Skip to content

Neuropathy

Neuropathy refers to any condition resulting from damage or disease affecting nerves, primarily those outside the brain and spinal cord, collectively known as the peripheral nervous system. This damage can disrupt the normal communication between the brain and other parts of the body, leading to a wide range of symptoms. Neuropathy can manifest in various forms, including sensory neuropathy (affecting sensation), motor neuropathy (affecting muscle movement), or autonomic neuropathy (affecting involuntary bodily functions). It can be caused by diverse factors, including physical injury, infections, exposure to toxins, systemic diseases such as diabetes, and certain medications, including D-drug-containing regimens and microtubule-targeting agents.[1]

The biological underpinnings of neuropathy involve damage to nerve cells (neurons), their axons (the long projections that transmit signals), or the myelin sheath that insulates them. Genetic factors play a significant role in an individual’s susceptibility to neuropathy, both inherited forms and those induced by external factors. Research has identified various genes associated with different types of neuropathy. For example, polymorphisms in genes such asIL2RA, ZNF648, RSPO4, and ANGPT4have been implicated in drug-induced peripheral neuropathy in specific patient populations.[1] Other genes are linked to nerve structure and function. FGD4 is involved in peripheral nerve myelination, and mutations in this gene are associated with autosomal recessive Charcot-Marie-Tooth Type 4H, a group of inherited peripheral nerve disorders.[1] Genes like KIF1B and KIF1A encode proteins crucial for axonal transport, with KIF1Amutations linked to spastic paraplegia and hereditary sensory neuropathy.[1] Mutations in LITAFhave been associated with Charcot-Marie-Tooth disease type 1C, whileNEFL, which encodes the neurofilament light polypeptide, is associated with Charcot-Marie-Tooth disease types 2E and 1F.[1] Furthermore, the gene S1PR1has been validated in genome-wide meta-analyses for its role in microtubule-targeting agent-induced sensory peripheral neuropathy.[2] Genes like CX3CL1 and CALUare also associated with expression and/or splicing quantitative trait loci, suggesting their relevance in regulating gene expression related to neuropathy.[2] Additionally, METTL4 may influence neural transmission through methyltransferase activity, potentially affecting sensory disturbances.[3]

The clinical presentation of neuropathy is highly variable, depending on the type and location of the affected nerves. Symptoms can range from mild numbness and tingling to severe chronic pain, muscle weakness, and even paralysis. Autonomic neuropathy can lead to issues with digestion, blood pressure regulation, and heart rate. Diagnosis typically involves a comprehensive neurological examination, nerve conduction studies, electromyography, and sometimes nerve biopsy. Treatment strategies focus on managing symptoms, addressing the underlying cause (if identifiable), and preventing further nerve damage. Understanding the genetic predispositions through studies like genome-wide association studies (GWAS) is crucial for identifying individuals at higher risk, particularly for drug-induced neuropathies, and for developing personalized treatment approaches.[1] Comprehensive phenotyping, which combines patient-reported and physician-reported outcomes, is essential for obtaining a complete clinical picture.[2]

Neuropathy carries significant social and economic implications due to its potential to severely impact an individual’s quality of life. Chronic pain, disability, and reduced mobility can hinder daily activities, employment, and social engagement. The economic burden includes healthcare costs, long-term medication, rehabilitation, and lost productivity. The prevalence of conditions that contribute to neuropathy, such as diabetes, underscores its public health importance. Furthermore, the occurrence of neuropathy as an adverse effect of life-saving medical treatments, such as those used in AIDS Clinical Trials Group protocols, highlights the critical need to identify genetic factors that can predict and mitigate these debilitating side effects, ensuring better patient outcomes and public health management.[1]

Methodological and Statistical Constraints

Section titled “Methodological and Statistical Constraints”

The primary genetic studies on peripheral neuropathy often operate with relatively small sample sizes, which inherently limits the statistical power to detect genuine genetic associations, especially for variants exerting modest effects.[1] Such constrained cohorts are less likely to yield robust genome-wide significant findings and can contribute to an overestimation of effect sizes for detected loci, making the interpretation of their true biological impact challenging.[4]This limitation frequently results in a higher susceptibility to false positives and a diminished ability to identify the full spectrum of genetic contributions to complex conditions like neuropathy.

A critical limitation across many genetic studies is the frequent absence of independent replication for initial suggestive findings.[5] Many genetic associations, particularly those involving variants with low allele frequencies, may fail to be validated in subsequent cohorts, suggesting that initial signals could be spurious or population-specific.[6] Furthermore, studies that rely on referral-based cohorts or clinical diagnoses rather than uniformly standardized research assessments can introduce sampling bias, affecting the generalizability and robustness of observed genetic associations.[7] Therefore, a lack of replication underscores the need for larger, collaborative efforts to confirm genetic associations and ensure their reliability across diverse populations.

