Sei sulla pagina 1di 18

G Model

NSL-31080; No. of Pages 18 ARTICLE IN PRESS


Neuroscience Letters xxx (2015) xxxxxx

Contents lists available at ScienceDirect

Neuroscience Letters
journal homepage: www.elsevier.com/locate/neulet

Review

Mechanisms of distal axonal degeneration in peripheral neuropathies


Christopher R. Cashman a,b , Ahmet Hke a,
a
Departments of Neuroscience and Neurology, USA
b
MSTP- MD/PhD Program, Johns Hopkins University, Baltimore, MD 21205, USA

a r t i c l e i n f o a b s t r a c t

Article history: Peripheral neuropathy is a common complication of a variety of diseases and treatments, including dia-
Received 27 October 2014 betes, cancer chemotherapy, and infectious causes (HIV, hepatitis C, and Campylobacter jejuni). Despite
Received in revised form 16 January 2015 the fundamental difference between these insults, peripheral neuropathy develops as a combination
Accepted 19 January 2015
of just six primary mechanisms: altered metabolism, covalent modication, altered organelle function
Available online xxx
and reactive oxygen species formation, altered intracellular and inammatory signaling, slowed axonal
transport, and altered ion channel dynamics and expression. All of these pathways converge to lead to
Keywords:
axon dysfunction and symptoms of neuropathy. The detailed mechanisms of axon degeneration itself
Peripheral neuropathy
GuillainBarr syndrome
have begun to be elucidated with studies of animal models with altered degeneration kinetics, includ-
Diabetes ing the slowed Wallerian degeneration (WldS ) and Sarm knockout animal models. These studies have
HIV neuropathy shown axonal degeneration to occur through a programmed pathway of injury signaling and cytoskeletal
Mitochondrial aging degradation. Insights into the common disease insults that converge on the axonal degeneration pathway
Campylobacter jejuni promise to facilitate the development of therapeutics that may be effective against other mechanisms of
Chemotherapy induced peripheral neurodegeneration.
neuropathy 2015 Elsevier Ireland Ltd. All rights reserved.
WldS
Mechanisms of neuropathy
Post-infectious neuropathy

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.1. Clinical overview of selected diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.1.1. Diabetic neuropathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.1.2. Chemotherapy induced peripheral neurotoxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.1.3. Human immunodeciency virus neuropathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
1.1.4. Non-HIV infectious causes: hepatitis C and Campylobacter jejuni . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2. Mechanisms of peripheral neuropathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.1. Metabolic dysregulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.2. Covalent modication . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.3. Organelle damage and reactive oxygen species . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.3.1. Impaired mitochondria function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.3.2. Endoplasmic reticulum dysfunction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.3.3. Reactive oxygen and nitrogen species . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.4. Intracellular and inammatory signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.5. Axonal transport defects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.6. Channelopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

Abbreviations: AGE, advanced glycation end products; AIDP, acute inammatory demyelinating polyradiculoneuropathy; AMAN, acute motor axonal neuropathy; CIDP,
chemotherapy induced peripheral neurotoxicity; ddC, 2 ,3 -dideoxycytidine; ETC, electron transport chain; G6P, glucose-6-phosphate; G6PD, glucose-6-phosphate dehydro-
genase; GBS, GuillainBarr syndrome; HCV, hepatitis C virus; IGF-1, insulin-like growth factor-1; IFN-/, interferon or ; MPTP, mitochondrial permeability transition
pore; Nmnat, nicotinamide mononucleotide adenylyltransferase; PARP, poly(ADP-ribose) polymerase; RAGE, receptor of advanced glycation end products; RNS, reactive
nitrogen species; ROS, reactive oxygen species; TLR, toll-like receptor; Ube4b, ubiquitination factor E4B; WldS , slowed Wallerian degeneration gene.
Corresponding author at: Department of Neurology, Johns Hopkins University School of Medicine, 855 North Wolfe St., Rangos 248, Baltimore, MD 21205, USA.
Tel.: +1 410 955 2227.
E-mail address: ahoke@jhmi.edu (A. Hke).

http://dx.doi.org/10.1016/j.neulet.2015.01.048
0304-3940/ 2015 Elsevier Ireland Ltd. All rights reserved.

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048
G Model
NSL-31080; No. of Pages 18 ARTICLE IN PRESS
2 C.R. Cashman, A. Hke / Neuroscience Letters xxx (2015) xxxxxx

3. New insights into mechanisms of degeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00


4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

1. Introduction support (hypoinsulinemia in type I and advanced type II diabetes)


and mitochondrial health and function, may also be important.
Peripheral nerve degeneration is a common disorder of the
nervous system whereby sensory and/or motor axons no longer
effectively communicate between the periphery and central ner- 1.1.2. Chemotherapy induced peripheral neurotoxicity
vous system. The prevalence of peripheral neuropathy in the United While improved cancer treatment regimes and higher rates of
States has been reported to be nearly 15% in adults over the age forty remission are a boon to modern medicine and cancer biologists,
[108]. many of the commonly used anti-neoplastics have long term toxic
Peripheral neuropathy is not a single, homogenous disease, effects that currently are not well mitigated during treatment [45].
but is instead a mix of different clinical presentations, natural Indeed, chemotherapy induced peripheral neurotoxicity (CIPN) has
histories, and pathologies. Patients may present with motor insuf- increased in incidence as cancer remission rates continue to climb
ciency (weakness), sensory abnormalities (numbness, paresthesias, due to improved cancer therapies (as reviewed in [12,45]). While
hyperalgesia/allodynia, pain), autonomic symptoms, or a combi- the incidence and prevalence of CIPN varies by agent, in general,
nation of all, often depending on the particular disease. These 3080% of treated patients develop a peripheral neuropathy (as
various constellations of neurological symptoms suggest motor, reviewed in [96]). Patients experience motor and sensory symp-
sensory, and autonomic axons have differing susceptibilities to toms, including numbness, pain, and weakness [248], so severely
various disease processes. Additionally, while most neuropathies that they may be dose-limiting. The wide variety of symptoms
are chronic, slowly progressive conditions, some neuropathies suggests chemotherapeutic agents may harm multiple types of
have a more acute onset and gradual recovery [111] (reviewed neurons, as well as different parts of neurons, including the axon
in [140,275,293]). The heterogeneity of peripheral neuropathies (axonopathy) or the soma/ganglion (ganglionopathy) (as reviewed
is likely secondary to the initiating event. Few neuropathies are in [118]). The most common causes of CIPN include taxanes, such
present in isolation, but, rather, are often secondary to other sys- as paclitaxel, platinum agents including cisplatin and oxaloplatin,
temic illnesses, including diabetes and infectious causes such as and proteasome inhibitors such as bortezomib and will, thus, be
human immunodeciency virus and hepatitis C virus. Additionally, the focus of discussion. For other compounds, readers are referred
peripheral neuropathies may be iatrogenic, arising from the toxicity to Argyriou et als excellent, recent review [12]. Within a pharma-
of drugs given as part of antiretroviral or chemotherapy regiments. cological class, there is also heterogeneity. For example, cisplatin
Despite the great variety of cause and symptoms, the patho- has chiey chronic affects, but oxaliplatin has toxic neurological
physiology of peripheral neuropathies is limited to only a handful effects in both the acute and chronic setting, suggesting different
of mechanisms. In this article, the pathophysiological mechanisms processes may underlie early and late pathology. Thus, the partic-
of various types of peripheral neuropathy diabetic neuropathy, ular pathophysiology of CIPN depends not only on the family of
chemotherapy induced peripheral neurotoxicity (CIPN), HIV- and chemotherapeutic but also on the specic member of the family.
non-HIV infectious neuropathies will briey be reviewed in an
effort to examine how many different disease process converge
onto a handful of cellular targets involved in the axonal degradation 1.1.3. Human immunodeciency virus neuropathy
pathway to produce peripheral nerve dysfunction. As it is among The global prevalence of the human immunodeciency virus
the most heavily studied, diabetic neuropathy will serve as the pro- (HIV) in 2009 was found to be 33.3 million [1], and nervous sys-
totypical example for many of the mechanisms, then additional tem defects, including a peripheral neuropathy, is common in
detail added for particular diseases. the disease [159,229]. Interestingly, while control of the virus has
improved over the past few decades, the incidence of neuropathy
1.1. Clinical overview of selected diseases has increased from 13% in 1993 to 42% in 2006 [229]. This increase
in prevalence during the era of effective treatments suggests that
1.1.1. Diabetic neuropathy the neuropathy is not caused by the virus alone, but also by the
Diabetes mellitus is a very common chronic disease. In 2014, drugs used to treat it [87]. HIV neuropathy is chiey a sensory
over 9% of the United States population (21 million people) has neuropathy of pain, paresthesias, and absent ankle reexes [67].
been diagnosed with diabetes [47], and the prevalence is expected Direct viral infection of neurons or Schwann cells has not been well
to increase [220]. Diabetes is the most common cause of peripheral demonstrated, but the virus has been recovered from some nerve
neuropathy, accounting for over half of the cases in a recent Dutch samples [52,74]. Viral concentration in these samples is remarkably
study [207]. low, suggesting viral particles and proteins must enter the nerve
Diabetic peripheral neuropathy is classically a sensory neuropa- through other cells, such as macrophages or T-cells. Indeed, careful
thy presenting as numbness and paresthesias in a length dependent studies of viral tropism have shown that particles recovered from
stocking glove distribution, whereby the feet are affected earlier nerve are consistent with macrophage and T-cell infectious par-
and more severely than the hands (as reviewed in [51]), but painful, ticles, [127] and that viral proteins colocalize with macrophages
autonomic, or motor neuropathies may also occur (reviewed in in vivo [115]. These ndings suggest the virus is brought to the
[249]). In two studies of patients with type-I diabetes, intense glu- nerve by immune cells, but does not directly infect neuronal cells
cose control reduced the risk of peripheral neuropathy by 60% [66,115]. The viral proteins gp120 and protein R (VPR) are espe-
[102,110]. This nding suggests that the elevated glucose levels cially important toward the observed neurotoxicity. The primary
in the body may be pathogenic. While hyperglycemia may be mechanisms of the neuropathy appear to be immune damage from
a large component of the pathogenesis of diabetic neuropathy, viral proteins and mitochondrial toxicity from the antiretrovirals
non- hyperglycemic effects of diabetes, including reduced trophic (as reviewed in [131,133]).

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048
G Model
NSL-31080; No. of Pages 18 ARTICLE IN PRESS
C.R. Cashman, A. Hke / Neuroscience Letters xxx (2015) xxxxxx 3

1.1.4. Non-HIV infectious causes: hepatitis C and Campylobacter glucose is instead diverted into the polyol pathway by the enzyme
jejuni aldose reductase to produce the alcohol sorbitol (see Fig. 1). Sorbitol
In addition to HIV, hepatitis C virus (HCV) and C. jejuni infection and other polyols accumulate in various tissues throughout the
are also associated with peripheral neuropathy. Hepatitis C is a viral body, including the sciatic nerve of experimental rat models [101].
infection that may become an intractable, chronic infection, best Sorbitol reduces the level of membrane component myo-inositol
known for causing liver cirrhosis and hepatocellular carcinoma. in cultured neuroblastoma cells exposed to high levels of glucose
In 2011, the best estimate of prevalence was 2.35% of the world by inhibiting its cellular import [290]. This disruption of the axonal
population (160 million people), making it ve times more com- membrane could reduce the ability of the axon to propagate an
mon than HIV [143]. HCV has long been known to be associated action potential [290] or impair the ability to regenerate following
with neurological complications including peripheral neuropa- injury since a wave of lipogenesis is necessary for regeneration
thy and encephalopathy [241]. A study in France of 321 patients [156], and nerves from patients with diabetes demonstrate areas
with hepatitis C infection found 9% and 10% develop a sensory of focal demyelination and remyelination, a lipid intensive process
or motor neuropathy, respectively [40]. HCV-associated periph- [245]. Additionally, myo-inositol depletion in a rat model of type II
eral neuropathy has been further divided into four subtypes based diabetes is associated with reduced Na+ -K+ -ATPase activity, leading
on distribution: polyneuropathy, mononeuritis multiplex, cranial to a nerve conduction decits [107].
neuropathy, and a combination of polyneuropathy/cranial neu- NADPH is one of the primary intracellular antioxidants, so its
ropathy, with axonal degeneration and demyelination on biopsy depletion reduces the ability of a cell to protect against oxidant
[180]. Importantly, the hepatitis C virus has not been shown to damage, ultimately leading to apoptosis. The polyol pathway also
replicate or infect muscle or nerves directly [14], so, as was the case depletes NADPH, since it is used in the conversion of glucose into
with HIV, neuropathy must develop through indirect, inammatory sorbitol (see Fig. 1). The pentose phosphate pathway is an anaero-
mechanisms instead of direct viral infection. bic metabolic pathway used to create NADPH reducing equivalents
In these studies of HCV and peripheral neuropathy, cryoglobu- and other biomolecules. G6PD is the gateway to this pathway, so
linemia was found to alter the complication prole associated with its inhibition by high glucose levels is a second hit against the
HCV infection [40] and be associated with more widespread neu- production of antioxidants, ultimately leading to increased rates
ropathy [180]. Cryoglobulins are immunoglobulins that reversibly of apoptosis [247,300]. Thus, dysregulation of the polyol path-
aggregate at cooler (less than 37 C) temperatures and are associ- way by hyperglycemia may cause nerve damage through multiple
ated with a variety of chronic and autoimmune diseases, with HCV mechanisms: membrane damage by reduced levels of myoinos-
infection being one of the best characterized (as reviewed in [95]). itol, and increased radical damage from a reduced antioxidant
Between 30 [213] and 78% [180] of patients with HCV have been capacity.
reported to have cryoglobulinemia. Although the presence of cryo-
globulinemia as a risk factor for neuropathy has been debated, it is 2.2. Covalent modication
interesting to consider Nemni, et als ndings that, while cryoglob-
ulinemia may not increase the severity of neuropathy, it changes In addition to the potential toxicity from alternative metabolic
the prole to one of a more generalized syndrome [180]. This obser- pathways in diabetes, glucose itself can be toxic as a result of
vation is pertinent since cryoglobulinemia is known to lead to non-enzymatic addition of glucose to proteins via the Amadori
systemic vasculitis, suggesting ischemic injury and inammation product [6] of the Maillard reaction [155] whereby glucose con-
may be important for the development and/or advancement of HCV denses with the amine group of amino acids to form modied
induced peripheral neuropathy [14] . proteins, advanced glycation end products (AGEs), as reviewed
The bacteria C. jejuni is a common cause of gastroenteritis and in [35]. AGEs have been observed in both the axons and Schwann
is associated [100] with a post-infectious neuropathy in 1 of 1000 cells of peripheral nerves, with enrichment of AGEs in samples
infections [5]. First described by Guillian, Barr, and Strohl [111], from patients and animal models of diabetes [261,234,84]. These
the neuropathy often occurs a week after infection and is classi- molecules work through direct modication of biomolecules.
cally an acute, ascending, motor neuropathy (as reviewed in [275]). Among the modied proteins is collagen, which leads to
With an estimated incidence of 1/100,000 [5], GuillianBarr syn- increased thickness [240], immune complex deposition [36], and
drome (GBS) is somewhat rare but may cause permanent disability LDL trapping in vitro [37] in comparison to non-modied colla-
or be fatal secondary to diaphragmatic paralysis and respiratory gen. All of these are characteristics of an atherosclerotic plaque,
failure [275]. While some viruses may also cause GBS, C. jejuni is which can lead to luminal narrowing of a vessel and greater risk
believed to cause 30% of all cases [5]. GBS has recently been appreci- of occlusion, thereby reducing blood ow and exposing the nerve
ated to have multiple subtypes [275], including acute inammatory to hypoxic conditions. This hypoxic risk is increased by AGEs that
demyelinating polyradiculoneuropathy (AIDP) and acute motor quench the vasodilator nitric oxide released from endothelial cells
axonal neuropathy (AMAN) [109,119,162]. The pathophysiology of [38], demonstrating how multiple aspects of the response to hyper-
AMAN is more thoroughly understood than AIDP, and, thus will be glycemic synergize to damage tissue.
used to discuss principles that may be generalized to other sub- One of the principal components of the basement membrane,
types. laminin is critical for the health and development of neurons
and Schwann cells (as reviewed in [62]) and is directly modi-
2. Mechanisms of peripheral neuropathy ed by AGEs. Modied laminin inhibits neurite outgrowth in vitro
when compared to nonmodied laminin [90], and this reduction
2.1. Metabolic dysregulation in outgrowth persists despite neurotrophin stimulation or precon-
In diabetes, hyperglycemia leads to dysregulation of the ditioning [84]. One can imagine the AGE modications of collagen
polyol, hexosamine, and pentose phosphate pathways that ulti- and laminin combining to produce recurrent injury to axons: the
mately leads to reactive intermediates that damage the axon and modied collagen facilitates hypoxic conditions which damages
Schwann cells. Glucose-6-phosphate (G6P) can be diverted by the the axon, but the modied laminin is not permissive to axon
enzyme glucose-6-phosphate dehydrogenase (G6PD) from glycol- regrowth, thereby limiting peripheral nerve recovery and produc-
ysis into the anaerobic pentose phosphate pathway to produce ing an axonopathy.
more NADPH. When intracellular glucose (and, thus, glucose- 6- Glyceraldehyde-3- phosphate, a metabolic intermediate of the
phosphate) levels are very high, G6PD is inhibited [300], and polyol and glycolytic pathway, can nonenzymatically bind to, and,

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048
G Model
NSL-31080; No. of Pages 18 ARTICLE IN PRESS
4 C.R. Cashman, A. Hke / Neuroscience Letters xxx (2015) xxxxxx

Fig. 1. Summary of metabolic pathways. (For interpretation of the references to colour in this gure legend, the reader is referred to the web version of this article.)
A variety of metabolic processes that branch off of glycolysis are dysregulated in diabetes, including the polyol, pentose phosphate, and hexosamine pathways. This dysreg-
ulation leads to protein modication, changes in extra- and intra-cellular signaling (effectors in blue text), and decreased antioxidant capacity secondary to reduced NADPH
levels (shown in green). Red arrow indicates inhibition of the pentose phosphate pathway by elevated glucose levels.

thus interfere with, other proteins [42] (as reviewed in [246]). oral hypoglycemic drugs [198]. This study conrms the ability of
Glyceraldehyde-3-phosphate may also activate the protein kinase AGE compounds to react with other biomolecules in vivo as well as
C (PKC) pathway, since it can be degraded into diacylglycerol, one illustrates how modied molecules remain in the tissue despite ini-
of the secondary messenger of the canonical PKC signaling cascade tiation of hypoglycemic therapy, although it remains unclear what
[203]. This pathway has been shown to be particularly relevant for effect the modied DNA has in these tissues.
vascular complications associated with diabetes (as reviewed in DNA modication is also believed to be one of the mecha-
[73]). nisms responsible for the chronic symptoms of CIPN, particularly
The hexosamine pathway produces glucosamine-6-phosphate following treatment with platinum containing compounds like cis-
and glutamate from fructose-6-phosphate and glutamine (see platin and oxaliplatin. Incubation of DRG neurons in vitro with
Fig. 1). Glucosamine-6- phosphate is ultimately converted into oxaliplatin and cisplatin decreases neurite outgrowth, increases
UDP-N- acetylglucosamine (UDP-GlcNAc) and other hexosamines platinum/DNA adducts [160,238], and apoptosismediated cell
to serve as substrates for glycoprotein sugar moieties (reviewed in death [238]. The exact mechanisms of toxicity is platinum adduct
[39]). UDP-GlcNAc can be covalently bound to proteins by O-GlcNAc formation with nuclear and mitochondrial DNA [160,197] which
transferase. These modications are believed to regulate protein initiates apoptosis [161]. Interestingly, the increased accumulation
function by competing with kinase phosphorylation [63]. Indeed, of platinum/DNA adducts is selective for DRG neurons, over cancer
O-GlcNAcylation has been observed on numerous proteins regulat- cells, in vitro [160], likely due to expression of a selective organic
ing transcription, including RNA polymerase II [63], Sp-1 [287], and cation transporter [231]. This pattern may explain the particular
insulin receptor substrates 1 and 2 (IRS1/2) [89]. In the case of Sp-1, vulnerability of sensory neurons to platinum compounds like cis-
O-GlcNAcylation leads to increased expression of more pathogenic platin and oxaliplatin.
proteins such as TGF [81]. Thus, over activity of the hexosamine
biosynthetic pathway modies the activity of numerous transcrip- 2.3. Organelle damage and reactive oxygen species
tion factors, ultimately leading to gross changes in cell state and
signaling. Mitochondria, the site of oxidative phosphorylation, are intrin-
In addition to proteins, other biomolecules, such as DNA, can sically linked to the development of reactive oxygen species (as
also be modied by AGE products. Plasmid DNA incubated with reviewed in [175]). Damage or pathology associated with these
reactive AGE products produced double strand DNA breaks and organelles may lead to cell death through apoptosis or ener-
base modications [174]. A small study of 31 type II diabetic getic failure, thereby increasing sensitivity to ischemia. Given the
patients in Turkey found higher AGE DNA adduct concentration constant ux of electrons and oxygen in mitochondria, it is not
in patients with diabetes compared to controls, despite the use of surprising that they are likely the primary site (more than 90%)

