2018;67(Supp 1):61–OR. https://​doi.​org/​10.​2337/​db18-61-OR.CrossRef
148.
Zurück zum Zitat Watanabe K, Hirano S, Kojima K, Nagashima K, Mukai H, Sato T, et al. Altered cerebral blood flow in the anterior cingulate cortex is associated with neuropathic pain. J Neurol Neurosurg Psychiatry. 2018;89:1082–7.PubMed Watanabe K, Hirano S, Kojima K, Nagashima K, Mukai H, Sato T, et al. Altered cerebral blood flow in the anterior cingulate cortex is associated with neuropathic pain. J Neurol Neurosurg Psychiatry. 2018;89:1082–7.PubMed
149.
Zurück zum Zitat Selvarajah D, Gandhi R, Hunter M, Emery C, Griffiths P, Tesfaye S, et al. A functional magnetic resonance imaging study demonstrating alterations in brain responses to acute pain stimulation in diabetic neuropathy. Diabetologia. 2007;50(Supp 1):448–S9. Selvarajah D, Gandhi R, Hunter M, Emery C, Griffiths P, Tesfaye S, et al. A functional magnetic resonance imaging study demonstrating alterations in brain responses to acute pain stimulation in diabetic neuropathy. Diabetologia. 2007;50(Supp 1):448–S9.
150.
Zurück zum Zitat Tseng MT, Chiang MC, Chao CC, Tseng WY, Hsieh ST. fMRI evidence of degeneration-induced neuropathic pain in diabetes: enhanced limbic and striatal activations. Hum Brain Mapp. 2013;34:2733–46.PubMed Tseng MT, Chiang MC, Chao CC, Tseng WY, Hsieh ST. fMRI evidence of degeneration-induced neuropathic pain in diabetes: enhanced limbic and striatal activations. Hum Brain Mapp. 2013;34:2733–46.PubMed
151.
Zurück zum Zitat •• Tesfaye S, Selvarajah D, Gandhi R, Greig M, Shillo P, Fang F, et al. Diabetic peripheral neuropathy may not be as its name suggests: evidence from magnetic resonance imaging. Pain. 2016;157(Supp 1):S72–80 Review of the MRI studies into painful-DPN. PubMed •• Tesfaye S, Selvarajah D, Gandhi R, Greig M, Shillo P, Fang F, et al. Diabetic peripheral neuropathy may not be as its name suggests: evidence from magnetic resonance imaging. Pain. 2016;157(Supp 1):S72–80 Review of the MRI studies into painful-DPN. PubMed
Metadaten
Titel
Painful and Painless Diabetic Neuropathies: What Is the Difference?
verfasst von
Pallai Shillo
Gordon Sloan
Marni Greig
Leanne Hunt
Dinesh Selvarajah
Jackie Elliott
Rajiv Gandhi
Iain D. Wilkinson
Solomon Tesfaye
Publikationsdatum
01.06.2019
Verlag
Springer US
Erschienen in
Current Diabetes Reports / Ausgabe 6/2019
Print ISSN: 1534-4827
Elektronische ISSN: 1539-0829
DOI
https://doi.org/10.1007/s11892-019-1150-5

Weitere Artikel der Ausgabe 6/2019

Current Diabetes Reports 6/2019 Zur Ausgabe

Macrovascular Complications in Diabetes (VR Aroda and A Getaneh, Section Editors)

Effect of Health Information Technologies on Cardiovascular Risk Factors among Patients with Diabetes

Economics and Policy in Diabetes (AA Baig and N Laiteerapong, Section Editors)

Updates in Gestational Diabetes Prevalence, Treatment, and Health Policy

Economics and Policy in Diabetes (AA Baig and N Laiteerapong, Section Editors)

Economic Impact of Diabetes in South Asia: the Magnitude of the Problem

Economics and Policy in Diabetes (AA Baig and N Laiteerapong, Section Editors)

Economics and Policy in Bariatric Surgery

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

CAR-M-Zellen: Warten auf das große Fressen

22.05.2024 Onkologische Immuntherapie Nachrichten

Auch myeloide Immunzellen lassen sich mit chimären Antigenrezeptoren gegen Tumoren ausstatten. Solche CAR-Fresszell-Therapien werden jetzt für solide Tumoren entwickelt. Künftig soll dieser Prozess nicht mehr ex vivo, sondern per mRNA im Körper der Betroffenen erfolgen.

Chronische Verstopfung: „Versuchen Sie es mit grünen Kiwis!“

22.05.2024 Obstipation Nachrichten

Bei chronischer Verstopfung wirken Kiwis offenbar besser als Flohsamenschalen. Das zeigen die Daten aus einer randomisierten Studie, die der Gastroenterologe Oliver Pech beim Praxis-Update vorstellte.

So häufig greift rheumatoide Arthritis auf Organe über

21.05.2024 Rheumatoide Arthritis Nachrichten

Im Verlauf von rheumatoider Arthritis entwickeln viele Patienten extraartikuläre Manifestationen. Schwedische Forscher haben sich mit der Inzidenz und den Risikofaktoren befasst.

Blutdrucksenkung könnte Uterusmyome verhindern

Frauen mit unbehandelter oder neu auftretender Hypertonie haben ein deutlich erhöhtes Risiko für Uterusmyome. Eine Therapie mit Antihypertensiva geht hingegen mit einer verringerten Inzidenz der gutartigen Tumoren einher.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.