Phenotypic Heterogeneity and Generalizability

Section titled “Phenotypic Heterogeneity and Generalizability”

The precise phenotyping of peripheral neuropathy presents a significant challenge, with current grading scales, such as the NCI-CTCAE, potentially underestimating symptom progression and exhibiting inconsistencies in interpretation.[2] This variability in phenotype definition and across different studies or clinical settings can introduce considerable noise, obscuring true genetic signals and making it difficult to combine data effectively in meta-analyses.[5] The use of varied assessment tools, even if validated, can also contribute to this heterogeneity, highlighting the need for more standardized and comprehensive phenotyping methods, ideally combining patient-reported and physician-reported outcomes, to capture the full spectrum of the condition.[2]While some studies incorporate patients from multiple ancestral groups, the specific demographic composition of cohorts, such as those within AIDS Clinical Trials Groups, may not fully represent the global diversity of individuals affected by neuropathy.[1] This can limit the generalizability of findings, as genetic risk factors and their effect sizes can vary significantly across different ancestral populations due to differences in allele frequencies, linkage disequilibrium patterns, and environmental exposures.[8] Consequently, findings may not be universally applicable, necessitating further research in more diverse and representative populations to ensure broad clinical relevance and to identify ancestry-specific genetic influences.

Unexplored Interactions and Biological Mechanisms

Section titled “Unexplored Interactions and Biological Mechanisms”

Current genetic studies often do not extensively explore complex gene-gene or gene-environment interactions, which are critical for fully elucidating the multifactorial etiology of peripheral neuropathy.[7]Unmeasured common factors, such as lifestyle, comorbidities, or socioeconomic status, can act as significant confounders or mediators, potentially introducing collider bias if not adequately accounted for in statistical models.[9]A comprehensive understanding of neuropathy requires moving beyond single-variant associations to investigate how genetic predispositions interact with environmental triggers and other genetic modifiers to influence disease risk and progression.

Although genetic associations may highlight regions within active regulatory elements, such as super-enhancers, the direct functional impact of specific variants on gene expression and protein function, particularly within relevant cell types like sensory neurons, often remains to be thoroughly validated.[2]Functional studies are frequently limited in scope, focusing on specific genes or pathways without fully exploring broader biological networks that might be perturbed in neuropathy.[2] Bridging the gap between statistical association and biological mechanism is essential for translating genetic findings into actionable clinical insights and therapeutic targets.

Genetic variations play a crucial role in influencing biological pathways that can impact neurological health, including susceptibility to various forms of neuropathy. Several single nucleotide polymorphisms (SNPs) across different genes are implicated in processes ranging from mitochondrial function and cellular stress responses to neural development and synaptic integrity. Understanding these variants helps to elucidate the complex genetic architecture underlying nerve disorders.

Variants associated with genes like PNPT1, EFEMP1, and ATP7B are involved in fundamental cellular processes vital for neuronal health. The PNPT1 gene encodes a mitochondrial RNA exonuclease, critical for mitochondrial RNA processing and overall mitochondrial function, which is essential for the high energy demands of neurons. Variations such as rs12464737 and rs12615158 in the PNPT1-EFEMP1region may alter mitochondrial efficiency or cellular stress responses, potentially contributing to neuronal damage or impaired nerve regeneration, which can manifest as neuropathy.[10] Similarly, the ATP7Bgene is responsible for copper transport, and its proper function is essential for preventing copper accumulation, which can be toxic to neurons and lead to conditions like Wilson’s disease, often presenting with neurological symptoms including neuropathy. The variantrs185185149 within ATP7B could influence copper homeostasis, thus modulating risk for neurological dysfunction and nerve damage.[11] Other variants impact genes crucial for cell cycle regulation and neural development. The CDKN2B-AS1 gene, also known as ANRIL, is a long non-coding RNA that influences the expression of genes involved in cell cycle progression and senescence, including CDKN2A and CDKN2B. Variants like rs6475604 and rs944801 within CDKN2B-AS1may affect cell proliferation and survival pathways, indirectly impacting nerve repair and regeneration after injury or disease. This gene region has been associated with various conditions, including intracranial aneurysms, highlighting its broader role in vascular and neurological health.[12] Disruption of these processes can contribute to the pathophysiology of neuropathies, where nerve cells fail to repair or maintain their integrity. The LNX1 gene, or Ligand of Numb Protein X 1, codes for an E3 ubiquitin ligase involved in protein degradation and signaling pathways critical for neuronal differentiation and synaptic plasticity. The variant rs71597855 in the LNX1-RPL21P44 region might alter these regulatory mechanisms, potentially affecting nerve development or the response to nerve injury.[13]Furthermore, genes involved in cellular signaling and structural integrity also harbor variants relevant to neuropathy.CNBD1(Cyclic Nucleotide Binding Domain Containing 1) is involved in cyclic nucleotide signaling, which plays roles in diverse cellular functions, including neuronal excitability and synaptic transmission. The variantrs75353718 could modify these signaling cascades, affecting nerve impulse conduction or sensory perception. LINC02780 represents a long intergenic non-coding RNA, and variants such as rs12137595 might influence gene expression in neural tissues, thereby indirectly modulating pathways essential for nerve health and function. UNC5D, a member of the UNC5 netrin receptor family, is involved in axon guidance and neuronal apoptosis, processes critical for the proper formation and maintenance of neural circuits. A variant like rs28485846 in UNC5Dcould impact these developmental and apoptotic pathways, potentially leading to neurodevelopmental issues or nerve degeneration. Finally,TRDN(Triadin) plays a role in calcium regulation within muscle cells, particularly in sarcoplasmic reticulum calcium release. While primarily known for its role in muscle, calcium dysregulation can significantly impact neuromuscular junction function and nerve excitability, suggesting that thers11154178 variant in TRDN could contribute to neuromuscular disorders that present with neuropathic symptoms.[14], [15]