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048
G Model
NSL-31080; No. of Pages 18 ARTICLE IN PRESS
C.R. Cashman, A. Hke / Neuroscience Letters xxx (2015) xxxxxx 5

of reactive oxygen species production (as reviewed in [19]). Mito- though apoptotic rates were unaffected [270]. Furthermore, calpain
chondrial dysfunction, both in its normal function and increased also degrades the neuronal calcium sensor-1 protein, leading to
ROS production, has been implicated in the pathogenesis of many reduced IP3 signaling [27,28]. In addition to direct toxic and apop-
different neuropathies. totic effects, the abnormal mitochondria may also be pathogenic
In addition to mitochondria, (dys) function of the endoplas- because of reduced respiratory ability, thereby contributing to
mic reticulum also facilitates development of neuropathy. As a energetic failure [280,301].
fundamental component of the protein synthesis and modica- Mitochondrial dysfunction has also been observed in axons
tion pathway, insults that may inhibit the proteasome or increase following CIPN caused by oxaliplatin and bortezomib [279,301].
the concentration of unfolded proteins may trigger stress, thereby Commonly used for multiple myeloma, bortezomib is a chemother-
releasing calcium and initiating the unfolded protein response, apeutic that inhibits the proteasome. Like platinum agents and
leading to cell death through multiple pathways (as reviewed taxanes, it is well known to cause painful sensory neuropathy in
in [282]). Impairment in both the endoplasmic reticulum and humans and animals [166]. Bortezomib, like paclitaxel, has been
mitochondria are a common mechanism to all of the pathologies shown to lead to mitochondrial calcium release leading to activa-
discussed. tion of the apoptotic cascade [142] and energetic collapse [302].
In HIV neuropathy, both the virus and the drugs used to treat
2.3.1. Impaired mitochondria function the infection lead to mitochondrial dysfunction. When cultured
While it is perhaps expected that mitochondria, as the pri- DRG neurons were exposed to HIV infected macrophages, mito-
mary energy production site for the mammalian cell, would display chondrial membrane potential in the soma decreased as reactive
certain pathology in the setting of a hyperglycemia and aberrant oxygen species formation increased, but there was no effect on
metabolism, recent work has suggested that the mitochondria play neurite mitochondria, suggesting there are distinct mechanisms
a much more active role in the development of neuropathy through of injury between the soma and axon [115]. This separation has
cell death and injury. been well characterized in studies on inammatory and apoptotic
In a murine model of type II diabetes, Vincent et al., found signaling (discussed below).
abnormal mitochondrial morphology in sensory neurons despite In addition to induction of apoptosis and increased calcium
an increased mitochondrial density in their axons, neurites, and cell from the MPTP (as reviewed in [206]), mitochondria are func-
bodies compared to healthy control mice [86,255]. Vincent et al., tionally impaired because of damaged DNA. In a recent study of
suggest increased mitochondrial ssion is necessary to meet the sural nerve mitochondria in HIV patients with and without sen-
greater energetic requirements of a hyperglycemic state, but too sory neuropathy, mitochondrial deletions were more common in
rapid mitogenesis leads to unhealthy mitochondria [255]. Alterna- those with neuropathy [144], with more mutations observed in
tively, mitochondria ssion may represent a response to toxicity. In distal mitochondria versus those in more proximal aspects [144].
a 2005 review, Youle and Karbowski proposed a model of the induc- These mutations were correlated with reduced mitochondrial res-
tion of apoptosis whereby the machinery necessary for ssion also piratory function [144]. Interestingly, mitochondrial defects were
mediates apoptosis [145,291]. not observed in the proximal sciatic nerve, only the sural nerve,
Mitochondrial dysfunction is also evident with an impaired res- which suggests the distal aspect of specic types of axons are more
piratory capacity. Indeed, in an animal model of type I diabetes, sensitive to toxicity [144]. Lehman et als study of mitochondrial
mitochondria in sensory neurons displayed depressed respiratory mutations and dysfunction lead to the development of a novel,
function compared to control animals due to lower activity of com- unifying theory of neuropathy based on mitochondrial aging and,
plex I and IV secondary to reduced protein expression of certain thus, distal axon susceptibility. Mitochondria must travel from the
complex components [56]. The reduced protein levels, in turn, soma to the periphery, which may take months to years. During this
are likely due to reduced PGC-1/AMPK axis signaling that occurs process, mitogenesis takes place, but in an imperfect manner, lead-
in the setting of hyperglycemia and metabolic derangement [55]. ing to an accumulation of mitochondrial DNA (mtDNA) mutations,
Impaired electron transport chain function also causes an increase and, thus decits. This process would make the distal most aspect
in ROS generation (as reviewed in [177]), which will be discussed of an axon more susceptible to further injury than the proximal
in more detail later. aspect, mirroring the length dependency of many neuropathies.
Mitochondrial dysfunction is a major mechanism of CIPN across Additionally, Lehman et al., also suggested that the higher density of
all of the various chemotherapeutic agents. The taxane family mitochondria in small, lightly or non-myelinated bers [144] will
of chemotherapeutics, including paclitaxel, exercises its antineo- make these bers especially sensitive to mitochondrial dysfunc-
plastic effects through inhibition of tubulin depolymerization (as tion, as is observed with the primarily sensory component of many
reviewed in [128]), thereby interfering with the stabile/labile bal- neuropathies. Lastly, one can imagine that processes that affect
ance of microtubules [128], including the axons of neurons [17]. axonal transport will only exacerbate mitochondrial dysfunction
Swollen and vacuolated mitochondria are commonly observed in distally since it takes longer to reach distal points. This hypothesis
the axons of sensory nerves of animal models CIPN caused by of mitochondrial aging is an exciting development toward a uni-
paclitaxel [98,279,280]. By interfering with microtubule dynam- fying explanation of many different peripheral neuropathies, but
ics, paclitaxel has been shown to interfere with calcium signaling more testing is needed, especially toward the development of ther-
in mitochondria and the endoplasmic reticulum by the opening of apeutics that protect mitochondria from chronic damage and aging.
the mitochondrial membrane permeability pore, leading to calcium This hypothesis suggests that agents that directly damage mito-
triggered calcium release from the ER [135,171]. The opening of chondrial DNA may exacerbate neuropathic symptoms, and this is
the mitochondrial permeability transition pore (MPTP) also leads indeed what is observed in the treatment of HIV infection.
to release of ROS and cytochrome C [252]. For these reasons, the Mitochondrial dysfunction in HIV neuropathy occurs through a
MPTP has been hypothesized to be a major, convergent effector variety of antiretroviral treatments (as reviewed in [71]). Treatment
of axonal degeneration [21] via induction of apoptosis [205] and of rabbits with a nucleoside analog (2 ,3 -dideoxycytidine;ddC)
downstream calcium activated effectors. The increase in calcium show robust dysmyelination, abnormal Schwann cell morphology,
may also facilitate cellular injury by activating the calcium depen- and slowing of nerve conduction velocity [9,91]. Treated patients
dent protease calpain. Indeed, paclitaxel treated animals that were display an increased density of swollen, vacuolated mitochon-
also administered a calpain inhibitor displayed improved neuro- dria with inclusions in the axons and Schwann cells of the sural
logical scores and fewer signs of sensory axon degeneration, even nerves in those treated with ddC [72]. These abnormal mitochon-

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048
G Model
NSL-31080; No. of Pages 18 ARTICLE IN PRESS
6 C.R. Cashman, A. Hke / Neuroscience Letters xxx (2015) xxxxxx

dria correlate with a reduced level of mtDNA [72], likely as a brane by bypassing ATP synthase, thereby uncoupling the hydrogen
result of inhibition of mtDNA polymerase [158] (as reviewed in ion gradient from the generation of ATP (as reviewed in [208,228]).
[130,131]) and subsequently reduced protein levels [144]. This ROS generation may be reduced by uncoupling since the electron
reduced amount of mtDNA may exacerbate the mtDNA damage transport chain (ETC) complexes that may generate superoxide
from aging as described in the hypothesis above. Indeed, the ratio of radical are bypassed (as reviewed in [173,228]). The uncoupling
mtDNA to nuclear DNA in nerve samples is reduced with infection protein (UCP) family includes three proteins, UCP-1, UCP-2, and
and drug over infection only, and marginally recovers upon drug UCP-3, which are all hydrogen ion channels or transporters. UCP-
cessation [68]. Like HIV itself, the antiretroviral-induced damage 1 was identied to be involved in thermogenesis from brown fat
to mtDNA also leads to impaired mitochondria function. [181], while UCP-2 and UCP-3 were found [85] to be important
Antiretrovirals are sufcient to interfere with mitochondrial for homeostasis [99] and thermogenesis [31] in a variety of tissues
function. By inhibiting replication of the mitochondrial genome, including white adipose tissue and brain for UCP-2 [99] and DRG,
a number of the necessary components of the electron transport brown adipose tissue, and skeletal muscle for UCP-3 [257,284].
chain may not be produced, thereby leading to uncoupled trans- UCP-2 and UCP-3 were also quickly identied as inhibitors of ROS
port and decreased oxidative phosphorylation, as is observed in generation in the mitochondria [179,253], since null mice displayed
neuronal mitochondria treated with ddC in vitro [132]. Interest- evidence of increased ROS production and damage (as reviewed in
ingly, patients on nucleoside analogs often display hyperlactemia, [30]). UCPs are also critical for the attenuation of damage from ROS
which resolves upon drug cessation. One could imagine sys- secondary to hyperglycemia. A variety of studies have shown all
temic mitochondrial inhibition would lead to decreased oxidative three UCP family members to protect cells from hyperglycemia
phosphorylation, and, thus a concomitant increase in glycolysis induced ROS generation when the proteins are overexpressed
and lactic acid fermentation. Thus, antiretroviral drugs and HIV [209,257]. Interestingly, in the setting of hyperglycemia, UCP-3
infection synergize to cause greater injury [305]. As with other expression is actually reduced [257]. These data suggest develop-
neuropathies, mitochondrial dysfunction may cause additional cel- ment of damage secondary to hyperglycemia may be facilitated by a
lular injury through the production of reactive radical species, as reduction of UCPs in vivo, or, alternatively, loss of UCPs in vivo may
outlined below. facilitate the development of diabetic complications. This relation-
ship makes UCP expression an enticing therapeutic target, but the
2.3.2. Endoplasmic reticulum dysfunction development of drugs to alter expression must move forward with
The endoplasmic reticulum (ER) is also vulnerable to various great caution since overexpression of UCP-2, for example, reduces
toxicities, as evidenced by the induction of the unfolded protein insulin secretion, thereby potentially worsening the hyperglycemia
response. While ER inhibition in diabetes is not well characterized, characteristic of diabetes [49,50]. Thus, while there is some conict-
bortezomib has been shown to induce a transient stress response ing evidence on the utility of increased UCP expression toward the
in the Schwann cell ER, leading to myelination deciency and an protection from, or development of, the complications of diabetes,
increased cytokine expression [224]. This off target effect is perhaps there is clear evidence that the coupling of the ETC to ATP produc-
unsurprising, given bortezomibs mechanism of action as a protea- tion becomes dysregulated in diabetes which may lead to increased
some inhibitor one would expect the unfolded protein response to ROS and altered energetics (as reviewed in [173]).
increase, as well. Superoxide production may be a unifying mechanism that initi-
While most of the work has examined mitochondrial dysfunc- ates, or least propagates, many of the mechanisms of neuropathy in
tion in the setting of HIV neuropathy and anti-retroviral therapy, diabetes. The aforementioned pathogenic elements from metabolic
the ER may also be adversely affected by viral proteins. Indeed, in derangements (sorbitol, AGEs, PKC signaling) decreased upon inhi-
a study by Hke, et al., the HIV envelope protein gp120 induced bition of superoxide [183]. Mitochondrial function and DNA health
ER calcium release by binding to the chemokine receptor to acti- was also improved upon inhibition of superoxide production in
vate the canonical IP3 signaling cascade leading to rapid increase the retina in a model of diabetes [242] . The ROS control of these
of intracellular calcium [123]. When calcium storage in the ER was mechanisms is effected by DNA damage activated poly(ADP-ribose)
experimentally reduced, the neurons were more resistant to gp120 polymerase (PARP). PARP catalyzes the inhibitory ADP-ribosylation
triggered cell death, reinforcing the important role of the ER in of proteins, including GAPDH [80] (as reviewed in [177]), inhibition
mediating degeneration [123]. A similar calcium response has been of which increases activity of the hexosamine pathway, PKC sig-
described upon DRG neuron exposure in vitro to Vpr [2]. While the naling activation, and AGE concentration [80]. Additionally, recent
downstream effect of the increased calcium has not been well stud- evidence has implicated PARP as not just a downstream signal of
ied, one could imagine a similar mechanism to what is observed in oxidative damage, but, instead, as a promulgator of ROS damage
CIPN with calpain protease activation and the induction of apopto- in a variety of tissues, including Schwann cells and the peripheral
sis (as reviewed in [116]). nerve [185]. PARP is directly neurotoxic due to the depletion of its
cofactor, NAD+ , when it is catalytically active [113]. Severe reduc-
2.3.3. Reactive oxygen and nitrogen species tion of NAD+ results in energetic collapse of the cell and necrotic
The electron transport chain (ETC) produces superoxide as a death [113].
byproduct of electron uncoupling followed by reduction of oxygen In addition to ADP- ribosylation, superoxide may directly oxi-
to form the free radical [20], but mitochondria also produce hydro- dize proteins to form carbonyl adducts that may modify protein
gen peroxide, hydroxide radicals, and nitric oxide, all of which are function (as reviewed in [177]). Indeed, these modied proteins
reactive species that can directly damage other proteins or DNA have been identied in the brain [70,202], although their function
through covalent modication (as reviewed in [177,227]). While is not well studied. In addition to oxygen modication, ROS may
superoxide production is a common feature of all eukaryotic cells, lead to PARP mediated up regulation of iNOS followed by reaction
the nervous system may be particularly vulnerable to free radical of nitric oxide with superoxide to produce the reactive nitrogen
damage because of large energetic demands and high lipid content radical [185]. Regardless of its source, nitric oxide may also modify
(as reviewed in [168]). proteins [185] to act in both damaging and protective capacities
Natural antioxidants help mitigate this damage, such that the [168]. Nitric oxide and its radical may be a particularly important
depletion of these molecules increases the susceptibility of the pathogenic mechanisms in the nervous system due to its use as
cell the oxidative damage. Uncoupling proteins serve to reduce both a neurotransmitter by neurons and regulator of blood ow
the electrochemical potential across the mitochondrial inner mem- secondary to nitric oxide mediated vasodilation [168]. The balance

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048
G Model
NSL-31080; No. of Pages 18 ARTICLE IN PRESS
C.R. Cashman, A. Hke / Neuroscience Letters xxx (2015) xxxxxx 7

between nitric oxide and its radical likely explains the observed increase in oxidative stress and NF-B induction upon exposure to
protective and damaging effects of nitric oxide, since nitric oxide AGEs [285]. Indeed, the antioxidant -lipoic acid protects Schwann
with its vasodilatory activity is likely protective, but its radical, cells from AGE induced apoptosis in vitro [219]. Thus, RAGE con-
that accumulates with increased ROS, likely mediates protein, and, verts the transient inammatory signal of the AGE into a sustained
thus, cellular, injury (as reviewed in [258]). Thus, the develop- inammatory response that may lead to a long term inammatory
ment of ROS and reactive nitrogen species (RNS) are a critical state, thereby hindering treatment [25,79].
component of diabetes-induced damage that extends beyond sim- After the identication of RAGE, three other receptors were
ple radical chemistry to prolonged alterations of cellular signaling identied (AGE-R1, AGE- R2, and AGE-R3) as part of a large AGE-
pathways. receptor complex [288,264], (as reviewed in [259]). As a complex,
CIPN, like diabetes, is also mediated by ROS (as reviewed in [65]). AGE- R1/R2/R3 has been shown to be up regulated in the presence
Both oxaliplatin and cisplatin have been shown to generate ROS of AGEs with AGE-R3 demonstrating the greatest induction after
[126], and mitochondria are damaged by ROS/RNS secondary to exposure, and AGE-R2 phosphorylation increased in vitro upon
drug treatment [125]. Regardless of the source, these ROS dam- exposure of human umbilical vein endothelial cells to AGE-BSA
age additional cellular components such as proteins and lipids [232]. Together, these data suggest that AGE-R1 may serve as a
[65,77], ultimately leading to energetic failure [125] and apopto- stable receptor, while AGE-R2 effects the intracellular response
sis [126,218]. The result of these derangements include increased through AGE-R3 following AGE-R2 tyrosine phosphorylation. The
nociception as well as distal degeneration of intraepidermal nerve role of intracellular signaling by the AGE-R complex and the rel-
bers, as is seen in animal models of CIPN caused by paclitaxel [23]. evance of the previously identied activities of each component
These data suggest that studies of CIPN must consider the distal in the larger complex remain to be elucidated. Interestingly, early
most aspect of axons, and not just proximal segments seen in the data on the AGE-R complex suggests AGE binding leads to a IL-
sciatic nerve or dorsal roots since damage is not uniform. 1 dependent late induction of progrowth cytokine insulin-like
ROS and RNS are also a prominent component of HIV, and its growth factor (IGF-1) in monocytes in vitro [137], linking RAGE
treatment-induced injury. The generation of these species appears cytokine release to AGE-R response.
to be most harmful to the neuronal cell body [115]. Antioxidant It should be noted that much of the work on the AGE-Rs has
treatment improves cell survival but not neurite outgrowth [115], been performed in endothelial cells, reinforcing the importance of
emphasizing the importance of ROS to somal, but not axonal, injury. vascular injury, with concomitant ischemia, in the development
Lehman and colleagues conrmed these observations in animal of neuropathy, as is seen with RAGE. Current models suggest a
models of HIV infection, whereby the virus increased hydrogen delicate balance between the pro-inammatory and tissue remod-
peroxide production as well as oxidative and nitrosative protein eling effects of RAGE and AGE clearance by the AGE-R complex
modication in the sural, but not the sciatic, nerve [144]. The selec- [150], whereby AGE binding facilitates tissue remodeling and
tivity of sural nerve versus sciatic nerve may be due to the increased inammatory cell recruitment, leading to vascular occlusion and
sensitivity of distal sensory bers to mitochondrial dysfunction, as neuronal/axonal injury. This model remains to be tested in vivo,
discussed previously [144]. however.
While the function of PARP in the regulation of metabolism
2.4. Intracellular and inammatory signaling through GAPDH modication and as an effector of ROS injury
was previously discussed, it is also responsible for the regula-
In diabetes, in addition to non-enzymatic modication of tion of inammatory signaling. PARP was elegantly demonstrated
biomolecules, AGEs are also bioactive as ligands for two major to, upon exposure to inammatory particles like LPS and TNF-,
types of cell surface receptors. The rst discovered receptor, recep- increase transcription factors that lead to an increased expression
tor for AGE (RAGE), was identied through the selective binding of inammatory cytokines and iNOS [112]. The expression of iNOS
of glycosylated albumin to the surface of mouse macrophages is particularly interesting, as its up regulation varied by cell type
with subsequently AGE/RAGE internalization [260,262], leading and stimulus [112]. Nitric oxide can also initiate damage through
to macrophage proliferation in a granulocyte/macrophage colony its radical as well as signal through in an independent cascade.
stimulating factor (GMCSF) dependent manner [292]. Since its Nitric oxide serves as both a vasodilator and neurotransmit-
identication, RAGE has been found to be expressed by a variety ter that has a complex activity in neuronal signaling, damage, and
of cell types, including neurons (as reviewed in [230]). Vlassara repair with mixed data describing its protection from or contribu-
et al., hypothesized that RAGE acts to clear harmful AGE modied tion toward cellular damage and neuropathy (as reviewed in [306]).
proteins from the body [260]. Interestingly, the RAGE receptor also As described above, iNOS is induced in glia by PARP [113] where it
has a soluble isoform that is believed to be a scavenger receptor for serves an inammatory function. However, in cultured endothe-
AGEs and found in many tissues, including the adult nervous system lial cells, endothelial nitric oxide synthase (eNOS) has been shown
[53]. A reduced level of this scavenger isoform is correlated with to be down regulated in the setting of hyperglycemia, as a result
increased risk of peripheral neuropathy in patients with type II dia- of O-linked N-acetylglucosamine modications secondary to acti-
betes [13]. RAGE, however, is not simply a net to clear AGEs, but also vation of the hexosamine pathway, thereby increasing the risk of
can initiate an inammatory response through cytokine release atherosclerosis and/or microvascular ischemia [82]. Interestingly,
[289], including NF-B mediated transcriptional activation of pro- an early study conicted with the previous data and found NOS
inammatory cytokines TNF-, IL-1, and MCP-1 [289], a pattern of and TNF- to be induced in endothelial cells upon exposure to gly-
cytokine expression that may facilitate tissue remodeling, as occurs cated albumin [7]. The authors of this study postulated that the
in atherosclerosis [263]. RAGE has been identied on endothe- NOS and AGE balance is critical for early and late development of
lial cells [178,217], and neurons [32], in vivo, which suggests an diabetic complications, where nitric oxide is increased early in the
immune response with the concomitant increase in ROS could be disease through the activity of NOS, thereby damaging glomeruli
initiated by AGEs in these tissues, thereby effecting additional vas- through hyperltration, whereas AGE products serve to quench NO
cular or neuronal injury [273]. In addition to tissue remodeling, the in the vasculature later in disease, thereby reducing vasodilatory
downstream effect of the induction of these cytokines may be the capacity [7]. Thus, in the setting of hyperglycemia, reactive nitro-
production of ROS, as monocytes from patients with diabetes have gen intermediates may be produced within the nervous system as
greater ROS production than control cells [79]. Cultured endothe- a result of PARP activation secondary to ROS damage while, concur-
lial cells and AGE infused mice also demonstrate a RAGE dependent rently, oxygen ow may be reduced through impaired mechanisms

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048
G Model
NSL-31080; No. of Pages 18 ARTICLE IN PRESS
8 C.R. Cashman, A. Hke / Neuroscience Letters xxx (2015) xxxxxx

of vasodilation. Together, the increase in iNOS and decrease in eNOS nerve and ganglia, which may then lead to tissue damage and more
are two hits toward a toxic knockout of neurons and glia. severe decits.
In addition to PARP and NOS signaling, neurotrophic factor sig- Inammatory signaling is a critical mechanism of HIV neu-
naling is also altered in diabetes. While all of the research discussed ropathy. Indeed, both HIV proteins and systemic inammation
thus far has focused on the effect of hyperglycemia on nerve pathol- secondary to the viral infection may damage neurons and axons (as
ogy, recent work has suggested that insulin, or the lack thereof, reviewed in [133,251]). Recent work has shown gp120 induces sep-
may also facilitate the development of the complications associ- arate processes of injury in the neuronal soma versus the axon. In a
ated with type I diabetes. Indeed, Sima et al., suggest different compartmentalized culture of DRG neurons, gp120 reduces axonal
nerve conduction properties between type I and type II diabetes length in an axon/Schwann cell only compartment and triggers
may be a result of the loss of insulin and C peptide that classi- apoptotic cell death when applied to the soma [165]. Interestingly,
cally occurs in type I diabetes [227]. C-peptide is derived from the in the absence of Schwann cells, DRG soma were protected from
same prohormone as insulin and is increasingly recognized as hav- apoptosis, but axons were still sensitive to the application of gp120
ing distinct, albeit complementary, effects to the action of insulin [165], suggesting different injury pathways were active in the two
(as reviewed in [265]). C-peptide will be lost at the same time as areas. Ultimately, Melli et al. found that this response is due to selec-
insulin, since they are both part of the same precursor peptide. The tive binding of gp120 to chemokine receptors on axons which leads
importance of the loss of C-peptide and insulin toward the devel- to caspase activation, and, thus, injury and degeneration [165]. The
opment of the complications of diabetes was supported by more engagement of apoptotic machinery in the soma, however, was
recent experiments showing diabetic animals had a reduced abil- distinct from the mechanisms in the axons as suggested by the
ity to up regulate neurotrophic factors (IGF-1, IGF-1 receptor, p75, Schwann cell dependence of soma apoptosis in response to gp120
and Trk A) in response to nerve crush injury, ultimately leading [165]. Additional work found gp120 binding to chemokine receptor
to reduced regenerative ability and decreased nerve ber density CXCR4 on Schwann cells in the DRG to result in the release of the
[190,196]. This deciency was largely selective for type I, not type ligand RANTES (regulated upon activation, normal T-cell expressed
II, diabetic models, suggesting that the deciency of insulin and and secreted). RANTES then binds to a different chemokine recep-
C-peptide, not hyperglycemia, leads to axonopathy and decreased tor (CCR5) on the neuron, leading to release of TNF- by neurons
nerve ber density [196]. Evidence for the importance of insulin and autocrine apoptotic signaling through its receptor and the c-
and C-peptide to the development of axonal defects in type I dia- Jun cascade [26,134]. The importance of CXCR4 and CCR5 to the
betes comes from rescue experiments where exogenous C-peptide development of HIV neuropathy also extends to the initiation of
improved conduction decits, myelination defects, regenerative inammation [127]. VPR has been shown to evoke very similar,
capacity, and nerve ber number [226]. pro-inammatory responses in DRG neurons [2]. In addition to the
Additional work has claried the protective role of the insulin activation of cell mediated inammation, Apostolski, et al. showed
induced neurotrophic factor insulin-like growth factor 1 (IGF- gp120 may mediate DRG neuron cell death by activation of the
1). Given a decrease an insulin, as is seen in type I diabetes, complement cascade [11]. Thus, HIV may enter the peripheral ner-
it is perhaps unsurprising that IGF expression is decreased in vous system through other cells, but its proteins and the immune
patients with type I diabetes versus normoglycemic controls [190], response may indirectly damage neuron cell bodies and axons,
Long appreciated for its anti-apoptotic activity in cancer and albeit through slightly different mechanisms.
injury (as reviewed in [256]), IGF-1 was recently found to pro- In addition to the inammation from the virus, recent work has
tect DRG neurons from hyperglycemia induced apoptotic signaling identied antiretrovirals to also increase inammatory signaling
[145]. [303] and activation of apoptotic machinery [26]. To this end, ddC
Other signaling axes have been identied to be dysregulated increases expression of TNF- in GFAP+ cells in the dorsal horn
in diabetes. Nerve growth factor (NGF) is well known to support and neurons in the DRG [303]. In addition to TNF-, BDNF has also
sensory ber survival in vitro and in vivo [5860,146,147] and was been shown to facilitate the development of mechanical and ther-
recently demonstrated to be transiently up regulated in the DRGs of mal hyperalgesia following treatment by a nucleoside analog by
an animal model of type II diabetes during the onset of mechanical increasing ring rates of dorsal horn neurons to stimuli [204]. This
allodynia, suggesting dysregulated NGF signaling may facilitate the nding suggests ddC increases BDNF release which ultimately leads
development of one of the hallmarks of diabetic neuropathy pain to enhanced synaptic connectivity between primary nociceptors
[54]. NGF levels may be elevated at the onset of injury [54], facilitat- and dorsal horn neurons. Thus, in addition to the inammatory
ing the development of neuropathic pain, but then fall later in the signaling from the virus itself, the antiretrovirals may also initi-
course of the disease [93], thereby reducing regenerative capacity, ate damage through modulation of the inammatory response via
although this pattern of expression may be dependent on the type TNF- and synaptic connectivity via BDNF.
of diabetes. Aberrant inammatory signaling is the primary pathogenic
Altered signaling to increase inammation is a prominent com- mechanism of HCV neuropathy. As discussed previously, while
ponent of CIPN. In animals models of paclitaxel toxicity, ATF3, cryoglobulinemia is not necessary for the development of periph-
a general, transient marker of neuronal injury [250], is up regu- eral neuropathy, it changes the symptom constellation from a
lated in DRG neurons within a day of drug administration, while localized neuropathy to a more generalized disorder affecting
glia increase expression of injury markers nearly a week after the multiple nerves. Cryoglobulinemia has long been appreciated to
rst dose of the drug [195]. Additionally, markers of macrophage precipitate peripheral neuropathy [48]. Sural nerve histology of a
(CD68) and microglia (CD11b) activation increase one week fol- patient with cryoglobulinemia showed small and medium vessel
lowing treatment [195]. These data show an evolving pathology perivascular inammation with some vessels completely occluded
whereby the DRG neurons are rst harmed, then supporting and myelin degeneration [48]. IgM and IgG were found deposited
glia in the DRG and nerve, which ultimately leads to recruit- on smaller vessels, while brinogen was readily stained on the
ment or activation of inammatory cells (macrophages, microglia) larger vessels [48]. The authors of this report suggest the patients
concomitant with symptom onset [194,195,272], and increased neuropathy was precipitated by the cryoglobulinemia, whereby
expression of many pro-inammatory cytokines (as reviewed in IgGs bound to myelin, leading to direct immune attack, as well
[271]). Macrophage activation leads to matrix metalloproteinases- as to the small and medium vessels, leading to vascular occlusion
3 and -9 and, as a result, increased tissue degradation [182,225]. and ischemia. A different study found that the antibodies bind-
Thus, the chemotherapeutic agents increase inammation in the ing to myelin were largely against myelin associated glycoprotein

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048
G Model
NSL-31080; No. of Pages 18 ARTICLE IN PRESS
C.R. Cashman, A. Hke / Neuroscience Letters xxx (2015) xxxxxx 9

(MAG) [244]. Thus, the IgG and IgM antibodies may cause damage between diabetic and control animals. While the exact mechanisms
through two mechanisms: activation of complement on myelin and of slowed axonal transport in diabetes, and how it may effect other
ischemia by precipitation of immune complexes and lymphocytic cellular processes, remains to be elucidated, one can imagine that
inammation within the vasa nervorum, causing luminal narrow- such large scale changes in the cell may alter its ability to respond
ing or occlusion [29,244]. While these mechanisms may explain the to its environment, perhaps making it more susceptible to damage
neuropathy in patients with HCV and cryoglobulinemia, recall that through other mechanisms or less efcient with cellular repair and
3070% of patients with HCV neuropathy may not have cryoglob- regeneration.
ulins. In these patients a different mechanism must be responsible Microtubule defects with the concomitant transport difcul-
for their symptoms. ties is a critical component of paclitaxel-induced CIPN with much
Interferon- (IFN- ) modulates the development of peripheral work focusing on how microtubule stabilization by taxanes, in
neuropathy in patients infected with HCV. Both IFN- and IFN- particular, may interfere with axonal transport (as reviewed in
are cytokines that are released during viral infection to induce an [106]). Incubation of DRG neurons with high levels of paclitaxel has
antiviral, intracellular state. IFN- can be induced by most cells, been shown to interfere with anterograde transport of HRP [243].
while IFN- is specically released by lymphocytes as part of the Interestingly, while paclitaxel binds to the lumen of microtubules
cell mediated immune response (as reviewed in [92]). In addition and, thus, cannot directly hinder motor movement [106], electron
to endogenous induction of IFN- by HCV, given its general antivi- microscopy shows decreased rates of microtubule crosslinks and
ral activity, exogenous IFN- has become a critical component reduced density of the cross- linking microtubule associated pro-
of combination therapy for certain HCV infections (as extensively teins upon DRG neuron exposure to paclitaxel in vitro [243]. The
reviewed in [222]). Interestingly, autoimmunity increases follow- dearth of these crosslinks could alter transport dynamics. Alterna-
ing IFN- monotreatment in HCV infection [88], despite a decrease tively, increased microtubule density and aggregation [46,106,243]
in cryoglobulins and viral titer. This suggests that IFN- produces may hinder motor movement. However, these microtubule abnor-
a pro-inammatory state that may facilitate damage to the vasa malities have not been observed in patient sural nerve biopsies
nevorum and myelin. Thus, it is a ne balance between inam- [211], perhaps due to lower drug concentrations.
mation to clear the virus and autoimmunity. This autoimmunity An alternative hypothesis of transport interference in paclitaxel
likely initiates the development of neuropathy in HCV; there is mediated CIPN is covalent modication of tubulin interfering with
a combination of ischemia due to occluded small vessels and molecular motor function. As a microtubule polymerizes, the tubu-
direct attack against the myelin. This model is consistent with lin components that are not on the elongating end can be acetylated,
the non-dependence of neuropathy on the presence of cryoglobu- glutamated, and detyrosinated to increase stability (as reviewed in
lins. While the cryoglobulins may increase vascular inammation, [106]). Investigations of dendrite/axon identity have found tubulin
leading to ischemia and more widespread neuropathy, anti-myelin detyrosination to organize distribution and movement of kinesin
proteins may still be present in the absence of cryoglobulins, so along the axon [83,139]. Paclitaxel enhances all three posttrans-
neuropathy may still develop. It is still unclear how ribavirin may lational modications, thereby disrupting axon/soma/dendrite
reduce the incidence of neuropathy, but one possibility is by lead- polarity [117,276]. The role of posttranslational modication is
ing to more complete viral clearance, ribavirin reduces systemic complex, however, as each modication can alter motor binding
inammation, including cryoglobulins and anti-MAG antibodies. and motility [106], and different motor subtypes may bind to differ-
By reducing these compounds, IFN- autoimmunity, and, thus, ent modied tubulins preferentially [41]. Nonetheless, interruption
neuropathy, is less likely to develop. This hypothesis remains to be of transport may lead to poor anterograde transport of vesicles
tested. and organelles, as well as retrograde transport of trophic factors
[106]. Transport dynamics may also be affected at the end of the
2.5. Axonal transport defects axon, where motors must release from and reattach to different
microtubules. Indeed, a recent paper has shown that in a drosophila
Diabetes also affects axonal health by modulation of axonal model of a degenerative motor neuron disease with a tubulin bind-
transport. Nearly 30 years ago, Medori et al. identied impaired ing domain mutation in a subunit of dynein, dynein accumulates
slow axonal transport of cytoskeletal proteins neurolament (light in the terminal bouton because of a failure to bind to the end of
and medium subunits), tubulin, and actin in an animal model of the microtubule [149]. The microtubule end and motor dynamics
type I diabetes versus healthy controls [164]. The authors also are critical for maintaining proper and equitable delivery of vesi-
observed increased axonal cross sectional area in the proximal cles to terminal boutons [277]. This model suggests dysfunction
aspect of the axon, but a reduced area in the distal aspect. They of transport could lead to derangements at the axon terminal and,
hypothesized that this pattern may be a result of reduced accu- thus, interfere with communication or organelle delivery and lead
mulation of cytoskeletal elements distally because of impaired to terminal degeneration, although this hypothesis has not been
transport [164]. Additional cytoskeletal protein defects include tested.
direct actin and neurolament glycation in the setting of hyper- Surprisingly, low dose paclitaxel has been shown to facilitate
glycemia in vitro [34], and in vivo [69,191] (as reviewed in [163]), regeneration, rather than degeneration, in the central nervous sys-
as well as reduced expression of heavy and light neurolament, and tem [120]. The authors of this initial study suggested paclitaxel
-tubulin in the DRGs of type I diabetic animals [172]. It should may facilitate regeneration by reducing scarring [120]. An addi-
be noted, however, that while actin glycation does not interfere tional hypothesis was proposed three years prior to the discovery
with its ability to polymerize [191], glycosylated tubulin has a of the regenerative benet of low dose paclitaxel. Witte et al.
reduced polymerization ability [274]. The expression and modica- suggested that since paclitaxel modies neuron polarity to cre-
tion of these cytoskeletal proteins (actin, neurolament, tubulin) is ate multiple axons from one cell, perhaps low dose paclitaxel may
believed to interfere with cytoskeletal assembly, and, thus, axonal be helpful to convert a nongrowing minor neurite into a growing
transport [163]. RAGE is predicted to interact with actin through axon to facilitate central nervous system regeneration [276]. Thus,
its effector mDia1 [124,129,157], but recent work has elegantly the microtubule stabilizing dynamics must be delicately balanced
shown that slowed axonal transport in diabetic animals is inde- between stable and unstable, polymer and monomer, such that
pendent of RAGE expression [129]. Despite the slowed transport paclitaxel may either cause axonal degeneration through interrup-
of the RAGE/actin effector protein mDia1 in diabetic animals (wild tion of axonal transport or regeneration by stabilization of growing
type and RAGE knockout), RAGE itself was not differentially present neurites.