RS IDGeneRelated Traits
rs12464737
rs12615158
PNPT1 - EFEMP1Inguinal hernia
neuropathy
rs185185149 ATP7Bneuropathy
rs6475604
rs944801
CDKN2B-AS1open-angle glaucoma
colorectal cancer
Antiglaucoma preparations and miotics use
glaucoma
neuropathy
rs71597855 LNX1 - RPL21P44neuropathy
rs75353718 CNBD1neuropathy
rs12137595 LINC02780neuropathy
rs28485846 UNC5Dneuropathy
rs11154178 TRDNneuropathy

Defining Neuropathy and Its Operationalization

Section titled “Defining Neuropathy and Its Operationalization”

Neuropathy fundamentally refers to any condition affecting the peripheral nervous system, leading to a range of symptoms depending on the nerve fibers involved. In research contexts, particularly large-scale studies utilizing health records, neuropathy is often operationally defined through the presence of specific diagnostic codes. For instance, studies have identified neuropathy through a comprehensive list of International Classification of Diseases, Ninth Revision, Clinical Modification (ICD-9-CM) codes. These codes encompass various presentations such as diabetic neuropathy (codes 356.9, 250.6), amyotrophy (358.1), cranial nerve palsy (951.0, 951.1, 951.3), mono-neuropathy (354.0–355.9), Charcot’s arthropathy (713.5), polyneuropathy (357.2), neurogenic bladder (596.54), autonomic neuropathy (337.0, 337.1), and orthostatic hypotension.[16], [458]This approach provides a standardized, albeit broad, framework for identifying individuals with neuropathy in large datasets, serving as a practical diagnostic criterion for epidemiological and genetic research.

Classification Systems and Subtypes of Neuropathy

Section titled “Classification Systems and Subtypes of Neuropathy”

Neuropathy is not a single entity but a broad category encompassing various subtypes, classified based on etiology, anatomical distribution, and specific clinical manifestations. The operational definitions used in studies highlight this diversity, distinguishing between conditions like diabetic neuropathy, which is defined by its underlying cause, and polyneuropathy or mono-neuropathy, which categorize based on the pattern of nerve involvement. Cranial nerve palsy represents involvement of specific cranial nerves, while autonomic neuropathy affects the involuntary nervous system, manifesting in conditions such as neurogenic bladder or orthostatic hypotension.[16]Further, specific inherited forms exist, such as Charcot-Marie-Tooth Disease Type 1A (CMT1A), which is a distinct nosological entity with its own genetic basis and characteristic subphenotypes, including arthritic-like pain, balance difficulties, and foot deformities.[17]These classifications are crucial for understanding the heterogeneous nature of neuropathy and guiding targeted research and clinical management.

Clinical Assessment and Severity Gradations

Section titled “Clinical Assessment and Severity Gradations”

The clinical assessment of neuropathy involves evaluating symptoms and signs to determine the type, extent, and severity of nerve damage. For inherited conditions like CMT1A, disease severity often progresses with age, and specific clinical signs can vary by sex, with females more prone to arthritic-like pain and difficulties with balance, while males may have a higher likelihood of foot deformity.[17]approaches for neuropathy severity can include objective assessments of muscle strength, such as the Medical Research Council (MRC) standards, which grade strength from 0 (no contraction) to 5 (normal contraction).[17] These objective measures allow for the establishment of severity gradations, where, for example, mild cases of CMT1A might be defined by a strength of 5, while severe cases could involve strength grades from 0 to 3 or 0 to 4.[17] Such detailed clinical criteria and quantitative measurements are vital for both clinical diagnosis and for characterizing phenotypes in research settings, enabling a more dimensional understanding beyond simple categorical presence or absence.

Neuropathy, a condition encompassing damage to the peripheral nervous system, often stems from a clear genetic basis, ranging from single-gene defects to complex inherited predispositions. A prominent example is Charcot-Marie-Tooth Disease Type 1A (CMT1A), which is a hereditary neuropathy directly caused by specific genetic anomalies.[17]Research into CMT1A has also identified various modifier gene candidates, indicating that an individual’s broader genetic makeup can significantly influence the severity and specific characteristics of their neuropathy.[17]These findings underscore how specific inherited variants can directly compromise nerve health and function, leading to clinical neuropathy.

Genetic Influence on Neuronal Health and Integrity

Section titled “Genetic Influence on Neuronal Health and Integrity”

Beyond direct Mendelian forms, a wider array of genetic factors contributes to neuropathy by impacting fundamental biological processes essential for nerve maintenance and resilience. Genes such asARHGEF28 are recognized for their critical roles in regulating neurofilaments and facilitating axon growth and branching, processes vital for healthy nerve function.[18] Dysregulation or mutation within ARHGEF28 can disrupt these pathways, contributing to neuronal dysfunction and aggregate formation as seen in motor neuron diseases, thereby impairing overall neuronal maintenance.[18] Furthermore, other genetic loci, including ZNF318 and NID2, have been associated with neurological functions, suggesting a polygenic landscape where multiple genetic variations collectively influence an individual’s susceptibility to nerve damage.[18]

Foundations of Peripheral Nerve Function and Dysfunction

Section titled “Foundations of Peripheral Nerve Function and Dysfunction”

Neuropathy, a condition affecting peripheral nerves, involves a breakdown in the intricate systems that maintain nerve structure and function. Myelination, the process by which nerve fibers are insulated to ensure rapid signal transmission, is crucial, and the protein frabin, encoded by theFGD4 gene, plays a key role in this process. Disruptions in FGD4 can lead to severe demyelinating conditions like Charcot-Marie-Tooth Type 4H, highlighting the importance of proper myelin sheath formation for nerve health.[19] Beyond insulation, the efficient transport of vital components along nerve axons is powered by motor proteins. For instance, KIF1A and KIF1B encode kinesin family proteins essential for the anterograde transport of organelles and other cargo along axonal microtubules. Mutations in KIF1Aare linked to hereditary sensory neuropathy and spastic paraplegia, while mutations inKIF family genes, including KIF1B, are associated with Charcot-Marie-Tooth disease type 2A.[1] The structural integrity of axons is further maintained by neurofilaments, with the neurofilament light polypeptide, encoded by NEFL, being a critical component; mutations in NEFLcan lead to Charcot-Marie-Tooth disease types 1F and 2E.[1]

Genetic and Epigenetic Influences on Neuropathy

Section titled “Genetic and Epigenetic Influences on Neuropathy”

The susceptibility to neuropathy is significantly shaped by an individual’s genetic makeup, with various genes and their regulatory elements influencing nerve health. Polymorphisms within or near genes such asFGD4, KIF1A, KIF1B, LITAF, and NEFLhave been associated with different forms of peripheral neuropathy, including various types of Charcot-Marie-Tooth disease.[1] These genetic variations can impact gene expression and protein function, as evidenced by associations of polymorphisms in FGD4, CX3CL1, and CALU with expression quantitative trait loci (eQTL) and splicing quantitative trait loci (sQTL).[2]Beyond direct gene mutations, epigenetic modifications, such as DNA methylation, can play a role in modulating neural transmission and sensory disturbances. TheMETTL4 gene, for example, is thought to be involved in methyltransferase activity, and nonsynonymous polymorphisms in this gene may lead to functional changes in its protein product.[3] Furthermore, regulatory regions like super-enhancers can influence gene activity; the genomic region annotated to S1PR1has been found within an active super-enhancer, suggesting a complex interplay between genetic variants and gene regulation in neuropathy development.[2]

Molecular Signaling and Cellular Pathologies

Section titled “Molecular Signaling and Cellular Pathologies”

Neuropathy often arises from disruptions in critical molecular signaling pathways and cellular processes within the nervous system. Sphingosine-1-phosphate receptor (S1P) signaling, particularly involvingS1PR1, has emerged as a key molecular driver in the development of sensory peripheral neuropathy, including that induced by microtubule-targeting agents.[2] Another receptor, S1PR3, is known to mediate sensations of itch and pain through distinct TRP channel-dependent pathways, illustrating the complexity of sensory nerve function.[20]Cellular damage and inflammation are also central to neuropathy’s pathophysiology. For instance,CX3CL1-mediated macrophage activation has been shown to contribute to neuronal apoptosis in dorsal root ganglia, leading to painful peripheral neuropathy.[21] Genes like RSPO4, possibly involved in Wnt/beta-catenin signaling, and ANGPT4, which plays a role in vascular development and angiogenesis, may also indirectly influence neuronal health by affecting cellular growth, differentiation, or the vascular supply to nerves.[1] While IL2RAencodes a regulator of immune responses, its direct relevance to neuronal physiology in the context of neuropathy is less clear, yet immune activation can undoubtedly contribute to nerve damage.[1]

Neuropathy manifests as a diverse group of conditions, ranging from inherited disorders to those induced by external factors like chemotherapy. Inherited neuropathies, such as Charcot-Marie-Tooth disease, encompass a wide spectrum of clinically and genetically distinct peripheral nerve disorders, often characterized by progressive muscle weakness and sensory loss.[19] These conditions underscore the critical role of specific gene functions, like those involved in myelination or axonal transport, in maintaining long-term peripheral nerve health.