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048
G Model
NSL-31080; No. of Pages 18 ARTICLE IN PRESS
10 C.R. Cashman, A. Hke / Neuroscience Letters xxx (2015) xxxxxx

Axonal transport defects are also observed in CIPN caused by cis- an increase in protein levels of TRPV1 in DRGs and spinal cord
platin and bortezomib. As with paclitaxel, in a model of bortezomib [201]. Interestingly, the transcript levels of TRPV1 and CGRP
CIPN, tubulin polymerization is increased in vivo and in vitro upon were reduced in these tissues, suggesting the protein levels likely
clinically appropriate drug concentration [167]. The exact mecha- increase because of bortezomibs inhibition of the proteasome
nism of bortezomib tubulin stabilization remains to be elucidated. [201], thereby potentially increasing neuronal response to noci-
In addition to bortezomib and paclitaxel, early studies on cisplatin ceptive stimuli. Additionally, patients treated with bortezomib
neurotoxicity showed impaired anterograde and retrograde fast had increased resting membrane potential, which would lead
transport in vitro with no gross morphologic abnormalities [210]. to increased neuronal response for a given input. This change
The authors of this study suggested cisplatin may be directly toxic in potential preceded axonal degeneration [176]. Degeneration
to kinesin and dynein through crosslinking or motor ATPase inhi- was suggested by the authors to be due to energetic collapse
bition [210], although little work has been pursued in this line of in the axon, from mitochondrial dysfunction leading to impaired
research. Na+ /K + ATPase activity and, thus, intraaxonal accumulation of
sodium ions [176] and increased calcium inux by reversal of the
2.6. Channelopathy sodium/calcium exchanger [233]. This inux of calcium may begin
the path to apoptosis. This hypothesis remains to be tested, how-
While diabetes does have some sodium/potassium channel dys- ever. Nonetheless, channel dysfunction and changes in membrane
function [107], as discussed with respect to metabolic deregulation polarization appear to be a critical mechanism for the development
and conduction velocity, the details of direct channel modulation of hyperexcitability and acute symptoms of CIPN, especially from
in diabetes is still unclear [254]. However, channel defects are some platinum chemotherapeutics and proteasome inhibitors.
of the fundamental dysfunctions in CIPN and GBS. While little work has examined the electrophysiological effect
In CIPN, channel toxicity is especially relevant for plat- of HIV or its proteins on DRGs, there do appear to be some subtle
inum compounds and proteasome inhibitors. Of the platinum changes. Chiey, both gp120 [188] and Vpr [2] have been shown
chemotherapeutics, oxaliplatin has the largest effect on channels. to increased DRG neuron excitability, perhaps through the inux
Indeed, recall that oxaliplatin has two types of neuropathy: an acute of calcium following activation of the chemokine receptors. In
as well as a more chronic form [22]. Thus far, the chronic neuropa- macrophages, gp120 binding to chemokine receptors has been
thy with numbness and functional impairment has been discussed. shown to directly regulate calcium, potassium, and chloride trans-
However, the acute neuropathy is characterized by transient cold port [148].
hypersensitivity and parasthesias, which generally resolve on their While channel dysfunction has not been identied as a
own and do not require dose changes [22]. Acute abnormalities pathogenic mechanism in HCV neuropathy, the node of Ranvier
of nerve, hippocampal neurons, and DRG neurons are observed is the primary insult in GBS. Much work by Yuki and others have
in vitro after just 45 min of oxaliplatin exposure, where sodium shown molecular mimicry between C. jejuni antigens and ganglio-
current is drastically increased to produce larger compound action sides results in immune attack of the node of Ranvier. The rst
potentials in response to depolarization [4]. These abnormalities clues to the pathophysiology were the association of anti-GM1 gan-
were abrogated with a sodium channel blocker, suggesting modi- glioside antibodies with the AMAN form of GBS following C. jejuni
cation of these channels likely contribute to the abnormality [4], infection [121,296]. The presence of these antibodies predicted a
and the increased refractory period of the neuron suggested that reversible conduction block, believed to be due to disruption of
it was the modication of the voltage gated ion channel inacti- the node of Ranvier [138,141]. The deposition of these antibodies
vation, in particular, that was altered by exposure to oxaliplatin leads to complement deposition on the axolemma [114] and, thus,
[4], with no effect on potassium channels [33]. The mechanisms disruption of axons/Schwann cell architecture at the node and para-
of specic sodium channel sensitization remain to be elucidated, nodal regions [43], ultimately leading to disruption of ion channel
however, and a general increase in sodium current does not explain organization at the node of Ranvier [235]. More specically, anti-
the specic cold hypersensitivity observed in oxaliplatin CIPN. body deposition and complement activation increases potassium
Cold hypersensitivity is instead most likely due to oxaliplatin current in the paranodal region and decreases sodium current in
triggered sensitivity in TRP channels, including the capsaicin recep- the node, thereby leading to conduction failure [239]. While this
tor (TRPV1) and cold receptors (TRPA1 or TRPM8) [8]. While may be the mechanism of block, it does not link C. jejuni to the
oxaliplatin fails to directly modulate these receptors, cAMP rapidly development of autoantibodies.
increases in concentration upon drug exposure, suggesting cAMP is A key breakthrough in the understanding of the pathophys-
a second messenger to a signaling cascade initiated by oxaliplatin iological link between C. jejuni and GBS came from Yuki and
that leads to TRP channel modication [8]. Additionally, Descoeur colleagues when they found rabbits that were inoculated with GM1
et al. identied two potassium channels, TREK1 and TRAAK, to ganglioside developed the axonal phenotype of GBS [295]. In a
be drastically reduced in expression, while the expression of two follow up study using this in vivo model, Yuki, et al. found the
depolarizing channels, the sodium channel and hyperpolarization lipooligosaccharide of C. jejuni to be sufcient to induce anti-GM1
activated channel (HCN), were slightly elevated [75]. These changes antibodies and paralysis consistent with GBS, and that the anti-
appeared to specic for cold sensing neurons [75]. Thus, oxaliplatin GM1 antibodies were sufcient to induce the symptoms [294]. The
treatment causes a global change in ion channel expression that ndings suggest GBS is the result of molecular mimicry between
leads to a higher membrane potential and, thus, more robust ring C. jejuni antigens and host gangliosides [294]. Much like Kochs
of TRP channels. Additionally, modeling experiments by Dimitrov postulates for infectious organisms, Ang, Jacobs, and Laman pro-
et al. have suggested [78] that the reduction in potassium current posed a number of criteria to dene molecular mimicry between
in internodal regions of myelinated axons could lead to the nerve pathogen antigens and host tissue as a cause of autoimmune dis-
ber defects rst observed by Adelsberger et al. [4], with action ease [10]. These include 1. Epidemiological association between
potential trains, and thus a stronger central response following a pathogen and disease, 2. Identication of antibodies against self
small peripheral stimulus. antigens, 3. Identication of the pathogens antigen that elicits the
In addition to cisplatin, bortezomib has also been shown to self antibodies, and 4. Recapitulation of the disease in an animal
interfere with ion channels. In a recent animal study with both model [10]. C. jejuni and GBS successfully lled all criteria and is
acute (single dose) and chronic (multiple dose) administration thus considered to be a genuine example of molecular mimicry
of bortezomib, the authors found mechanical hyperalgesia and between a pathogen and host molecular that leads to an autoim-

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048
G Model
NSL-31080; No. of Pages 18 ARTICLE IN PRESS
C.R. Cashman, A. Hke / Neuroscience Letters xxx (2015) xxxxxx 11

mune disease [10] . After the identication of the C. jejuni/GM1 to be critical for axonal protection [16,216,298], the classic prod-
relationship, other pathogen/antigen links were identied for GBS, uct of Nmnat reactions, NAD, is not required intracellularly for
including anti-GD1a ganglioside [122], anti-neurofascin [76,200] as resistance to degeneration [216], despite protection from degener-
well as antibodies against other nodal/paranodal proteins including ation with exogenous, extracellular NAD application in vitro [267].
gliomedin and contactin [76]. The motor selectivity of the AMAN These data suggest that both extracellular NAD and preserved
subtype of GBS may be due to differential ganglioside expression Nmnat function protect axons from degeneration but through two
between motor and sensory axons and myelin [186], with GM1 distinct mechanisms [216,298]. The secondary activity of Nmnat
found mainly in motor myelin [187]. However, these results were may be chaperone activity [299] or regulation of mitochondrial
challenged in a later, larger study [237], suggesting more work is metabolism [283]. Nmnat has been shown to be active within the
needed to separate the ganglioside composition of both motor and mitochondria. Several studies have shown Nmnat to reduce cal-
sensory axons and myelin in both healthy controls and patients cium spikes after axotomy [3,15], which ties the early stages of
who develop GBS. Nonetheless, anti- ganglioside antibodies that axonal degradation signaling to the later calcium dependent phase
interfere with the node of Ranvier appear to be common in many discussed above. Additionally, the WldS gene product has been
of the sensory and motor subtypes of GBS, with specic ganglioside shown to modulate energy metabolism within the axon to main-
targets being more prevalent in certain subtypes [236]. tain ATP levels following injury [221] as well as to reduce ROS
Thus, a variety of diseases and neuropathies are caused by only a production [184,199].
few mechanisms: mitochondrial dysfunction and ROS, altered sig- This arrangement of the pathway suggests that, in contrast to the
naling and inammation, and nally reduced axonal transport and late injury signal of calcium, Nmnat may produce a survival signal
channel dysfunction. While these pathophysiological mechanisms that is lost during axonal injury. Indeed, when the MPTP is activated
inevitably will be valuable for therapeutic design, recent advances in WldS mice, axons degenerate, which conrms the upstream posi-
into the downstream effectors of degeneration reveal fundamental tion of Nmnat to calcium signaling [21]. If Nmnat is transported
mechanisms of axonal degeneration and promise to be essential for throughout the axon [105], reduced transport may increase dis-
the development of treatments that may be applied universally to tal axon sensitivity to degeneration due to reduced Nmnat levels,
degeneration caused by disease as well as trauma. tying this mechanism to the axonal transport defect pathways.
Additionally, E3 ligases have been shown to balance degeneration
and regeneration, where increased activity of an E3 ligase reduces
3. New insights into mechanisms of degeneration Nmnat expression, thereby initiating degeneration [18,281], while
inhibition of the proteasome system resists Wallerian degeneration
Wallerian degeneration is the process whereby an injured axon [297]. Thus, Nmnat likely acts as a survival signal that is actively
degrades and debris is cleared to facilitate axonal regeneration and distributed throughout the axon. Once an axon is injured, Nmnat
recovery. Axonal degeneration is not a passive process like a wire levels decrease, ultimately leading to calcium inux and activation
being broken, however. Rather, it is an active process that requires of calcium dependent proteases and degradation of the axon (as
axon to soma signaling. This type of self destructive pathway is reviewed in [61]). WldS interferes with this process by resisting
reminiscent of apoptosis, but the pathways of axonal degenera- decreases in NMNAT concentration.
tion have been shown to be independent of caspases and apoptotic A loss of function mutation of a component of Toll-like recep-
signaling [97]. One can imagine this signaling may be through an tor signaling has been found to phenocopy the gain of function of
active injury signal or the loss of a survival signal. Early studies the WldS gene product, presenting an additional therapeutic target
suggested axonal degeneration is a result of the former case. After for protection from axon degeneration. A member of the Toll-like
axotomy, there is a delayed inux of calcium with subsequent acti- receptor (TLR) adaptor family, SARM/MyD88-5, was rst identi-
vation of the calcium activated protease calpain and other proteases ed to negatively regulate TLR signaling [44] and to be expressed
that is necessary and sufcient for axon degeneration and digestion in vivo in neuronal mitochondria and microtubules where it asso-
of the cytoskeleton [103,153,286]. Interestingly, inhibition of cal- ciates with JNK3 in vitro [136], a pathway known to be critical for
pain does delay Wallerian degeneration, but chelation has a much axon degeneration [223]. SARM null mice are resistant to cell death
more robust effect, suggesting the inux of calcium likely has multi- following oxygen and glucose deprivation, suggesting SARM, like
ple roles, in addition to the activation of calpain [297]. This calcium Nmnat, may be critical for neuron degeneration and death signal-
inux marks a late step toward axonal degeneration (once proteins ing [136]. These ndings were extended to axon degeneration more
begin to be degraded, the process cannot be reversed), suggesting specically, where SARM loss of function resulted in resistance
it is the result of up stream signaling, but what is this signal, and to axon degeneration following axon transection [189] and with-
what are the components of this pathway? drawal of trophic support in vitro [104]. SARM may also be activated
A curious inbred mouse strain was reported to have delayed by CaM kinase, linking this pathway to the calcium inux char-
Wallerian degeneration where the axons in a transected nerve acteristic of axonal degeneration [189]. It is likely that SARM is a
remained largely intact three days after injury, a time point common pathway toward axon degeneration from various insults,
where robust degeneration normally has begun [151]. Later studies as is the case with the downstream JNK signaling, which may be
identied the mutant gene responsible for this slowed Walle- activated by SARM or by an additional emerging pathway involv-
rian degeneration, WldS , [152,193] as a fusion of ubiquitination ing dual leucine kinase (DLK) [170]. Thus, the axon degeneration
factor E4B (Ube4b) and nicotinamide mononucleotide adenylyl- pathway appears to contain many points of convergence such that
transferase (Nmnat). The fusion protein is chiey found in the many different types of insults may result in initiation of a common
nucleus of these animals [154], although its protective activity degradation pathway.
depends on its activity in the axon [57,214]. The Ube4b:Nmnat These data suggest a model of the active process of axon
fusion results in increased stability of Nmnat versus wild type degradation. The balance of proteasomal degradation and active
Nmnat [105]. Studies in mice and drosophila have shown that the transport maintain Nmnat expression at a constant, steady state
axon protective effects of the WldS gene product requires the activ- level. In the setting of an injury, either one of these processes
ity of Nmnat [16,215], while the Ube4b fusion allows the WldS may be affected such that Nmnat levels fall below a threshold. The
gene product to bind to another protein, which may localize the decrease in Nmnat then triggers calcium inux and release from
protein fusion to the axon or mitochondria and/or allow it to inter- mitochondria, which activates calpain. Alternatively, calpain may
act with additional, unknown proteins [16]. While Nmnat appears be activated by SARM (which is itself stimulated by injury) with JNK

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048
G Model
NSL-31080; No. of Pages 18 ARTICLE IN PRESS
12 C.R. Cashman, A. Hke / Neuroscience Letters xxx (2015) xxxxxx

Fig. 2. Summary of the mechanisms of peripheral nerve degeneration. (For interpretation of the references to colour in this gure legend, the reader is referred to the web
version of this article.)
Peripheral neuropathy in a wide range of disease, including diabetes, CIPN, viral infections, and GBS, develops as a result of the interaction of various combinations of just
six processes: altered metabolism, covalent modication, impaired organelle function with ROS/RNS, altered signaling, slowed axonal transport, and altered ion channel
dynamics. The gure delineates which process is active in which disease by color, as shown in the key. All of these processes interact to ultimately converge on the neuronal
death and/or axonal degeneration pathways, with the major interactions outlined in the above gure. The different black, dark gray, and light gray coloring of the arrows is
for clarity only.

signaling. Calpain then leads to protein degradation. Additionally, degeneration marked by the increase in calcium and protease acti-
DLK may activate JNK in parallel to Nmnat loss or act to signal a dif- vation. While the details of this processes are still being elucidated,
ferent type of insult. Clearly, the details of the axonal degeneration current data suggest that within the axon, Nmnat acts as a survival
process continue to be elucidated and much work must be done to signal that decreases during axonal injury. This fall leads to the initi-
clarify how WldS (Nmnat), SARM, and DLK converge or diverge. Any ation of the degeneration pathway. While the mechanism whereby
insult that activates this pathway or modulates any of the compo- Nmnat modulates downstream effectors such as calcium is still
nents, as in mitochondrial dysfunction or slowed axonal transport, being investigated, its potential role in mitochondrial metabolism
will lead to axonal degeneration, as is seen in diabetes, CIPN, HIV is particularly interesting, given the importance of this organelle
neuropathy, HCV neuropathy, and GBS (Fig. 2). Interestingly, Nmnat to the generation of neuropathy through many diseases. The mul-
has been shown to be regulated by PKC and to modulate PARP, tipurpose role of Nmnat in NAD metabolism, chaperone activity,
which links Nmnat activity to two additional points of injury in and mitochondrial function regulation suggests that it may protect
neuropathy, as discussed above [24]. different pathologies in a variety of ways, with one modality not
being protective for all diseases (as proposed in [64] and observed
4. Conclusion in transection versus CharcotMarieTooth disease [169]). Clearly,
the protective effect of the WldS transgene is multifaceted and only
While a variety of insults, including diabetes, chemotherapy, now starting to be understood.
HIV, HCV, and C. jejuni may lead to peripheral neuropathy, there The promise of the WldS and SARM models of resistance
are only a handful of mechanisms that cause it. These include of axonal degeneration in the generation of pan-disease neuro-
altered metabolism, covalent modication of proteins, reduced protectants is reected in the many experiments showing the
organelle efciency with concomitant increases in ROS and RNS WldS gene to protect against neuropathy in a variety of dis-
production, altered intracellular signaling (e.g. PKC, RAGE) and eases, including models of spinocerebellar ataxia [299], CIPN
increased inammation, reduced axonal transport, and modulation [268,269], diabetes [278,304], myelinopathy [212], progressive
of ion channel expression and/or dynamics (Fig. 2). Alterations in motor neuronopathy [94], CharcotMarieTooth disease [169],
metabolism, as seen in diabetes, may modulate the ux through dif- traumatic axon injury [169,266], and spinal muscular atrophy
ferent metabolic pathways with concomitant changes in reactive [169]. However, the WldS gene does not cure all decits of
metabolites. Many of these mechanisms are intrinsically linked, these diseases. Indeed, WldS does not protect against disease
such as slowed mitochondrial transport exacerbating the accumu- onset or synapse abnormalities in the model of spinal muscular
lation of mitochondrial mutations during the normal aging process, atrophy [169], and it loses its effectiveness in protection from
or the increase in ROS associated with mitochondrial dysfunc- degeneration with advanced age [192]. While research contin-
tion. ues to uncover the axonal degeneration pathway, the initiating
Downstream of the damage from the pathophysiological pro- apparatus of this pathway and how it interacts with other path-
cesses, in a paradigm reminiscent of apoptosis, degeneration occurs ways (i.e., DLK) or components (e.g., SARM) have still not been
in an early, non-committed (survival) and late (death) stage of uncovered.

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048
G Model
NSL-31080; No. of Pages 18 ARTICLE IN PRESS
C.R. Cashman, A. Hke / Neuroscience Letters xxx (2015) xxxxxx 13