Furthermore, drug-induced neuropathies, such as microtubule targeting agent-induced sensory peripheral neuropathy, represent a significant clinical challenge, often being a dose-limiting toxicity that severely impacts patient quality of life.[2]The study of these conditions often focuses on specific neuronal populations, such as sensory neurons and dorsal root ganglia (DRG), which are particularly vulnerable to damage and play a central role in transmitting pain and sensory information.[21]Understanding these varied disease mechanisms, from genetic predispositions to environmental triggers, is crucial for developing effective prevention and treatment strategies.

Neuropathy often originates from dysregulated receptor-mediated signaling pathways that govern neuronal survival, growth, and function. A key example is the sphingosine-1-phosphate receptor 1 (S1PR1) signaling pathway, which has been identified as a molecular driver in the development of microtubule targeting agent-induced peripheral neuropathy. Activation ofS1PR1 initiates intracellular cascades, notably involving the RhoA/ROCK pathway, which in turn leads to the phosphorylation of CRMP2 and subsequent neuronal retraction. This process of structural dismantling contributes significantly to neuropathic symptoms and positions S1PR1 as a promising therapeutic target for intervention.[22] Furthermore, S1PR1activation can modulate central sensitization, as demonstrated by its role in inhibiting neuropathic pain behaviors in models of multiple sclerosis, highlighting its broad impact on pain processing.[23]

Neuroinflammatory Pathways and Intercellular Crosstalk

Section titled “Neuroinflammatory Pathways and Intercellular Crosstalk”

Neuroinflammation and complex intercellular signaling crosstalk are fundamental mechanisms contributing to the initiation and progression of neuropathy. The chemokineCX3CL1 (fractalkine) and its receptor CX3CR1 play a pivotal role in these processes; CX3CL1-mediated macrophage activation has been shown to induce dorsal root ganglion (DRG) neuronal apoptosis and painful peripheral neuropathy, particularly in the context of chemotherapy agents like paclitaxel.[24] This pathway is often driven by NF-κB activation, which upregulates CX3CL1in DRG, exacerbating peripheral sensitization and chronic pain induced by agents such as oxaliplatin.[25] Beyond direct neuro-immune interactions, broader immune components such as the complement system are implicated in neurodegenerative processes, mediating early synapse loss, which contributes to overall neuronal dysfunction and degeneration observed in conditions like visual system degeneration.[26]

Genetic and Epigenetic Regulation of Neuronal Function

Section titled “Genetic and Epigenetic Regulation of Neuronal Function”

Genetic and epigenetic regulatory mechanisms profoundly influence an individual’s susceptibility to and the manifestation of neuropathy by controlling gene expression and protein function. Genome-wide meta-analyses have identified specific single nucleotide polymorphisms (SNPs) such asrs74497159 near S1PR1, rs10771973 near FGD4, and rs11076190 near CX3CL1that are linked to chemotherapy-induced peripheral neuropathy. These genetic variants are often associated with expression quantitative trait loci (eQTLs) or splicing quantitative trait loci (sQTLs), indicating their direct involvement in regulating the transcription and alternative splicing of these critical genes, thereby impacting their functional output in sensory neurons.[22] Such genetic predispositions can alter the baseline activity or responsiveness of key signaling pathways, contributing to the development and progression of neuropathic conditions.

Furthermore, epigenetic modifications and broader transcriptional regulators contribute to neuropathy pathogenesis. For example, a nonsynonymous polymorphism inMETTL4can lead to an amino acid substitution, potentially altering the protein’s function.METTL4is hypothesized to be involved in methyltransferase activity, which could epigenetically modify genomic DNA near neuropathy-related genes, thereby modulating neural transmission and contributing to sensory disturbances.[3] Genes like ZNF648, which may be involved in transcriptional regulation, and IL2RA, a regulator of immune responses, further highlight the complex interplay between genetic control, immune system activity, and neuronal health in the context of peripheral neuropathy.[1]

Neuronal Structural Integrity and Metabolic Homeostasis

Section titled “Neuronal Structural Integrity and Metabolic Homeostasis”

The physical integrity and dynamic organization of neuronal structures are paramount for proper function, and their disruption represents a core mechanism in neuropathy. Microtubule targeting agents (MTAs), commonly used in cancer chemotherapy, induce sensory peripheral neuropathy by interfering with microtubule dynamics. Microtubules are vital for maintaining neuronal morphology, facilitating axonal transport of essential proteins and organelles, and supporting synaptic function. The resulting collapse of this intricate cellular infrastructure leads to axonal degeneration, impaired neurovascular repair, and overall neuronal dysfunction, which are hallmarks of various neuropathic conditions and contribute significantly to dose-limiting toxicities.[22]Beyond direct structural damage, maintaining cellular homeostasis in neurons requires continuous metabolic support and regulated biosynthetic and catabolic processes. While specific metabolic pathway dysregulations leading to neuropathy are diverse and context-dependent, the overall energy state and efficient cellular resource management are critical for neuronal survival and resilience against stressors. For instance, the disruption of axonal transport by MTAs can indirectly impede the delivery of mitochondria and other metabolically active components to distant axonal regions, leading to localized energy deficits and contributing to neuronal degeneration. This highlights how direct structural insult can precipitate broader failures in cellular maintenance and metabolic equilibrium, exacerbating neuropathic pathology.