Clearly, while there are many new exciting avenues of research [22] B.K. Bennett, S.B. Park, C.S.-Y. Lin, M.L. Friedlander, M.C. Kiernan, D.
into the unifying mechanisms of neuropathy, and how modulation Goldstein, Impact of oxaliplatin-induced neuropathy: a patient perspective,
Support Care Cancer 20 (2012) 29592967.
of these mechanisms may help treat or prevent neuropathy, there is [23] G.J. Bennett, G.K. Liu, W.H. Xiao, H.W. Jin, C. Siau, Terminal arbor
still much work that needs to be done in understanding how axons degeneration a novel lesion produced by the antineoplastic agent
become more susceptible to injury and degeneration with age, as paclitaxel, Eur. J. Neurosci. 33 (2011) 16671676.
[24] F. Berger, C. Lau, M. Ziegler, Regulation of poly(ADP-ribose) polymerase 1
well as what, if any, commonality there is between the pathways activity by the phosphorylation state of the nuclear NAD biosynthetic
of development, degeneration, and regeneration (Fig. 2). enzyme NMN adenylyl transferase 1, Proc. Natl. Acad. Sci. 104 (2007)
37653770.
[25] A. Bierhaus, P.M. Humpert, M. Morcos, T. Wendt, T. Chavakis, B. Arnold, D.M.
Acknowledgements Stern, P.P. Nawroth, Understanding RAGE, the receptor for advanced
glycation end products, J. Mol. Med. 83 (2005) 876886.
[26] A. Bodner, P.T. Toth, R.J. Miller, Activation of c-Jun N-terminal kinase
This work was supported by the Dr. Miriam and Sheldon G. Adel- mediates gp120IIIB- and nucleoside analogue-induced sensory neuron
son Medical Research Foundation, NINDS (R01 NS-43991), and the toxicity, Exp. Neurol. 188 (2004) 246253.
Foundation for Peripheral Neuropathy. The authors wish to thank [27] W. Boehmerle, U. Spittgerber, M.B. Lazarus, K.M. McKenzie, D.G. Johnston,
D.J. Austin, B.E. Ehrlich, Paclitaxel induces calcium oscillations via an inositol
Catherine Kiefe for her artistic talents and expertise in the produc-
1,4,5-triphosphate receptor and neuronal calcium sensor 1-dependent
tion of gure two. mechanism, Proc. Natl. Acad. Sci. 103 (2006) 1835618361.
[28] W. Boehmerle, K. Zhang, M. Sivula, F.M. Heidrich, Y. Lee, S.-E. Jordt, B.E.
Ehrlich, Chronic exposure to paclitaxel diminishes phosphoinositide
References signaling by calpain-mediated neuronal calcium sensor-1 degradation, Proc.
Natl. Acad. Sci. 104 (2007) 1110311108.
[1] U.J.P.o.H.A. (UNAIDS), Global report: UNAIDS report on the global AIDS [29] B. Bonetti, F. Invernizzi, N. Rizzuto, M.L. Bonazzi, G. Zanusso, G. Chinaglia, S.
epidemic: 2010. Monaco, T-cell-mediated epineural vasculitis and humoral-mediated
[2] S. Acharjee, F. Noorbakhsh, P.L. Stemkowski, C. Olechowski, A. . Cohen, K. microangiopathy in cryoglobulinemic neuropathy, J. Neuroimmunol. 73
Ballanyi, B. Kerr, C. Pardo, P.A. Smith, C. Power, HIV-1 viral protein R causes (1997) 145154.
peripheral nervous system injury associated with in vivo neuropathic pain, [30] O. Boss, T. Hagen, B.B. Lowell, Uncoupling proteins 2 and 3: potential
FASEB J. 24 (2010). regulators of mitochondrial energy metabolism, Diabetes 49 (2000)
[3] R. Adalbert, G. Morreale, M. Paizs, L. Conforti, S.A. Walker, H.L. Roderick, 143156.
M.D. Bootman, L. Sikls, M.P. Coleman, Intra-axonal calcium changes after [31] O. Boss, S. Samec, A. Paoloni-Giacobino, C. Rossier, A. Dulloo, J. Seydoux, P.
axotomy in wild-type and slow Wallerian degeneration axons, Muzzin, J.-P. Giacobino, Uncoupling protein-3: a new member of the
Neuroscience 225 (2012) 4454. mitochondrial carrier family with tissue specic expression, FEBS Lett. 408
[4] H. Adelsberger, S. Quasthoff, J. Grosskreutz, A. Lepier, F. Eckel, C. Lersch, The (1997) 3942.
chemotherapeutic oxaliplatin alters voltage-gated Na+ channel kinetics on [32] J. Brett, A.M. Schmidt, S.D. Wan, Y.S. Zou, E. Weidman, D. Pinsky, R.
rat sensory neurons, Eur. J. Pharmacol. 406 (2000) 2532. Nowygrod, M. Neeper, C. Przysiecki, A. Shaw, A. Migheli, D. Stern, Survey of
[5] B.M. Allos, Campylobacter jejuni infections: update on emerging issues and the distribution of a newly characterized receptor for advanced glycation
trends, Clin. Infect. Dis. 32 (2001) 12011206. end products in tissues, Am. J. Pathol. 143 (1993) 16991712.
[6] M. Amadori, Atti R. Accad, Lincei 2 (1925) 337342. [33] A. Broomand, E. Jerremalm, J. Yachnin, H. Ehrsson, F. Elinder, Oxaliplatin
[7] A. Amore, P. Cirina, S. Mitola, L. Peruzzi, B. Gianoglio, I. Rabbone, C. Sacchetti, neurotoxicity-no general ion channel surface-charge effect, J. Negat. Results
F. Cerutti, C. Grillo, R. Coppoa, Nonenzymatically glycated albumin (Amadori Biomed. 8 (2009).
adducts) enhances nitric oxide synthase activity and gene expression in [34] M.R. Brown, T.J. Keith, H.R. Knull, Nonenzymatic incorporation of glucose
endothelial cells, Kidney Int. 51 (1997) 2735. and galactose into brain cytoskeletal proteins in vitro, Neurochem. Int. 21
[8] U. Anand, W.R. Otto, P. Anand, Sensitization of capsaicin and icilin responses (1992) 177183.
in oxaliplatin treated adult DRG neurons, Mol. Pain 6 (2010) 111. [35] M. Brownlee, Advanced protein glycosylation in diabetes and aging, Annu.
[9] T.D. Anderson, A. Davidovich, R. Arceo, C. Brosnan, J. Arezzo, H. Schaumburg, Rev. Med. 46 (1995) 223234.
Peripheral neuropathy induced by 2 ,3 -dideoxycytidine. A rabbit model of [36] M. Brownlee, S. Pongor, A. Cerami, Covalent attachment of solube proteins
2 ,3-dideoxycytidine neurotoxicity, Lab. Invest. 66 (1992) 6374. by nonenzymatically glycosylated collagen: role in the in situ formation of
[10] C.W. Ang, B.C. Jacobs, J.D. Laman, The GuillainBarr syndrome: a true case immune complexes, J. Exp. Med. 158 (1983) 17391744.
of molecular mimicry, Trends Immunol. 25 (2004) 6166. [37] M. Brownlee, H. Vlassara, A. Cerami, Nonenzymatic glycosylation products
[11] S. Apostolski, T. McAlarney, A.P. Hays, N. Latov, Complement dependent on collagen covalently trap low-density lipoprotein, Diabetes 34 (1985)
cytotoxicity of sensory ganglion neurons mediated by the gp120 938941.
glycoprotein of HIV-1, Immunol. Invest. 23 (1994) 4752. [38] R. Bucala, K.J. Tracey, A. Cerami, Advanced glycosylation products quench
[12] A.A. Argyriou, A. Kyritsis, T. Makatsoris, H. Kalofonos, nitric oxide and mediate defective endothelium-dependent vasodilatation
Chemotherapy-induced peripheral neuropathy in adults: a comprehensive in experimental diabetes, J. Clin. Invest. 87 (1991) 432438.
update of the literature, Cancer Manag. Res. 6 (2014) 135147. [39] M.G. Buse, Hexosamines, insulin resistance, and the complications of
[13] C.E. Aubert, P.-L. Michel, P. Gillery, S. Jaisson, M. Fonfrede, F. Morel, A. diabetes: current status, Am. J. Physiol. Endocrinol. Metab. 290 (2006)
Hartemann, O. Bourron, Association of peripheral neuropathy with E1E8.
circulating advanced glycation end products, soluble receptor for advanced [40] P. Cacoub, C. Renou, E. Rosenthal, P. Cohen, I. Loury, V. Loustaud-Ratti, A.-M.
glycation end products and other risk factors in patients with type 2 Yamamoto, A.-C. Camproux, P. Hausfater, L. Musset, P. Veyssier, G. Raguin,
diabetes, Diabetes Metab. Res. Rev. 30 (2014) 674685. J.-C. Piette, GERMIVIC, Extrahepatic manifestations associated with hepatitis
[14] F.-J. Authier, G. Bassez, C. Payan, L. Guillevin, J.-M. Pawlotsky, J.-D. Degos, C virus infection: a prospective multicenter study of 321 patients, Medicine
R.K. Gherardi, L. Belec, Detection of genomic viral RNA in nerve and muscle 79 (2000) 4756.
of patients with HCV neuropathy, Neurology 60 (2003) 808812. [41] D. Cai, D.P. McEwan, J.R. Martens, E. Meyhofer, K.J. Verhey, Single molecule
[15] M.A. Avery, T.M. Rooney, J.D. Pandya, T.M. Wishart, T.H. Gillingwater, J.W. imaging reveals differences in microtubule track selection between kinesin
Geddes, P.G. Sullivan, M.R. Freeman, WldS prevents axon degeneration motors, PLoS Biol. 7 (2009) 114.
through increased mitochondrial ux and enhanced mitochondrial Ca2+ [42] A.-V. Cantero, M. Portero-Otn, V. Ayala, N. Auge, M. Sanson, M. Elbaz, J.-C.
buffering, Curr. Biol. 22 (2012) 596600. Thiers, R. Pamplona, R. Salvayre, A. Ngre-Salvayre, Methylglyoxal induces
[16] M.A. Avery, A.E. Sheehan, K.S. Kerr, J. Wang, M.R. Freeman, WldS requires advanced glycation end product (AGEs) formation and dysfunction of PDGF
Nmnat1 enzymatic activity and N16-VCP interactions to suppress Wallerian receptor-: implications for diabetic atherosclerosis, FASEB J. 21 (2007)
degeneration, J. Cell Biol. 184 (2009) 501513. 30963106.
[17] P.W. Baas, M.M. Black, Individual microtubules in the axon consist of [43] M. Capasso, C.M. Caporale, F. Pomilio, P. Gandol, A. Lugaresi, A. Uncini,
domains that differ in both composition and stability, J. Cell Biol. 111 (1990) Acute motor conduction block neuropathy: another GuillainBarr variant,
495509. Neurology 61 (2003) 617622.
[18] E. Babetto, B. Beirowski, E.V. Russler, J. Milbrandt, A. DiAntonio, The Phr1 [44] M. Carty, R. Goodbody, M. Schrder, J. Stack, P.N. Moynagh, A.G. Bowie, The
ubiquitin ligase promotes injury- induced axon self-destruction, Cell Rep. 3 human adaptor SARM negatively regulates adaptor protein TRIF-dependent
(2013) 14221429. toll-like receptor signaling, Nat. Immunol. 7 (2006) 10741081.
[19] R.S. Balaban, S. Nemoto, T. Finkel, Mitochondria, oxidants, and aging, Cell [45] G. Cavaletti, Chemotherapy-induced peripheral neurotoxicity (CIPN): what
120 (2005) 483495. we need and what we know, J. Peripher. Nerv. Syst. 19 (2014) 6676.
[20] G. Barja, A. Herrero, Localization at complex I and mechanism of the higher [46] G. Cavaletti, E. Cavaletti, P. Montaguti, N. Oggioni, O. De Negri, G. Tredici,
free radical production of brain nonsynaptic mitochondria in the short-lived Effect on the peripheral nervous system of the short-term intravenous
rat than in the longevous pigeon, J. Bioenerg. Biomembr. 30 (1998) 235243. admininstration of paclitaxel in the rat, Neurotoxicology 18 (1997) 137145.
[21] S.A. Barrientos, N.W. Martinez, S. Yoo, J.S. Jara, S. Zamorano, C. Hetz, J.L. [47] CDC, National diabetes statistics report: Estimates of diabetes and its burden
Twiss, J. Alvarez, F.A. Court, Axonal degeneration is mediated by the in the United States, 2014. U.S. Department of Health and Human Services,
mitochondrial permeability transition pore, J. Neurosci. 31 (2011) 966978. Atlanta, GA, (2014).

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048
G Model
NSL-31080; No. of Pages 18 ARTICLE IN PRESS
14 C.R. Cashman, A. Hke / Neuroscience Letters xxx (2015) xxxxxx

[48] D. Chad, K. Pariser, W.G. Bradley, L.S. Adelman, V.W. Pinn, The pathogenesis [76] J.J. Devaux, M. Odaka, N. Yuki, Nodal proteins are target antigens in
of cryglobulinemic neuropathy, Neurology 32 (1982) 725729. GuillainBarr syndrome, J. Peripher. Nerv. Syst. 17 (2012) 6271.
[49] C.B. Chan, D. De Leo, J.W. Joseph, T.S. McQuaid, X.F. Ha, F. Xu, R.G. Tsushima, [77] L. Di Cesare Mannelli, M. Zanardelli, P. Failli, C. Ghelardini,
P.S. Pennefather, A.M.G. Salapatek, M.B. Wheeler, Increased uncoupling Oxaliplatin-induced neuropathy: oxidative stress as pathological
protein-2 levels in -cells are associated with impaired glucose-stimulated mechanism. Protective effect of silibinin, J. Pain 13 (2012) 276284.
insulin secretion: mechanism of action, Diabetes 50 (2001) 13021310. [78] A.G. Dimitrov, N.A. Dimitrova, A possible link of oxaliplatin-induced
[50] C.B. Chan, P.E. MacDonald, M.C. Saleh, D.C. Johns, E. Marbn, M.B. Wheeler, neuropathy with potassium channel decit, Muscle Nerve 45 (2012)
Overexpression of uncoupling protein 2 inhibits glucose-stimulated insulin 403411.
secretion from rat islets, Diabetes 48 (1999) 14821486. [79] Y. Ding, A. Kantarci, H. Hasturk, P.C. Trackman, A. Malabanan, T.E. Van Dyke,
[51] G. Charnogursky, H. Lee, N. Lopez, Diabetic neuropathy, in: J. Biller, J.M. Ferro Activation of RAGE induces elevated O2 generation by mononuclear
(Eds.), Handbook of Clinical Neurology, vol. 120, Elsevier, 2014, pp. 773785. phagocytes in diabetes, J. Leukocyte Biol. 81 (2007) 520527.
[52] M.-P. Chaunu, H. Ratinahirana, M. Raphael, D. Hnin, C. Leport, F. [80] X. Du, T. Matsumura, D. Edelstein, L. Rossetti, Z. Zsengellr, C. Szab, M.
Brun-Vezinet, J.-M. Lger, P. Brunet, J.-J. Hauw, The spectrum of changes on Brownlee, Inhibition of GAPDH activity by poly(ADP- ribose) polymerase
20 nerve biopsies in patients with HIV infection, Muscle Nerve 12 (1989) activates three major pathways of hyperglycemic damage in endothelial
452459. cells, J. Clin. Invest. 112 (2003) 10491057.
[53] C. Cheng, K. Tsuneyama, R. Kominami, H. Shinohara, S. Sakurai, H. Yonekura, [81] X.-L. Du, D. Edelstein, L. Rossetti, I.G. Fantus, H. Goldberg, F. Ziyadeh, J. Wu,
T. Watanabe, Y. Takano, H. Yamamoto, Y. Yamamoto, Expression proling of M. Brownlee, Hyperglycemia-induced mitochondrial superoxide
endogenous secretory receptor for advanced glycation end products in overproduction activates the hexosamine pathway and induces
human organs, Mod. Pathol. 18 (2005) 13851396. plasminogen activator inhibitor-1 expression by increasing Sp1
[54] H.T. Cheng, J.R. Dauch, J.M. Hayes, Y. Hong, E.L. Feldman, Nerve growth glycosylation, Proc. Natl. Acad. Sci. 97 (2000) 1222212226.
factor mediates mechanical allodynia in a mouse model of type 2 diabetes, J. [82] X.L. Du, D. Edelstein, S. Dimmeler, Q. Ju, C. Sui, M. Brownlee, Hyperglycemia
Neuropathol. Exp. Neurol. 68 (2009) 12291243. inhibits endothelial nitric oxide synthase activity by posttranslational
[55] S.K.R. Chowdhury, D.R. Smith, A. Saleh, J. Schapansky, A. Marquez, S. Gomes, modication at the Akt site, J. Clin. Invest. 108 (2001) 13411348.
E. Akude, D. Morrow, N.A. Calcutt, P. Fernyhough, Impaired adenosine [83] S. Dunn, E.E. Morrison, T.B. Liverpool, C. Molina-Pars, R.A. Cross, M.C.
monophosphate-activated protein kinase signalling in dorsal root ganglia Alonso, M. Peckham, Differential trafcking of Kif5c on tyrosinated and
neurons is linked to mitochondrial dysfunction and peripheral neuropathy detyrosinated microtubules in live cells, J. Cell Sci. 121 (2008) 10851095.
in diabetes, Brain 135 (2012) 17511766. [84] B. Duran-Jimenez, D. Dobler, S. Moffatt, N. Rabbani, C.H. Streuli, P.J.
[56] S.K.R. Chowdhury, E. Zherebitskaya, D.R. Smith, E. Akude, S. Chattopadhyay, Thornalley, D.R. Tomlinson, N.J. Gardiner, Advanced glycation end products
C.G. Jolivalt, N.A. Calcutt, P. Fernyhough, Mitochondrial respiratory chain in extracellular matrix proteins contribute to the failure of sensory nerve
dysfunction in dorsal root ganglia of streptozotocin-induced diabetic rats regeneration in diabetes, Diabetes 58 (2009) 28932903.
and its correction by insulin treatment, Diabetes 59 (2010) 10821091. [85] K.S. Echtay, E.W. Inkler, K. Frischmuth, M. Klingenberg, Uncoupling proteins
[57] M.S. Cohen, A.K. Ghosh, H.J. Kim, N.L. Jeon, S.R. Jaffrey, Chemical 2 and 3 are highly active H+ transporters and highly nucelotide sensitive
genetic-mediated spatial regulation of protein expression in neurons when activated by coenzyme Q (ubiquinone), Proc. Natl. Acad. Sci. 98 (2001)
reveals an axonal function for WldS , Chem. Biol. 19 (2012) 179187. 14161421.
[58] S. Cohen, R. Levi-Montalcini, A nerve growth-stimulating factor isolated [86] J.L. Edwards, A. Quattrini, S.I. Lentz, C. Figueroa-Romero, F. Cerri, C. Backus,
from snake venom, Proc. Natl. Acad. Sci. 42 (1956) 571574. Y. Hong, E.L. Feldman, Diabetes regulates mitochondrial biogenesis and
[59] S. Cohen, R. Levi-Montalcini, Purication and properties of a nerve ssion in mouse neurons, Diabetologia 53 (2010) 160169.
growth-promoting factor isolated from mouse sarcoma 180, Cancer Res. 17 [87] R.J. Ellis, D. Rosario, D.B. Clifford, J.C. McArthur, D. Simpson, T. Alexander,
(1957) 1520. B.B. Gelman, F. Vaida, A. Collier, C.M. Marra, B. Ances, J.H. Atkinson, R.H.
[60] S. Cohen, R. Levi-Montalcini, V. Hamburger, A nerve growth-stimulating Dworkin, S. Morgello, I. Grant, CHARTER Study Group, Continued high
factor isolated from sarcomas 37 and 180, Proc. Natl. Acad. Sci. 40 (1954) prevalence and adverse clinical impact of human immunodecency virus-
10141018. associated sensory neuropathy in the era of combination antiretroviral
[61] M. Coleman, Axon degeneration mechanisms: commonality amid diversity, therapy: the CHARTER study, Arch. Neurol. 67 (2010) 552558.
Nat. Rev. Neurosci. 6 (2005) 889898. [88] G. Fattovich, G. Giustina, S. Favarato, A. Ruol, Investigators of the Italian
[62] H. Colognato, C. ffrench-Constant, M.L. Feltri, Human diseases reveal novel Associaton for the Study of the Liver, A survey of adverse events in 11 241
roles for neural laminins, Trends Neurosci. 28 (2005) 480486. patients with chronic viral hepatitis treated with alfa interferon, J. Hepatol.
[63] F.I. Comer, G.W. Hart, Reciprocity between O-GlcNAc and O-phosphate on 24 (1996) 3847.
the carboxy terminal domain of RNA polymerase II, Biochemistry 40 (2001) [89] M. Federici, R. Menghini, A. Mauriello, M.L. Hribal, F. Ferrelli, D. Lauro, P.
78457852. Sbraccia, L.G. Spagnoli, G. Sesti, R. Lauro, Insulin dependent activation of
[64] L. Conforti, J. Gilley, M.P. Coleman, Wallerian degeneration: an emerging endothelial nitric oxide synthase is impaired by O-linked glycosylation
axon death pathway linking injury and disease, Nat. Rev. Neurosci. 15 modication of signaling proteins in human coronary endothelial cells,
(2014) 394409. Circulation 106 (2002) 466472.
[65] K.A. Conklin, Chemotherapy-associated oxidative stress: impact on [90] H.J. Federoff, D. Lawrence, M. Brownlee, Nonenzymatic glycosylation of
chemotherapeutic effectiveness, Integr. Cancer Ther. 3 (2004) 294300. laminin and the laminin peptide CIKVAVS inhibits neurite outgrowth,
[66] L. Conti, L. Fantuzzi, M. Del Corn, F. Belardelli, S. Gessani, Diabetes 42 (1993) 509513.
Immunomodulatory effects of the HIV-1 gp120 protein on antigen [91] D. Feldman, C. Brosnan, T.D. Anderson, Ultrastructure of peripheral
presenting cells: implications for AIDS pathogenesis, Immunobiology 209 neuropathy induced in rabbits by 2,3-dideoxycytidine, Lab. Invest. 66
(2004) 99115. (1992) 7585.
[67] D.R. Cornblath, J.C. McArthur, Predominantly sensory neuropathy in patients [92] V. Fensterl, G.C. Sen, Interferons and viral infections, Biofactors 35 (2009)
with AIDS and AIDS-related complex, Neurology 38 (1988) 794796. 1420.
[68] H.C.F. Ct, Z.L. Brumme, K.J.P. Craib, M. Math, C.S. Alexander, B. Wynhoven, [93] P. Fernyhough, L.T. Diemel, J. Hardy, W.J. Brewster, L. Mohiuddin, D.R.
L. Ting, H. Wong, M. Harris, P.R. Harrigan, M.V. OShaughnessy, J.S.G. Tomlinson, Human recombinant nerve growth factor replaces decient
Montaner, Changes in mitochondrial DNA as a marker of nucleoside toxicity neurotrophic support in the diabetic rat, Eur. J. Neurosci. 7 (1995)
in HIV-infected patients, N. Engl. J. Med. 346 (2002) 811820. 11071110.
[69] N.A. Cullum, J. Mahon, K. Stringer, W.G. McLean, Glycation of rat sciatic nerve [94] A. Ferri, J.R. Sanes, M.P. Coleman, J.M. Cunningham, A.C. Kato, Inhibiting axon
tubulin in experimental diabetes mellitus, Diabetologia 34 (1991) 387389. degeneration and synapse loss attenuates apoptosis and disease progression
[70] A. Cumaoglu, G. Ozansoy, A.M. Irat, A. Aricioglu, C. Karasu, N. Ari, Effect of in a mouse model of motoneuron disease, Curr. Biol. 13 (2003) 669673.
long term, non cholesterol lowering dose of uvastatin treatment on [95] C. Ferri, A.L. Zignego, S.A. Pileri, Cryoglobulins, J. Clin. Pathol. 55 (2002) 413.
oxidative stress in brain and peripheral tissues of streptozotocin-diabetic [96] J. Ferrier, V. Pereira, J. Busserolles, N. Authier, D. Balayssac, Emerging trends
rats, Eur. J. Pharmacol. 654 (2011) 8085. in understanding chemotherapy-induced peripheral neuropathy, Curr. Pain
[71] M.C. Dalakas, Peripheral neuropathy and antiretroviral drugs, J. Peripher. Headache Rep. 17 (2013) 364372.
Nerv. Syst. 6 (2001) 1420. [97] J.T. Finn, M. Weil, F. Archer, R. Siman, A. Srinivasan, M.C. Raff, Evidence that
[72] M.C. Dalakas, C. Semino-Mora, M. Leon-Monzon, Mitochondrial alterations Wallerian degeneration and localized axon degeneration induced by local
with mitochondrial DNA depletion in the nerves of AIDS patients with neurotrophin deprivation do no involve caspases, J. Neurosci. 20 (2000)
peripheral neuropathy induced by 2 3 -dideoxycytidine (ddC), Lab. Invest. 13331341.
81 (2001) 15371544. [98] S.J.L. Flatters, G.J. Bennett, Studies of peripheral sensory nerves in
[73] N. Das Evcimen, G.L. King, The role of protein kinase C activation and the paclitaxel-induced painful peripheral neuropathy: evidence for
vascular complications of diabetes, Pharmacol. Res. 55 (2007) 498510. mitochondrial dysfunction, Pain 122 (2006) 245257.
[74] S. de la Monte, D.H. Gabuzda, D.D. Ho, R.H. Brown Jr., E.T. Hedley-Whyte, R.T. [99] C. Fleury, M. Neverova, S. Collins, S. Raimbault, O. Champigny, C.
Schooley, M.S. Hirsch, A.K. Bhan, Peripheral neuropathy in the acquired Levi-Meyrueis, F. Bouillaud, M.F. Seldin, R.S. Surwit, D. Ricquier, C.H.
immunodeciency syndrome, Ann. Neurol. 23 (1988) 485492. Warden, Uncoupling protein-2: a novel gene linked to obesity and
[75] J. Descoeur, V. Pereira, A. Pizzoccaro, A. Francois, B. Ling, V. Maffre, B. hyperinsulinemia, Nat. Genet. 15 (1997) 269272.
Couette, J. Busserolles, C. Courteix, J. Noel, M. Lazdunski, A. Eschalier, N. [100] S. Fujimoto, K. Amako, Guillain-Barr syndrome and Campylobacter jejuni
Authier, E. Bourinet, Oxaliplatin-induced cold hypersensitivity is due to infection, Lancet 335 (1990) 1350.
remodelling of ion channel expression in nociceptors, EMBO Mol. Med. 3 [101] K.H. Gabbay, L.O. Merola, R.A. Field, Sorbitol pathway: presence in nerve and
(2011) 266278. cord with substrate accumulation in diabetes, Science 151 (1966) 209210.

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048
G Model
NSL-31080; No. of Pages 18 ARTICLE IN PRESS
C.R. Cashman, A. Hke / Neuroscience Letters xxx (2015) xxxxxx 15

[102] S. Genuth, Insights from the diabetes control and complications [128] M.A. Jordan, L. Wilson, Microtubules as a target for anticancer drugs, Nat.
trial/epidemiology of diabetes interventions and complications study on the Rev. Cancer 4 (2004) 253265.
use of intensive glycemic treatment to reduce the risk of complications of [129] J.K. Juranek, M.S. Geddis, R. Rosario, A.M. Schmidt, Impaired slow axonal
type 1 diabetes, Endocr. Pract. 12 (2006) 3441. transport in diabetic peripheral nerve is independent of RAGE, Eur. J.
[103] E.B. George, J.D. Glass, J.W. Grifn, Axotomy-induced axonal degeneration is Neurosci. 38 (2013) 31593168.
mediated by calcium inux through ion-specic channels, J. Neurosci. 15 [130] T.N. Kakuda, Pharmacology of nucleoside and nucleotide reverse
(1995) 64456452. transcriptase inhibitor-induced mitochondrial toxicity, Clin. Ther. 22 (2000)
[104] J. Gerdts, D.W. Summers, Y. Sasaki, A. DiAntonio, J. Milbrandt, 685708.
Sarm1-mediated axon degeneration requires both SAM and TIR interactions, [131] P.R. Kamerman, P.J. Moss, J. Weber, V.C.J. Wallace, A.S.C. Rice, W. Huang,
J. Neurosci. 33 (2013) 1356913580. Pathogenesis of HIV-associated sensory neuropathy: evidence from in vivo
[105] J. Gilley, M.P. Coleman, Endogenous Nmnat2 is an essential survival factor and in vitro experimental models, J. Peripher. Nerv. Syst. 17 (2012) 1931.
for maintenance of healthy axons, PLoS Biol. 8 (2010) e1000300. [132] S. Keilbaugh, G.A. Hobbs, M.V. Simpson, Effect of 2,3-dideoxycytidine on
[106] E. Gornstein, T.L. Schwarz, The paradox of paclitaxel neurotoxicity: oxidative phosphorylation in the PC2 cell, a neuronal model, Biochem.
mechanisms and unanswered questions, Neuropharmacology 76 (2014) Pharmacol. 53 (1997) 14851492.
175183. [133] S.C. Keswani, C.A. Pardo, C.L. Cherry, A. Hoke, J.C. McArthur, HIV-associated
[107] D.A. Greene, S. Yagihashi, S.A. Lattimer, A.A.F. Sima, Nerve Na+-K+-ATPase, sensory neuropathies, AIDS 16 (2002) 21052117.
conduction, and myo-inositol in the insulin-decient BB rat, Am. J. Physiol. [134] S.C. Keswani, M. Polley, C.A. Pardo, J.W. Grifn, J.C. McArthur, A. Hoke,
247 (1984) E534E539. Schwann cell chemokine receptors mediate HIV-1 gp120 toxicity to sensory
[108] E.W. Gregg, P. Sorlie, R. Paulose-Ram, Q. Gu, M.S. Eberhardt, M. Wolz, V. neurons, Ann. Neurol. 54 (2003) 287296.
Burt, L. Curtin, M. Engelgau, L. Geiss, Prevalence of lower-extremity disease [135] J.F. Kidd, M.F. Pilkington, M.J. Schell, K.E. Fogarty, J.N. Skepper, C.W. Taylor, P.
in the U.S. adult population 40 years of age with and without diabetes, Thorn, Paclitaxel affects cytosolic calcium signals by opening the
Diabetes Care 27 (2004) 15911597. mitochondrial permeability transition pore, J. Biol. Chem. 277 (2002)
[109] J.W. Grifn, C.Y. Li, T.W. Ho, P. Xue, C. Macko, C.Y. Gao, C. Yang, M. Tian, B. 65046510.
Mishu, D.R. Cornblath, G.M. McKhann, A.K. Asbury, GuillainBarr syndrome [136] Y. Kim, P. Zhou, L. Qian, J.-Z. Chuang, J. Lee, C. Li, C. Iadecola, C. Nathan, A.
in northern China: the spectrum of neuropathological changes in clinically Ding, MyD88-5 links mitochondria, microtubules and JNK3 in neurons and
dened cases, Brain 118 (1995) 577595. regulates neuronal survival, J. Exp. Med. 204 (2007) 20632074.
[110] TDCACTR, Group, The effect of intensive treatment of diabetes on the [137] M. Kirstein, C. Aston, R. Hinz, H. Vlassara, Receptor-specic induction of
development and progression of long-term complications in insulin-like growth factor I in human monocytes by advanced glycosylation
insulin-dependent diabetes mellitus, N. Engl. J. Med. 329 (1993) 977986. end product-modied proteins, J. Clin. Invest. 90 (1992) 439446.
[111] G. Guillain, J.-A. Barr, A. Strohl, Sur un syndrome de radiculonvrite avec [138] N. Kokubun, M. Nishibayashi, A. Uncini, M. Odaka, K. Hirata, N. Yuki,
hyperalbuminose du liquide cphalo-rachidien sans raction cellulaire: Conduction block in acute motor axonal neuropathy, Brain 133 (2010)
remarques sur les caractres cliniques et graphiques des rexes tendineux, 28972908.
Bull. Soc. Md. Hp. Paris 40 (1916) 14621470. [139] Y. Konishi, M. Setou, Tubulin tyrosination navigates the kinesin-1 motor
[112] H.C. Ha, L.D. Hester, S.H. Snyder, Poly(ADP-ribose) polymerase-1 domain to axons, Nat. Neurosci. 12 (2009) 559567.
dependence of stress-induced transcription factors and associated gene [140] S. Kuwabara, N. Yuki, Axonal GuillainBarr syndrome: concepts and
expression in glia, Proc. Natl. Acad. Sci. 99 (2002) 32703275. controversies, Lancet Neurol. 12 (2013) 11801188.
[113] H.C. Ha, S.H. Snyder, Poly(ADP-ribose) polymerase is a mediator of necrotic [141] S. Kuwabara, N. Yuki, M. Koga, T. Hattori, D. Matsuura, M. Miyake, M. Noda,
cell death by ATP depletion, Proc. Natl. Acad. Sci. 96 (1999) 1397813982. IgG anti-GM1 antibody is associated with reversible conduction failure and
[114] C. Hafer-Macko, S.-T. Hsieh, C.Y. Li, T.W. Ho, K. Sheikh, D.R. Cornblath, G.M. axonal degeneration in GuillainBarr syndrome, Ann. Neurol. 44 (1998)
McKhann, A.K. Asbury, J.W. Grifn, Acute motor axonal neuropathy: an 202208.
antibody-mediated attack on axolemma, Ann. Neurol. 40 (1996) 635644. [142] T.H. Landowski, C.J. Megli, K.D. Nullmeyer, R.M. Lynch, R.T. Dorr,
[115] K. Hahn, B. Robinson, C. Anderson, W. Li, C.A. Pardo, S. Morgello, D. Simpson, Mitochondrial-mediated disregulation of Ca2+ is a critical determinent of
A. Nath, Differential effects of HIV infected macrophages on dorsal root velcade (PS-341/bortezomib) cytotoxicity in myeloma call lines, Cancer Res.
ganglia neurons and axons, Exp. Neurol. 210 (2008) 3040. 65 (2005) 28363828.
[116] G. Hajnczky, E. Davies, M. Madesh, Calcium signaling and apoptosis, [143] D. Lavanchy, Evolving epidemiology of hepatits C virus, Clin. Microbiol.
Biochem. Biophys. Res. Commun. 304 (2003) 445454. Infect. 17 (2011) 107115.
[117] J.W. Hammond, C.-F. Huang, S. Kaech, C. Jacobson, G. Banker, K.J. Verhey, [144] H.C. Lehmann, W. Chen, J. Borzan, J.L. Mankowski, A. Hke, Mitochondrial
Posttranslational modications of tubulin and the polarized transport of dysfunction in distal axons contributes to human immunodeciency virus
kinesin-1 in neurons, Mol. Biol. Cell 21 (2010) 572583. sensory neuropathy, Ann. Neurol. 69 (2011) 100110.
[118] Y. Han, M.T. Smith, Pathobiology of cancer chemotherapy- induced [145] G.M. Leinninger, C. Backus, A.M. Sastry, Y.-B. Yi, C.-W. Wang, E.L. Feldman,
peripheral neuropathy (CIPN), Front. Pharmacol. 4 (2013) 116. Mitochondria in DRG neurons undergo hyperglycemic mediated injury
[119] W. Haymaker, J.W. Kernohan, The LandryGuillainBarr syndrome: a through BimBax and the ssion protein Drp1, Neurobiol. Dis. 23 (2006)
clinicopathologic report of fty fata cases and a critique of the literature, 1122.
Medicine 28 (1949) 59141. [146] R. Levi-Montalcini, Effects of mouse tumor transplantation on the nervous
[120] F. Hellal, A. Hurtado, J. Ruschel, K.C. Flynn, C.J. Laskowski, M. Umlauf, L.C. system, Ann. N. Y. Acad. Sci. 55 (1952) 330344.
Kapitein, D. Strikis, V. Lemmon, J. Bixby, C.C. Hoogenraad, F. Bradke, [147] R. Levi-Montalcini, S. Cohen, In vitro and in vivo effects of a nerve
Microtubule stabilization reduces scarring and causes axon regeneration growth-stimulating agent isolated from snake venom, Proc. Natl. Acad. Sci.
after spinal cord injury, Science 331 (2011) 928931. 42 (1956) 695699.
[121] T.W. Ho, B. Mishu, C.Y. Li, C.Y. Gao, D.R. Cornblath, J.W. Grifn, A.K. Asbury, [148] Q.-H. Liu, D.A. Williams, C. McManus, F. Baribaud, R.W. Doms, D. Schols, E.
M.J. Blaser, G.M. McKhann, GuillainBarr syndrome in northern China: De Clercq, M.I. Kotlikoff, R.G. Collman, B.D. Freedman, HIV-1 gp120 and
relationship to Campylobacter jejuni infection and anti-glycolipid antibodies, chemokines activate ion channels in primary macrophages through CCR5
Brain 118 (1995) 597605. and CXCR4 stimulation, Proc. Natl. Acad. Sci. 97 (2000) 48324837.
[122] T.W. Ho, H.J. Willison, I. Nachamkin, C.Y. Li, J. Veitch, H. Ung, G.R. Wang, R.C. [149] T.E. Lloyd, J. Machamer, K. OHara, J.H. Kim, S.E. Collins, M.Y. Wong, B. Sahlin,
Liu, D.R. Cornblath, A.K. Asbury, J.W. Grifn, G.M. McKhann, Anti-GD1a W. Imlach, Y. Yang, E.S. Levitan, B.D. McCabe, A.L. Kolodkin, The p150(Glued)
antibody is associated with axonal but not demyelinating forms of CAP-Gly domain regulates initiation of retrograde transport at synaptic
GuillainBarr syndrome, Ann. Neurol. 45 (1999) 168173. termini, Neuron 74 (2012) 344360.
[123] A. Hke, M. Morris, N.J. Haughey, GPI-1046 protects dorsal root ganglia from [150] C. Lu, J.C. He, W. Cai, H. Liu, L. Zhu, H. Vlassara, Advanced glycation
gp120-induced axonal injury by modulating store-operated calcium entry, J. endproduct (AGE) receptor 1 is a negative regulator of the inammatory
Peripher. Nerv. Syst. 14 (2009) 2735. response to AGE in mesangial cells, Proc. Natl. Acad. Sci. 101 (2004)
[124] B.I. Hudson, A.Z. Kalea, M. Del Mar Arriero, E. Harja, E. Boulanger, V. DAgati, 1176711772.
A.M. Schmidt, Interaction of the RAGE cytoplasmic domain with [151] E.R. Lunn, V.H. Perry, M.C. Brown, H. Rosen, S. Gordon, Absence of Wallerian
diaphanous-1 is required for ligand-stimulated cellular migration through degeneration does not hinder regeneration in peripheral nerve, Eur. J.
activation of Rac1 and Cdc42, J. Biol. Chem. 283 (2008) 3445734468. Neurosci. 1 (1989) 2733.
[125] K. Janes, T. Doyle, L. Bryant, E. Esposito, S. Cuzzocrea, J. Ryerse, G.J. Bennett, [152] M.F. Lyon, B.W. Ogunkolade, M.C. Brown, D.J. Atherton, V.H. Perry, A gene
D. Salvemini, Bioenergetic decits in peripheral nerve sensory axons during affecting Wallerian nerve degeneration maps distally on mouse
chemotherapy-induced neuropathic pain resulting from chromosome 4, Proc. Natl. Acad. Sci. 90 (1993) 97179720.
peroxynitrite-mediated post-translational nitration of mitochondrial [153] M. Ma, T.A. Ferguson, K.M. Schoch, J. Li, Y. Qian, F.S. Shofer, K.E. Saatman,
superoxide dismutase, Pain 154 (2013) 24322440. R.W. Neumar, Calpains mediate axonal cytoskeleton disintegration during
[126] Y. Jiang, C. Guo, M.R. Vasko, M.R. Kelley, Implications of Wallerian degeneration, Neurobiol. Dis. 56 (2013) 3446.
apurinic/apyrimidinic endonuclease in reactive oxygen signaling reponse [154] T.G.A. Mack, M. Reiner, B. Beirowski, W. Mi, M. Emanuelli, D. Wagner, D.
after cisplatin treatment of dorsal root ganglion neurons, Cancer Res. 68 Thomson, T. Gillingwater, F.A. Court, L. Conforti, F.S. Fernando, A. Tarlton, C.
(2008) 64256434. Andressen, K. Addicks, G. Magni, R.R. Ribchester, V.H. Perry, M.P. Coleman,
[127] G. Jones, Y. Zhu, C. Silva, S. Tsutsui, C.A. Pardo, O.T. Keppler, J.C. McArthur, C. Wallerian degeneration of injured axons and synapses is delayed by a
Power, Peripheral nerve-derived HIV-1 is predominantly CCR5-dependent Ube4b/Nmnat chimeric gene, Nat. Neurosci. 4 (2001) 11991206.
and causes neuronal degeneration and neuroinammation, Virology 334 [155] L.C. Maillard, Action des acides amins sur les scures: formation des
(2005) 178193. mslanodines par voie mthodique, C. R. Acad. Sci. 154 (1912) 6668.