Pharmacogenetics explores how an individual’s genetic makeup influences their response to drugs, including susceptibility to adverse drug reactions like neuropathy. Genetic variations can affect drug metabolism, transport, and the function of drug targets or related pathways, leading to differences in drug efficacy and toxicity among patients. Understanding these genetic factors is crucial for personalizing treatment strategies and mitigating the risk of drug-induced neuropathy.

Genetic Modulators of Drug Metabolism and Disposition

Section titled “Genetic Modulators of Drug Metabolism and Disposition”

Variations in genes encoding drug-metabolizing enzymes and transporters can significantly impact the pharmacokinetics of drugs associated with neuropathy. For instance, the cytochrome P450 enzymeCYP2C8plays a role in the metabolism of certain microtubule-targeting agents (MTAs), such as paclitaxel, which are known to cause sensory peripheral neuropathy.[27] Genotypes of CYP2C8have been associated with both the risk of developing peripheral neuropathy and the need for early dose reduction in paclitaxel-treated breast cancer patients.[27]These genetic differences can lead to altered drug exposure, where individuals with reduced metabolic capacity might experience higher systemic drug levels, consequently increasing their susceptibility to dose-limiting toxicities like neuropathy. While specific drug transporter variants linked to neuropathy were not detailed in available research, their general role in drug absorption, distribution, and excretion suggests they could similarly influence drug concentrations at neuronal sites.

Target and Pathway Variants Influencing Neuropathy Risk

Section titled “Target and Pathway Variants Influencing Neuropathy Risk”

Beyond drug metabolism, genetic variations in drug target proteins and related signaling pathways can directly influence an individual’s susceptibility to neuropathy. A genome-wide meta-analysis identified a significant role for sphingosine-1-phosphate receptor subtype 1 (S1PR1) in microtubule-targeting agent-induced sensory peripheral neuropathy (MTA-induced PN).[2]Specifically, the single nucleotide polymorphism (SNP)rs74497159 within the genomic region annotated to S1PR1 was strongly associated with this toxicity.[2] Functional studies have further demonstrated that S1PR1activation is required for paclitaxel-induced neuropathic pain, and dysregulation of sphingolipid metabolism, which involvesS1PR1, contributes to bortezomib-induced neuropathic pain.[2] Other SNPs, such as rs10771973 in FGD4 and rs11076190 in CX3CL1, are also linked to chemotherapy-induced peripheral neuropathy (CIPN) and are associated with expression or splicing quantitative trait loci, indicating potential roles in gene expression regulation.[2] CX3CL1-mediated macrophage activation, for example, has been implicated in paclitaxel-induced neuronal apoptosis and painful peripheral neuropathy, highlighting how these target and pathway variants can modulate drug pharmacodynamics and contribute to adverse neurological outcomes.[2]

Clinical Considerations and Implementation Challenges

Section titled “Clinical Considerations and Implementation Challenges”

The pharmacogenetic insights into neuropathy, particularly MTA-induced PN, hold promise for personalized prescribing, yet their clinical implementation faces several challenges. The validation ofS1PR1 signaling provides pharmacogenetic evidence that supports ongoing clinical investigations aimed at targeting this pathway for the prevention or treatment of CIPN.[2] This suggests a future where genetic testing could guide drug selection or prophylactic interventions for at-risk patients. However, current pharmacogenetic studies, including large-scale meta-analyses, have sometimes been limited by insufficient power and difficulties in replicating top-ranking SNPs across diverse cohorts, which can be attributed to differences in treatment regimens, sample sizes, and phenotyping methods.[2]The use of broad clinical grading scales for peripheral neuropathy, such as NCI-CTCAE, may also underestimate symptom progression and introduce inconsistencies, further complicating the establishment of robust genotype-phenotype associations for clinical guidelines.[2]Overcoming these limitations through larger, well-phenotyped studies and consistent genetic validation is essential for translating pharmacogenetic findings into actionable dosing recommendations and personalized treatment strategies to minimize neuropathy risk.

Frequently Asked Questions About Neuropathy

Section titled “Frequently Asked Questions About Neuropathy”

These questions address the most important and specific aspects of neuropathy based on current genetic research.


1. My parent has nerve problems; will I get them too?

Section titled “1. My parent has nerve problems; will I get them too?”

Yes, some forms of neuropathy are directly inherited. Mutations in genes likeFGD4 (involved in nerve insulation) or KIF1A(crucial for nerve signal transport) can lead to inherited conditions such as Charcot-Marie-Tooth disease. If your parent has an inherited form, you might have a higher chance of inheriting the genetic predisposition, though severity can vary.

2. Why did my treatment cause nerve damage, but not my friend’s?

Section titled “2. Why did my treatment cause nerve damage, but not my friend’s?”

It’s often due to your unique genetic makeup. Genes like IL2RA, ZNF648, RSPO4, ANGPT4, and S1PR1have been linked to an increased risk of drug-induced peripheral neuropathy. These genetic variations can make your nerves more susceptible to damage from certain medications, even if your friend takes the same treatment without issues.

3. Why do my nerve symptoms feel so different from others?

Section titled “3. Why do my nerve symptoms feel so different from others?”

Neuropathy symptoms are highly variable because different types of nerves can be affected, and your genetic background influences this. You might have sensory neuropathy (affecting sensation) while someone else has motor (muscle movement) or autonomic neuropathy. Genetic factors, such as mutations in genes likeNEFL or LITAF, contribute to these distinct presentations.

4. Am I just more likely to get nerve issues in general?

Section titled “4. Am I just more likely to get nerve issues in general?”

Your genetics can play a significant role in your overall susceptibility to neuropathy. Beyond inherited forms, certain gene variations can make you more prone to developing nerve damage from external factors like toxins, infections, or even systemic diseases like diabetes. This genetic predisposition means your nerves might be inherently more vulnerable compared to others.

5. Could a special test show my risk for nerve problems?

Section titled “5. Could a special test show my risk for nerve problems?”

Yes, genetic testing can identify specific gene variants linked to neuropathy risk. For example, it can show if you carry variations associated with inherited conditions or an increased susceptibility to drug-induced neuropathy. This information can be crucial for personalized risk assessment and treatment planning.

6. Why does my mild numbness sometimes get much worse?

Section titled “6. Why does my mild numbness sometimes get much worse?”

The progression and severity of neuropathy symptoms can be influenced by multiple factors, including your underlying genetic susceptibility. While external triggers like physical injury or certain medications can worsen symptoms, your genetic profile might affect how your nerves respond to stress or damage. This can lead to fluctuations in symptom intensity.

7. Can my genes affect my ability to do my job daily?

Section titled “7. Can my genes affect my ability to do my job daily?”

Absolutely. If you have a genetic predisposition to motor neuropathy, it can lead to muscle weakness and difficulty with movement, directly impacting your physical capabilities at work. Similarly, sensory neuropathy can cause chronic pain or numbness, affecting fine motor skills or prolonged standing. Your genetic profile dictates the type and severity of nerve damage, which in turn influences your daily functioning.

8. Does my family background change my risk for nerve issues?

Section titled “8. Does my family background change my risk for nerve issues?”

Yes, your family background, especially your genetic ancestry, can influence your risk. Certain genetic variations linked to neuropathy might be more prevalent in specific populations. This can impact both your susceptibility to inherited forms of neuropathy and how you respond to environmental triggers or medications.

9. Why do some people never get nerve damage from certain drugs?

Section titled “9. Why do some people never get nerve damage from certain drugs?”

It often comes down to individual genetic differences. While some people have genetic variations in genes like S1PR1, CX3CL1, or CALUthat make them more vulnerable to drug-induced neuropathy, others lack these specific predispositions. Their genetic makeup provides a degree of protection, allowing them to tolerate medications that might cause nerve damage in genetically susceptible individuals.

10. Do my genes impact how my body handles digestion problems?

Section titled “10. Do my genes impact how my body handles digestion problems?”

Yes, your genes can influence your susceptibility to autonomic neuropathy, which affects involuntary bodily functions like digestion. If you have genetic factors that make you prone to this type of nerve damage, you might experience issues like altered gut motility or digestive discomfort. While diet plays a role, your genetic predisposition can make you more vulnerable to these digestive challenges.


This FAQ was automatically generated based on current genetic research and may be updated as new information becomes available.

Disclaimer: This information is for educational purposes only and should not be used as a substitute for professional medical advice. Always consult with a healthcare provider for personalized medical guidance.

[1] Leger, P. D., et al. “Genome-wide association study of peripheral neuropathy with D-drug-containing regimens in AIDS Clinical Trials Group protocol 384.”J Neurovirol, vol. 20, no. 2, 2014, pp. 164-173.

[2] Chua, K. C., et al. “Genome-Wide Meta-Analysis Validates a Roless for S1PR1 in Microtubule Targeting Agent-Induced Sensory Peripheral Neuropathy.”Clin Pharmacol Ther, vol. 110, no. 3, 2021, pp. 783-792.

[3] Kobayashi, D., et al. “Genome-wide association study of sensory disturbances in the inferior alveolar nerve after bilateral sagittal split ramus osteotomy.” Mol Pain, vol. 9, no. 1, 2013, pp. 49.

[4] Meyer, H. V., et al. “Genetic and functional insights into the fractal structure of the heart.” Nature, vol. 584, no. 7821, 2020, pp. 433–439.

[5] Traylor, M., et al. “Genome-wide meta-analysis of cerebral white matter hyperintensities in patients with stroke.”Neurology, vol. 85, no. 24, 2015, pp. 2164–2170.

[6] Ishigaki, K., et al. “Large-scale genome-wide association study in a Japanese population identifies novel susceptibility loci across different diseases.” Nat Genet, vol. 52, no. 7, 2020, pp. 669–679.