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048
G Model
NSL-31080; No. of Pages 18 ARTICLE IN PRESS
16 C.R. Cashman, A. Hke / Neuroscience Letters xxx (2015) xxxxxx

[156] G. Majno, M.L. Karnovsky, A biochemical and morphologic study of ganglion of rats with paclitaxel-induced neuropathy, Cancer Sci. 99 (2008)
myelination and demyelination: II. Lipogenesis in vitro by rat nerves 16181625.
following transection, J. Exp. Med. 108 (1958) 197214. [183] T. Nishikawa, D. Edelstein, X.-L. Du, T. S.-i. Yamagishi, Matsumura, Y.
[157] Y. Mao, FORMIN a link between kinetochores and microtubule ends, Trends Kaneda, M.A. Yorek, D. Beebe, P.J. Oates, H.-P. Hammes, I. Giardino, M.
Cell Biol. 21 (2011) 625629. Brownlee, Normalizing mitochondrial superoxide production blocks three
[158] J.L. Martin, C.E. Brown, N. Matthews-Davis, J.E. Reardon, Effects of antiviral pathways of hyperglycaemic damage, Nature 404 (2000) 787790.
nucleoside analogs on human DNA polymerases and mitochondrial DNA [184] K.C. ODonnell, M.E. Vargas, Sagasti, WldS and PGC- 1 regulated
synthesis, Antimicrob. Agents Chemother. 38 (1994) 27432749. mitochondrial transport and oxidation state after axonal injury, J. Neurosci.
[159] J.C. McArthur, B.J. Brew, A. Nath, Neurological complications of HIV 33 (2013) 1477814790.
infection, Lancet Neurol. 4 (2005) 543555. [185] I.G. Obrosova, V.R. Drel, P. Pacher, O. Ilnytska, Z.Q. Wang, M.J. Stevens, M.A.
[160] E.S. McDonald, K.R. Randon, A. Knight, A.J. Windebank, Cisplatin Yorek, Oxidative-nitrosative stress and poly(ADP- ribose) polymerase
preferentially binds to DNA in dorsal root ganglion neurons in vitro and in (PARP) activation in experimental diabetic neuropathy: the relation is
vivo: a potential mechanism for neurotoxicity, Neurobiol. Dis. 18 (2005) revisted, Diabetes 54 (2005) 34353441.
305313. [186] K. Ogawa-Goto, N. Funamoto, T. Abe, K. Nagashima, Different ceramide
[161] E.S. McDonald, A.J. Windebank, Cisplatin-induced apoptosis of DRG neurons compositions of gangliosides between human motor and sensory nerves, J.
involves bax redistribution and cytochrome c release but not fas receptor Neurochem. 55 (1990) 14861493.
signaling, Neurobiol. Dis. 9 (2002) 220233. [187] K. Ogawa-Goto, N. Funamoto, Y. Ohta, T. Abe, K. Nagashima, Myelin
[162] G.M. McKhann, D.R. Cornblath, J.W. Grifn, T.W. Ho, C.Y. Li, Z. Jiang, H.S. Wu, gangliosides of human peripheral nervous system: an enrichment of GM1 in
G. Zhaori, Y. Liu, L.P. Jou, T.C. Liu, C.Y. Gao, J.Y. Mao, M.J. Blaser, B. Mishu, A.K. the motor nerve myelin isolated from cauda equina, J. Neurochem. 59
Asbury, Acute motor axonal neuropathy: a frequent cause of acute accid (1992) 18441849.
paralysis in China, Ann. Neurol. 33 (1993) 333342. [188] S.B. Oh, P.B. Tran, S.E. Gillard, R.W. Hurley, D.L. Hammond, R.J. Miller,
[163] W.G. McLean, The role of the axonal cytoskeleton in diabetic neuropathy, Chemokines and glycoprotein120 produce pain hypersensitivity by directly
Neurochem. Res. 22 (1997) 951956. exciting primary nociceptive neurons, J. Neurosci. 21 (2001) 50275035.
[164] R. Medori, L. Autilio-Gambetti, S. Monaco, P. Gambetti, Experimental [189] J.M. Osterloh, J. Yang, T.M. Rooney, A.N. Fox, R. Adalbert, E.H. Powell, A.E.
diabetic neuropathy: impairment of slow transport with changes in axon Sheehan, M.A. Avery, R. Hackett, M.A. Logan, J.M. MacDonald, J.S. Ziegenfuss,
cross- sectional area, Proc. Natl. Acad. Sci. 82 (1985) 77167720. S. Milde, Y.-J. Hou, C. Nathan, A. Ding, R.H. Brown Jr., L. Conforti, M. Coleman,
[165] G. Melli, S.C. Keswani, A. Fischer, W. Chen, A. Hke, Spatially distinct and M. Tessier-Lavigne, S. Zchner, M.R. Freeman, dSarm/Sarm1 is required for
functionally independent mechanisms of axonal degeneration in a model of activation of an injury-induced axon death pathway, Science 337 (2012)
HIV-associated sensory neuropathy, Brain 129 (2006) 13301338. 481484.
[166] C. Meregalli, A. Canta, V.A. Carozzi, A. Chiorazzi, N. Oggioni, A. Gilardini, C. [190] M. Palta, T. LeCaire, M. Sadek-Badawi, V. Herrera, K.K. Danielson, The
Ceresa, F. Avezza, L. Crippa, P. Marmiroli, G. Cavaletti, Bortezomib-induced trajectory of IGF-1 across age and duration of type 1 diabetes, Diabetes
painful neuropathy in rats: a behavioral, neurophysiological and Metab. Res. Rev. 30 (2014).
pathological study in rats, Eur. J. Pain 14 (2010) 343350. [191] C. Pekiner, N.A. Cullum, J.N. Highes, A.J. Hargreaves, J. Mahon, I.F. Casson,
[167] C. Meregalli, A. Chiorazzi, V. Carozzi, A. Canta, B. Sala, M. Colombo, N. W.G. McLean, Glycation of brain actin in experimental diabetes, J.
Oggioni, C. Ceresa, D. Foudah, F. La Russa, M. Miloso, G. Nicolini, P. Neurochem. 61 (1993) 436442.
Marmiroli, D.L.H. Bennett, G. Cavaletti, Evaluation of tubulin polymerization [192] V.H. Perry, M.C. Brown, J.W. Tsao, The effectiveness of the gene which slows
and chronic inhibition of protesome as citotoxicity mechanisms in the rate of Wallerian degeneration in C57BL/Ola mice declines with age, Eur.
bortezomib-induced peripheral neuropathy, Cell Cycle 13 (2014) 612621. J. Neurosci. 4 (1992) 10001002.
[168] D. Metodiewa, C. Koska, Reactive oxygen species and reactive nitrogen [193] V.H. Perry, E.R. Lunn, M.C. Brown, S. Cahusac, S. Gordon, Evidence that the
species: relevance to cyto(neuro) toxic events and neurologic disorders. An rate of Wallerian degeneration is controlled by a single autosomal dominant
overview, Neurotox. Res. 1 (1999) 197233. gene, Eur. J. Neurosci. 2 (1990) 408413.
[169] G. Meyer zu Horste, T.A. Miesbach, J.I. Muller, R. Fledrich, R.M. Stassart, B.C. [194] C.M. Peters, J.M. Jimenez-Andrade, B.M. Jonas, M.A. Sevcik, N.J. Koewler, J.R.
Kieseier, M.P. Coleman, M.W. Sereda, The WldS transgene reduces axon loss Ghilardi, G.Y. Wong, P.W. Mantyh, Intravenous paclitaxel in the rat induces
in a CharcotMarieTooth disease 1A rat model and nicotinamide delays a peripheral sensory neuropathy characterized by macrophage inltration
post-traumatic axonal degeneration, Neurobiol. Dis. 42 (2011) 18. and injury to sensory neurons and their supporting cells, Exp. Neurol. 203
[170] B.R. Miller, C. Press, R.W. Daniels, Y. Sasaki, J. Milbrandt, A. DiAntonio, A dual (2007) 4254.
leucine kinase-dependent axon self- destruction program promotes [195] C.M. Peters, J.M. Jimenez-Andrade, M.A. Kuskowski, J.R. Ghilardi, P.W.
Wallerian degeneration, Nat. Neurosci. 12 (2009) 387389. Mantyh, An evolving cellular pathology occurs in dorsal root ganglia,
[171] S.L. Mironov, M.V. Ivannikov, M. Johansson, [Ca2+ ]i signaling between peripheral nerve and spinal cord following intravenous administration of
mitochondria and endoplasmic reticulum in neurons is regulated by paclitaxel in the rat, Brain Res. 1168 (2007) 4659.
microtubules: from mitochondrial permeability transition pore to [196] C.R. Pierson, W. Zhang, Y. Murakawa, A.A.F. Sima, Insulin deciency rather
Ca2+ -induced Ca2+ release, J. Biol. Chem. 280 (2005) 715721. than hyperglycemia accounts for impaired neurotrophic responses and
[172] L. Mohiuddin, P. Fernyhough, D.R. Tomlinson, Reduced levels of mRNA nerve ber regeneration in type 1 diabetic neuropathy, J. Neuropathol. Exp.
encoding endoskeletal and growth-associated proteins in sensory ganglia in Neurol. 62 (2003) 260271.
experimental diabetes, Diabetes 44 (1995) 2530. [197] J.L. Podratz, A.M. Knight, L.E. Ta, N.P. Staff, J.M. Gass, K. Genelin, A. Schlattau,
[173] Z. Mokini, M.L. Marcovecchio, F. Chiarelli, Molecular pathology of oxidative L. Lathroum, A.J. Windebank, Cisplatin induced mitochondrial DNA damage
stress in diabetic angiopathy: role of mitochondrial and cellular pathways, in dorsal root ganglion neurons, Neurobiol. Dis. 41 (2011) 661668.
Diabetes Res. Clin. Pract. 87 (2010) 313321. [198] F.C. Polizzi, G. Andican, E. Cetin, S. Civelek, V. Yumuk, G. Burcak, Increased
[174] E.A. Mullokandov, W.A. Franklin, M. Brownlee, DNA damage by the glycation DNA-glycation in type 2 diabetic patients: the effect of thiamine and
products of glyceraldehyde 3-phosphate and lysine, Diabetologia 37 (1994) pyridoxine therapy, Exp. Clin. Endocr. Diab. 120 (2012) 329334.
145149. [199] C. Press, J. Milbrandt, Nmnat delays axonal degeneration caused by
[175] M.P. Murphy, How mitochondria produce reactive oxygen species, Biochem. mitochondrial and oxidative stress, J. Neurosci. 28 (2008) 48614871.
J. 417 (2009) 113. [200] H. Prss, J.M. Schwab, C. Derst, A. Grtzen, R.W. Veh, Neurofascin as target of
[176] S. Nasu, S. Misawa, C. Nakaseko, K. Shibuya, S. Isose, Y. Sekiguchi, S. autoantibodies in Guillain-Barr syndrome, Brain 134 (2011) 12.
Mitsuma, S. Ohmori, Y. Iwai, M. Beppu, N. Shimizu, C. Ohwada, Y. Takeda, Y. [201] M. Quartu, V.A. Carozzi, S.G. Dorsey, M.P. Serra, L. Poddighe, C. Picci, M. Boi,
Fujimaki, S. Kuwabara, Bortezomib-induced neuropathy: axonal membrane T. Melis, M. Del Fiacco, C. Meregalli, A. Chiorazzi, C.L. Renn, G. Cavaletti, P.
depolarization precedes development of neuropathy, Clin. Neurophysiol. Marmiroli, Bortezomib treatment produces nocifensive behavior and
125 (2014) 381387. changes in the expression of TRPV1, CGRP, and substance P in the rat DRG,
[177] A. Naudi, M. Jove, V. Ayala, A. Cassanye, J. Serrano, H. Gonzalo, J. Boada, J. spinal cord, and sciatic nerve, Biomed. Res. Int. 2014 (2014).
Prat, M. Portero-Otin, R. Pamplona, Cellular dysfunction in diabetes as [202] B.V. Ramana, V.V. Kumar, P.N.R. Krishna, C.S. Kumar, P.U.M. Reddy, T.N. Raju,
maladaptive response to mitochondrial oxidative stress, Exp. Diabetes Res. Effect of quercetin on galactose-induced hyperglycaemic oxidative stress in
2012 (2012) 16. hepatic and neuronal tissues of Wistar rats, Acta Diabetol. 43 (2006)
[178] M. Neeper, A.M. Schmidt, J. Brett, S.D. Yan, F. Wang, Y.-C.E. Pan, K. Elliston, D. 135141.
Stern, A. Shaw, Cloning and expression of a cell surface receptor for [203] K.V. Ramana, B. Friedrich, R. Tammali, M.B. West, A. Bhatnagar, S.K.
advanced glycosylation end products of proteins, J. Biol. Chem. 267 (1992) Srivastava, Requirement of aldose reductase for the hyperglycemic
1499815004. activation of protein kinase C and formation of diacylglycerol in vascular
[179] A. Ngre-Salvayre, C. Hirtz, G. Carrera, R. Cazenave, M. Troly, R. Salvayre, L. smooth muscle cells, Diabetes 54 (2005) 818829.
Pnicaud, L. Casteilla, A role for uncoupling protein-2 as a regulator of [204] C.L. Renn, C.C. Leitch, S. Lessans, P. Rhee, W.C. McGuire, B.A. Smith, R.J.
mitochondrial hydrogen peroxide generation, FASEB J. 11 (1997) 809815. Traub, S.G. Dorsey, Brain-derived neurotrophic factor modulates
[180] R. Nemni, L. Sanvito, A. Quattrini, G. Santuccio, M. Camerlingo, N. Canal, antiretroviral-induced mechanical allodynia in the mouse, J. Neurosci. Res.
Peripheral neuropathy in hepatitis C virus infection with and without 89 (2011) 15511565.
cryoglobulinaemia, J. Neurol. Neurosurg. Psychiatry 74 (2003) 12671271. [205] D.J. Rodi, R.W. Janes, H.J. Sanganee, R.A. Holton, B.A. Wallace, L. Makowski,
[181] D.G. Nicholls, R.M. Locke, Thermogenic mechanisms in brown fat, Physiol. Screening of a library of phage-displayed peptides identies human bcl-2 as
Rev. 64 (1984) 164. a taxol-binding partner, J. Mol. Biol. 285 (1999) 197203.
[182] K. Nishida, S. Kuchiiwa, S. Oiso, T. Futagawa, S. Masuda, Y. Takeda, K. [206] B. Romani, S. Engelbrecht, Human immunodeciency virus type 1 Vpr:
Yamada, Up-regulation of matrix metalloproteinase-3 in the dorsal root functions and molecular interactions, J. Gen. Virol. 90 (2009) 17951805.

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048
G Model
NSL-31080; No. of Pages 18 ARTICLE IN PRESS
C.R. Cashman, A. Hke / Neuroscience Letters xxx (2015) xxxxxx 17

[207] N.R. Rosenberg, P. Portegies, M. de Visser, M. Vermeulen, Diagnostic [232] A.W. Stitt, C. He, H. Vlassara, Characterization of the advanced glycation
investigation of patients with chronic polyneuropathy: evalualtion of a end-product receptor complex in human vascular endothelial cells,
clinical guideline, J. Neurol. Neurosurg. Psychiatry 71 (2001) 205209. Biochem. Biophys. Res. Commun. 256 (1999) 549556.
[208] S. Rousset, M.-C. Alves-Guerra, J. Mozo, B. Miroux, A.-M. Cassard-Doulcier, F. [233] P.K. Stys, S.G. Waxman, B.R. Ransom, Ionic mechanisms of anoxic injury in
Bouillaud, D. Ricquier, The biology of mitochondrial uncoupling proteins, mammalian CNS white matter: role of Na+ channels and Na+-Ca+ exchanger,
Diabetes 53 (2004) S130S135. J. Neurosci. 12 (1992) 430439.
[209] G. Rudofsky, A. Schroedter, A. Schlotterer, O.E. Voronko, M. Schlimme, J. [234] K. Sugimoto, Y. Nishizawa, S. Horiuchi, S. Yagihashi, Localization in human
Tafel, B.H. Isermann, P.M. Humpert, M. Morcos, A. Bierhaus, P.P. Nawroth, A. diabetic peripheral nerve of N-e-carboxymethyllysine-protein adducts, an
Hamann, Functional polymorphisms of UCP2 and UCP3 are associated with advanced glycation endproduct, Diabetologia 40 (1997) 13801387.
a reduced prevalence of diabetic neuropathy in patients with type 1 [235] K. Susuki, M.N. Rasband, K. Tohyama, K. Koibuchi, S. Okamoto, K. Funakoshi,
diabetes, Diabetes Care 29 (2006) 8994. K. Hirata, H. Baba, N. Yuki, Anti-GM1 antibodies cause complement-
[210] J.W. Russell, A.J. Windebank, M.A. McNiven, D.J. Brat, W.S. Brimijoin, Effect mediated disruption of sodium channel clusters in peripheral motor nerve
of cisplatin and ACTH49 on neural transport in cisplatin induced bers, J. Neurosci. 27 (2007) 39563967.
neurotoxicity, Brain Res. 676 (1995) 258267. [236] K. Susuki, N. Yuki, D.P. Schafer, K. Hirata, G. Zhang, K. Funakoshi, M.N.
[211] Z. Sahenk, R. Barohn, P. New, J.R. Mendell, Taxol neuropathy: Rasband, Dysfunction of nodes of Ranvier: a mechanism for anti-ganglioside
electrodiagnostic and sural nerve biopsy ndings, Arch. Neurol. 51 (1994) antibody-mediated neuropathies, Exp. Neurol. 233 (2012) 534542.
726729. [237] L. Svennerholm, K. Bostrm, P. Fredman, B. Jungbjer, A. Lekman, J.-E.
[212] M. Samsam, W. Mi, C. Wessig, J. Zielasek, K.V. Toyka, M.P. Coleman, R. Mnsson, B.-M. Rynmark, Gangliosides and allied glycosphingolipids in
Martini, The WldS mutation delays robust loss of motor and sensory axons human peripheral nerve and spinal cord, Biochim. Biophys. Acta 1214
in a genetic model for myelin-related axonopathy, J. Neurosci. 23 (2003) (1994) 115123.
28332839. [238] L.E. Ta, L. Espeset, J. Podratz, A.J. Windebank, Neurotoxicity of oxaliplatin
[213] L. Santoro, F. Manganelli, C. Briani, F. Giannini, L. Benedetti, E. Vitelli, A. and cisplatin for dorsal root ganglion neurons correlates with
Mazzeo, E. Beghi, H.P.N.S. Group, Prevalence and characteristics of platinum-DNA binding, Neurotoxicology 27 (2006) 9921002.
peripheral neuropathy in hepatitis C virus population, J. Neurol. Neurosurg. [239] T. Takigawa, H. Yasuda, R. Kikkawa, Y. Shigeta, T. Saida, H. Kitasato,
Psychiatry 77 (2006) 626629. Antibodies against GM1 ganglioside affect K+ and Na+ currents in isolated
[214] Y. Sasaki, J. Milbrandt, Axonal degeneration is blocked by nicotinamide rat myelinated nerve bers, Ann. Neurol. 37 (1995) 436442.
mononucleotide adenylyltransferase (Nmnat) protein transduction into [240] S. Tanaka, G. Avigad, B. Brodsky, E.F. Eikenberry, Glycation induces
transected axons, J. Biol. Chem. 285 (2010) 4121141215. expansion of the molecular packing of collagen, J. Mol. Biol. 203 (1988)
[215] Y. Sasaki, B.P.S. Vohra, R.H. Baloh, J. Milbrandt, Transgenic mice expressing 495505.
the Nmnat1 protein manifest robust delay in axonal degeneration in vivo, J. [241] J.I. Tembl, J.M. Ferrer, M.T. Sevilla, A. Lago, F. Mayordomo, J.J. Vilchez,
Neurosci. 29 (2009) 65266534. Neurological complications associated with hepatitis C virus infection,
[216] Y. Sasaki, B.P.S. Vohra, F.E. Lund, J. Milbrandt, Nicotinamide mononucleotide Neurology 53 (1999) 861864.
adenylyl transferase-mediated axonal protection requires enzymatic [242] S. Tewari, J.M. Santos, R.A. Kowluru, Damaged mitochondrial DNA
activity but not increased levels of neuronal nicotinamide adenine replication system and the development of diabetic retinopathy, Antioxid.
dinucleotide, J. Neurosci. 29 (2009) 55255535. Redox. Signaling 17 (2012) 492504.
[217] A.M. Schmidt, M. Vianna, M. Gerlach, J. Brett, J. Ryan, J. Kao, C. Esposito, H. [243] C. Theiss, K. Meller, Taxol impairs anterograde axonal transport of
Hegarty, W. Hurley, M. Clauss, F. Wang, Y.-C.E. Pan, T.C. Tsang, D. Stern, microinjected horseradish peroxidase in dorsal root ganglia neurons in
Isolation and characterization of two binding proteins for advanced vitro, Cell Tissue Res. 299 (2000) 213224.
glycosylation end products from bovine lung which are present on the [244] F.P. Thomas, R.E. Lovelace, X.-S. Ding, S.A. Sadiq, G.W. Petty, W.H. Sherman,
endothelial cell surface, J. Biol. Chem. 267 (1992) 1498714997. N. Latov, A.P. Hays, Vasculitic neuropathy in a patient with cryglobulinemia
[218] A. Scuteri, A. Galimberti, D. Maggioni, M. Ravasi, S. Pasini, G. Nicolini, M. and anti-MAG IgM monoclonal gammopathy, Muscle Nerve 15 (1992)
Bossi, M. Miloso, G. Cavaletti, G. Tredici, Role of MAPKs in platinum-induced 891898.
neuronal apoptosis, Neurotoxicology 30 (2009) 312319. [245] P.K. Thomas, R.G. Lascelles, Schwann-cell abnormalities in diabetic
[219] H. Sekido, T. Suzuki, T. Jomori, M. Takeuchi, C. Yabe-Nishimura, S. Yagihashi, neuropathy, Lancet 285 (1965) 13551357.
Reduced cell replication and induction of apoptosis by advanced glycation [246] P.J. Thornalley, Pharmacology of methylglyoxal: formation, modication of
end products in rat Schwann cells, Biochem. Biophys. Res. Commun. 320 proteins and nucleic acids, and enzymatic detoxication a role in
(2004) 241248. pathogenesis and antiproliferative chemotherapy, Gen. Pharmacol. 27
[220] J.E. Shaw, R.A. Sicree, P.Z. Zimmet, Global estimates of the prevalence of (1996) 565573.
diabetes for 2010 and 2030, Diabetes Res. Clin. Pract. 87 (2010) 414. [247] M. Tiedge, S. Lortz, J. Drinkgern, S. Lenzen, Relation between antioxidant
[221] H. Shen, K.L. Hyrc, M.P. Goldberg, Maintaining energy homeostasis is an enzyme gene expression and antioxidant defense status of
essential component of WldS -mediated axon protection, Neurobiol. Dis. 59 insulin-producing cells, Diabetes 46 (1997) 17331742.
(2013) 6979. [248] C. Tofthagen, Patient perceptions associated with chemotherapy-induced
[222] J. Shepard, N. Waugh, P. Hewitson, Combination therapy (interferon alfa and peripheral neuropathy, Clin. J. Oncol. Nurs. 14 (2010) E22E28.
ribavirin) in the treatment of chronic hepatitis C: a rapid and systemic [249] J.A. Tracy, P.J.B. Dyck, The spectrum of diabetic neuropathies, Phys. Med.
review, Health Technol. Assess. 4 (2000) 167. Rehabil. Clin. N. Am. 19 (2008) 126.
[223] J.E. Shin, B.R. Miller, E. Babetto, Y. Cho, Y. Sasaki, S. Qayum, E.V. Russler, V. [250] H. Tsujino, E. Kondo, T. Fukuoka, Y. Dai, A. Tokunaga, K. Miki, K. Yonenobu, T.
Cavalli, J. Milbrandt, A. DiAntonio, SCG10 is a JNK target in the axonal Ochi, K. Noguchi, Activating transcription factor 3 (ATF3) induction by
degeneration pathway, Proc. Natl. Acad. Sci. 109 (2012) E3696E3705. axotomy in sensory and motoneurons: a novel neuronal marker of nerve
[224] Y.K. Shin, S.Y. Jang, H.K. Lee, J. Jung, D.J. Suh, S.-Y. Seo, H.T. Park, Pathological injury, Mol. Cell Neurosci. 15 (2000) 170182.
adaptive responses of Schwann cells to endoplasmic reticulum stress in [251] W.R. Tyor, S.L. Wesselingh, J.W. Grifn, J.C. McArthur, D.E. Grifn, Unifying
bortezomib- induced peripheral neuropathy, Glia 58 (2010) hypothesis for the pathogenesis of HIV-associated dementia complex
19611976. vacuolar myelopathy and sensory neuropathy, J. Acquir. Immune Dec.
[225] V.I. Shubayev, M. Angert, J. Dolkas, W.M. Campana, K. Palenscar, R.R. Myers, Syndr. Hum. Retrovirol. 9 (1995) 379388.
TNF-induced MMP-9 promotes macrophage recruitment into injured [252] G. Varbiro, B. Veres, F. Gallyas Jr., B. Sumegi, Direct effect of Taxol on free
peripheral nerve, Mol. Cell Neurosci. 31 (2006) 407415. radical formation and mitochondrial permeability transition, Free Radical
[226] A.A.F. Sima, W. Zhang, K. Sugimoto, D. Henry, Z. Li, J. Wahren, G. Grunberger, Biol. Med. 31 (2001) 548558.
C-peptide prevents and improves chronic type I diabetic polyneuropathy in [253] A.J. Vidal-Puig, D. Grujic, C.-Y. Zhang, T. Hagen, O. Boss, Y. Ido, A. Szczepanik,
the BB/Wor rat, Diabetologia 44 (2001) 889897. J. Wade, V. Mootha, R. Cortright, D.M. Muoio, B.B. Lowell, Energy metabolism
[227] A.A.F. Sima, W. Zhang, G. Xu, K. Sugimoto, D. Guberski, M.A. Yorek, A in uncoupling protein 3 gene knockout mice, J. Biol. Chem. 275 (2000)
comparison of diabetic polyneuropathy in type II diabetic BBZDR/Wor rats 1625816266.
and in type I diabetic BB/Wor rats, Diabetologia 43 (2000) [254] A.M. Vincent, B. Calabek, L. Roberts, E.L. Feldman, Biology of diabetic
786793. neuropathy, in: G. Said, C. Krarup (Eds.), Handbook of Clinical Neurology,
[228] V.P. Skulachev, Uncoupling: new approaches to an old problem of vol. 115, Elsevier, 2013, pp. 591606.
bioenergetics, Biochim. Biophys. Acta 1363 (1998) 100124. [255] A.M. Vincent, J.L. Edwards, L.L. McLean, Y. Hong, F. Cerri, I. Lopez, A.
[229] K. Smyth, J.S. Affandi, J.C. McArthur, C. Bowtell-Harris, A.M. Mijch, K. Quattrini, E.L. Feldman, Mitochondrial biogenesis and ssion in axons in cell
Watson, K. Costello, I.J. Woolley, P. Price, S.L. Wesselingh, C.L. Cherry, culture and animal models of diabetic neuropathy, Acta Neuropathol. 120
Prevalence of and risk factors for HIV-associated neuropathy in Melbourne, (2010) 477489.
Australia 19932006, HIV Med. 8 (2007) 367373. [256] A.M. Vincent, E.L. Feldman, Control of cell survival by IGF signaling
[230] K.C. Sourris, J.M. Forbes, Interactions between advanced glycation pathways, Growth Horm. IGF Res. 12 (2002) 193197.
end-products (AGE) and their receptors in the development and progression [257] A.M. Vincent, J.A. Olzmann, M. Brownlee, W.I. Sivitz, J.W. Russell,
of diabetic nephropathy are these receptors valid therapeutic targets, Curr. Uncoupling proteins prevent glucose- induced neuronal oxidative stress and
Drug Targets 10 (2009) 4250. programmed cell death, Diabetes 53 (2004) 726734.
[231] J.A. Sprowl, G. Ciarimboli, C.S. Lancaster, H. Giovinazzo, A.A. Gibson, G. Du, [258] A.M. Vincent, J.W. Russell, P. Low, E.L. Feldman, Oxidative stress in the
L.J. Janke, G. Cavaletti, A.F. Shields, A. Sparreboom, Oxaliplatin-induced pathogenesis of diabetic neuropathy, Endocr. Rev. 25 (2004) 612628.
neurotoxicity is dependent on the organic cation transporter OCT2, Proc. [259] H. Vlassara, The AGE-receptor in the pathogenesis of diabetic complications,
Natl. Acad. Sci. 110 (2013) 1119911204. Diabetes Metab. Res. Rev. 17 (2001) 436443.

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048
G Model
NSL-31080; No. of Pages 18 ARTICLE IN PRESS
18 C.R. Cashman, A. Hke / Neuroscience Letters xxx (2015) xxxxxx

[260] H. Vlassara, M. Brownlee, A. Cerami, High-afnity-receptor-mediated promoter polymorphism 866 G/A affects peripheral nerve dysfunction in
uptake and degradation of glucose-modied proteins: a potential Japanese type 2 diabetic patients, Diabetes Care 29 (2006) 888894.
mechanism for removal of senescent macromolecules, Proc. Natl. Acad. Sci. [285] S.D. Yan, A.M. Schmidt, G.M. Anderson, J. Zhang, J. Brett, Y.S. Zou, D. Pinsky,
82 (1985) 55885592. D. Stern, Enhanced cellular oxidant stress by the interaction of advanced
[261] H. Vlassara, M. Brownlee, A. Cerami, Nonenzymatic glycosylation of glycation end products with their receptors/binding proteins, J. Biol. Chem.
peripheral nerve protein in diabetes mellitus, Proc. Natl. Acad. Sci. 78 (1981) 269 (1994)
51905192. 98899897.
[262] H. Vlassara, M. Brownlee, A. Cerami, Novel macrophage receptor for [286] J. Yang, R.M. Weimer, D. Kallop, O. Olsen, Z. Wu, N. Renier, K. Uryu, M.
glucose-modied proteins is distinct from previously described scavenger Tessier-Lavigne, Regulation of axon degeneration after injury and in
receptors, J. Exp. Med. 164 (1986) 13011309. development by the endogenous calpain inhibitor calpastatin, Neuron 80
[263] H. Vlassara, M. Brownlee, K.R. Manogue, C.A. Dinarello, A. Pasagian, (2013) 11751189.
Cachectin/TNF and IL-1 induced by glucose-modied proteins: role in [287] X. Yang, K. Su, M.D. Roos, Q. Chang, A.J. Paterson, J.E. Kudlow, O-linkage of
normal tissue remodeling, Science 240 (1988) 15461548. N-acetylglucosamine to Sp1 activation domain inhibits its transcriptional
[264] H. Vlassara, Y.M. Li, F. Imani, D. Wojciechowicz, Z. Yang, F.-T. Liu, A. Cerami, capability, Proc. Natl. Acad. Sci. 98 (2001) 66116616.
Identication of galectin-3 as a high-afnity binding protein for advanced [288] Z. Yang, Z. Makita, Y. Horii, S. Brunelle, A. Cerami, P. Sehajpal, M.
glycation end products (AGE): a new member of the AGE-receptor complex, Suthanthiran, H. Vlassara, Two novel rat liver membrane proteins that bind
Mol. Med. 1 (1995) 634646. advanced glycosylation endproducts: relationship to macrophage receptor
[265] J. Wahren, K. Ekberg, J. Johansson, M. Henriksson, A. Pramanik, B.-L. for glucose-modied proteins, J. Exp. Med. 174 (1991) 515524.
Johansson, R. Rigler, H. Jrnvall, Role of C-peptide in human physiology, Am. [289] C.-H. Yeh, L. Sturgis, J. Haidacher, X.-N. Zhang, S.J. Sherwood, R.J. Bjercke, O.
J. Physiol. Endocrinol. Metab. 278 (2000) E759E768. Juhasz, M.T. Crow, R.G. Tilton, L. Denner, Requirement for p38 and p44/p42
[266] C.-H. Wang, B. Wang, R.-L. Wendu, H.-e. Bi, G.-F. Cao, C. Ji, Q. Jiang, J. Yao, mitogen-activated protein kinases in RAGE-mediated nuclear factor-kB
Protective role of Wallerian degeneration slow (Wlds ) gene against retinal transcriptional activation and cytokine secretion, Diabetes 50 (2001)
ganglion cell body damage in a Wallerian degeneration model, Exp. Ther. 14951504.
Med. 5 (2013) 621625. [290] M.A. Yorek, J.A. Dunlap, B.H. Ginsberg, myo-Inositol metabolism in 41A3
[267] J. Wang, Q. Zhai, Y. Chen, E. Lin, W. Gu, M.W. McBurney, Z. He, A local neuroblastoma cells: effects of high glucose and sorbitol levels, J.
mechanism mediates NAD-dependent protection of axon degeneration, J. Neurochem. 48 (1987) 5361.
Cell Biol. 170 (2005) 349355. [291] R.J. Youle, M. Karbowski, Mitochondrial ssion in apoptosis, Nat. Rev. Mol.
[268] M.-S. Wang, Y. Wu, D.G. Culver, J.D. Glass, The gene for slow Wallerian Cell Biol. 6 (2005) 657663.
degeneration (WldS ) is also protective against vincristine neuropathy, [292] S. Yui, T. Sasaki, N. Araki, S. Horiuchi, M. Yamazaki, Induction of macrophage
Neurobiol. Dis. 8 (2001) 155161. growth by advanced glycation end products of the Maillard reaction, J.
[269] M.S. Wang, A.A. Davis, D.G. Culver, J.D. Glass, WldS mice are resistant to Immunol. 152 (1994) 19431949.
paclitaxel (Taxol) neuropathy, Ann. Neurol. 52 (2002) 442447. [293] N. Yuki, H.-P. Hartung, GuillainBarr syndrome, N. Eng. J. Med. 366 (2012)
[270] M.S. Wang, A.A. Davis, D.G. Culver, Q. Wang, J.C. Powers, J.D. Glass, Calpain 22942304.
inhibition protects against Taxol-induced sensory neuropathy, Brain 127 [294] N. Yuki, K. Susuki, M. Koga, Y. Nishimoto, M. Odaka, K. Hirata, K. Taguchi, T.
(2004) 671679. Miyatake, K. Furukawa, T. Kobata, M. Yamada, Carbohydrate mimicry
[271] X.-M. Wang, T.J. Lehky, J.M. Brell, S.G. Dorsey, Discovering cytokines as between human ganglioside GM1 and Campylobacter jejuni
targets for chemotherapy-induced painful peripheral neuropathy, Cytokine lipooligosaccharide causes GuillainBarr syndrome, Proc. Natl. Acad. Sci.
59 (2012) 39. 101 (2004) 1140411409.
[272] R.A. Warwick, M. Hanani, The contribution of satellite glial cells to [295] N. Yuki, M. Yamada, M. Koga, M. Odaka, K. Susuki, Y. Tagawa, S. Ueda, T.
chemotherapy-induced neuropathic pain, Eur. J. Pain 17 (2013) 571580. Kasama, A. Ohnishi, S. Hayashi, H. Takahashi, M. Kamijo, K. Hirata, Animal
[273] J.-L. Wautier, M.-P. Wautier, A.-M. Schmidt, G.M. Anderson, O. Hori, C. model of axonal GuillainBarr syndrome induced by sensitization with
Zoukourian, L. Capron, O. Chappey, S.-D. Yan, J. Brett, P.-J. Guillausseau, D. GM1 ganglioside, Ann. Neurol. 49 (2001) 712720.
Stern, Advanced glycation end products (AGEs) on the surface of diabetic [296] N. Yuki, H. Yoshino, S. Sato, T. Miyatake, Acute axonal polyneuropathy
erythrocytes bind to the vessel wall via a specic receptor inducing oxidant associated with anti-GM1 antibodies following Campylobacter enteritis,
stress in the vasculature: a link between surface-associated AGEs and Neurology 40 (1990) 19001902.
diabetic complications, Proc. Natl. Acad. Sci. 91 (1994) 77427746. [297] Q. Zhai, J. Wang, A. Kim, Q.-H. Liu, R. Watts, E. Hoopfer, T. Mitchison, L. Luo,
[274] S.K. Williams, N.L. Howarth, J.J. Devenny, M.W. Bitensky, Structural and Z. He, Involvement of the ubiquitin-proteasome system in the early stages of
functional consequences of increased tubulin glycosylation in diabetes Wallerian degeneration, Neuron 39 (2003) 217225.
mellitus, Proc. Natl. Acad. Sci. 79 (1982) 65466550. [298] R.G. Zhai, Y. Cao, P.R. Hiesinger, Y. Zhou, S.Q. Mehta, K.L. Schulze, P.
[275] J.B. Winer, An update on GuillainBarr syndrome, Autoimmune Dis. 2014 Verstreken, H.J. Bellen, Drosophila NMNAT maintains neural integrity
(2014) 16. independent of its NAD synthesis activity, PLoS Biol. 4 (2006) 23362348.
[276] H. Witte, D. Neukirchen, F. Bradke, Microtubule stabilization specices [299] R.G. Zhai, F. Zhang, P.R. Hiesinger, Y. Cao, C.M. Haueter, H.J. Bellen, NAD
initial neuronal polarization, J. Cell Biol. 180 (2008) 619632. synthase NMNAT acts as a chaperone to protect against neurodegeneration,
[277] M.Y. Wong, C. Zhou, D. Shakiryanova, T.E. Lloyd, D.L. Deitcher, E.S. Levitan, Nature 452 (2008) 887892.
Neuropeptide delivery to synapses by long-range vesicle circulation and [300] Z. Zhang, C.W. Liew, D.E. Handy, Y. Zhang, J.E. Leopold, J. Hu, L. Guo, R.N.
sporadic capture, Cell 148 (2012) 10291038. Kulkarni, J. Loscalzo, R.C. Stanton, High glucose inhibits
[278] J. Wu, F. Zhang, M. Yan, D. Wu, Q. Yu, Y. Zhang, B. Zhou, M.W. McBurney, Q. glucose-6-phosphate dehydrogenase, leading to increased oxidative stress
Zhai, WldS enhances insulin transcription and secretion via a and -cell apoptosis, FASEB J. 24 (2010) 14971505.
SIRT1-dependent pathway and improves glucose homeostasis, Diabetes 60 [301] H. Zheng, W.H. Xiao, G.J. Bennett, Functional decits in peripheral nerve
(2011) 31973207. mitochondria in rats with paclitaxel- and oxaliplatin-evoked peripheral
[279] W.H. Xiao, H. Zheng, G.J. Bennett, Characterization of oxaliplatin-induced neuropathy, Exp. Neurol. 232 (2011) 154161.
chronic painful peripheral neuropathy in the rat and comparison with [302] H. Zheng, W.H. Xiao, G.J. Bennett, Mitotoxicity and bortezomib-induced
neuropathy induced by paclitaxel, Neuroscience 203 (2012) 194196. chronic painful peripheral neuropathy, Exp. Neurol. 238 (2012) 225234.
[280] W.H. Xiao, H. Zheng, F.Y. Zheng, R. Nuydens, T.F. Meert, G.J. Bennett, [303] X. Zheng, H. Ouyang, S. Liu, M. Mata, D.J. Fink, S. Hao, TNF is involved in
Mitochondrial abnormality in sensory but not motor axons in neuropathic pain induced by nucleoside reverse transcriptase inhibitor in
paclitaxel-evoked painful peripheral neuropathy in the rat, Neuroscience rats, Brain Behav. Immun. 25 (2011) 16681676.
199 (2011) 461469. [304] S.S. Zhu, Y. Ren, M. Zhang, J.Q. Cao, Q. Yang, X.Y. Li, H. Bai, L. Jiang, Q. Jiang,
[281] X. Xiong, Y. Hao, K. Sun, J. Li, X. Li, B. Mishra, P. Soppina, C. Wu, R.I. Hume, Z.G. He, Q. Chen, WldS protects against peripheral neuropathy and
C.A. Collins The Highwire ubiquitin ligase promotes axonal degeneration by retinopathy in an experimental model of diabetes in mice, Diabetologia 54
tuning levels of Nmnat protein, PLoS Biol., 10, (2012), e1001440. (2011) 24402450.
[282] C. Xu, B. Bailly-Maitre, J.C. Reed, Endoplastmic reticulum stress: cell life and [305] Y. Zhu, J.M. Antony, J.A. Martinez, D.M. Glerum, V. Brussee, A. Hoke, D.
death decisions, J. Clin. Invest. 115 (2005) 26562664. Zochodne, C. Power, Didanosine causes sensory neuropathy in an HIV/AIDS
[283] N. Yahata, S. Yuasa, T. Araki, Nicotinamide mononucleotide animal model: impaired mitochondrial and neurotrophic factor gene
adenylyltransferase expression in mitochondrial matrix delays Wallerian expression, Brain 130 (2007) 20112023.
degeneration, J. Neurosci. 29 (2009) 62766284. [306] D.W. Zochodne, D. Levy, Nitric oxide in damage, disease and repair of the
[284] H. Yamasaki, H. Sasaki, K. Ogawa, T. Shono, S. Tamura, A. Doi, M. Sasahara, H. peripheral nervous system, Cell. Mol. Biol. (Noisy-le-grand) 51 (2005)
Kawashima, T. Nakao, H. Furuta, M. Nishi, K. Nanjo, Uncoupling protein 2 255267.

Please cite this article in press as: C.R. Cashman, A. Hke, Mechanisms of distal axonal degeneration in peripheral neuropathies, Neurosci.
Lett. (2015), http://dx.doi.org/10.1016/j.neulet.2015.01.048

Potrebbero piacerti anche