[7] Chung, S. J., et al. “Genomic determinants of motor and cognitive outcomes in Parkinson’s disease.”Parkinsonism Relat Disord, vol. 18, no. 5, 2012, pp. 603–608.

[8] Malik, R., et al. “Multiancestry genome-wide association study of 520,000 subjects identifies 32 loci associated with stroke and stroke subtypes.”Nat Genet, vol. 50, no. 4, 2018, pp. 524–533.

[9] Fitzgerald, K. C., et al. “Early complement genes are associated with visual system degeneration in multiple sclerosis.”Brain, vol. 142, no. 8, 2019, pp. 2225–2237.

[10] Pankratz N, et al. “Genomewide association study for susceptibility genes contributing to familial Parkinson disease.” Hum Genet. PMID: 18985386.

[11] Wain LV, et al. “Genome-wide association study identifies six new loci influencing pulse pressure and mean arterial pressure.” Nat Genet. PMID: 21909110.

[12] Foroud T, et al. “Genome-wide association study of intracranial aneurysms confirms role of Anril and SOX17 in disease risk.” Stroke. PMID: 22961961.

[13] Jiang Y, et al. “Propensity score-based nonparametric test revealing genetic variants underlying bipolar disorder.” Genet Epidemiol. PMID: 21254220.

[14] Fanous AH, et al. “Genome-wide association study of clinical dimensions of schizophrenia: polygenic effect on disorganized symptoms.” Am J Psychiatry. PMID: 23212062.

[15] Thompson SD, et al. “Genome-wide association analysis of juvenile idiopathic arthritis identifies a new susceptibility locus at chromosomal region 3q13.” Arthritis Rheum. PMID: 22354554.

[16] Vujkovic, M. et al. “Discovery of 318 new risk loci for type 2 diabetes and related vascular outcomes among 1.4 million participants in a multi-ancestry meta-analysis.” Nat Genet, 2020.

[17] Tao, F. et al. “Modifier Gene Candidates in Charcot-Marie-Tooth Disease Type 1A: A Case-Only Genome-Wide Association Study.”J Neuromuscul Dis, 2019.

[18] Wells, H. R. R., et al. “GWAS Identifies 44 Independent Associated Genomic Loci for Self-Reported Adult Hearing Difficulty in UK Biobank.” Am J Hum Genet, vol. 105, no. 4, 2019, pp. 788-802. PMID: 31564434.

[19] Delague, Valerie, et al. “Mutations in FGD4 Encoding the Rho GDP/GTP Exchange Factor FRABIN Cause Autosomal Recessive Charcot-Marie-Tooth Type 4H.” American Journal of Human Genetics, vol. 81, no. 1, 2007, pp. 1-16.

[20] Hill, R. Z., et al. “S1PR3 Mediates Itch and Pain via Distinct TRP Channel-Dependent Pathways.”Journal of Neuroscience, vol. 38, no. 36, 2018, pp. 7833-7843.

[21] Huang, Z. Z., et al. “CX3CL1-mediated Macrophage Activation Contributed to Paclitaxel-induced DRG Neuronal Apoptosis and Painful Peripheral Neuropathy.”Brain, Behavior, and Immunity, vol. 40, 2014, pp. 155-165.

[22] Chua, K. C., et al. “Genome-Wide Meta-Analysis Validates a Roless for S1PR1in Microtubule Targeting Agent-Induced Sensory Peripheral Neuropathy.”Clin Pharmacol Ther, vol. 108, no. 3, 2020, pp. 605–615.

[23] Doolen, Sarah, et al. “Fingolimod Reduces Neuropathic Pain Behaviors in a Mouse Model of Multiple Sclerosis by a Sphingosine-1 Phosphate Receptor 1-Dependent Inhibition of Central Sensitization in the Dorsal Horn.”Pain, vol. 159, no. 2, 2018, pp. 224-238.

[24] Wang, Jing, et al. “CX3CL1-Mediated Macrophage Activation Contributed to Paclitaxel-Induced DRG Neuronal Apoptosis and Painful Peripheral Neuropathy.”Brain, Behavior, and Immunity, vol. 40, 2014, pp. 155-165.

[25] Wang, Jing, et al. “Upregulation of CX3CL1 Mediated by NF-κB Activation in Dorsal Root Ganglion Contributes to Peripheral Sensitization and Chronic Pain Induced by Oxaliplatin Administration.”Molecular Pain, vol. 13, 2017, pp. 1-10.

[26] Ramakrishnan, S., et al. “Complement and Microglia Mediate Early Synapse Loss in Alzheimer Mouse Models.” Science, vol. 352, 2016, pp. 712-716.

[27] Lam, S. W., et al. “Genotypes of CYP2C8 and FGD4 and their association with peripheral neuropathy or early dose reduction in paclitaxel-treated breast cancer patients.”British Journal of Cancer, vol. 115, no. 11, 2016, pp. 1335-1342.

[28] Benjamin, E. J., et al. “Genome-wide association with select biomarker traits in the Framingham Heart Study.” BMC Med Genet, vol. 8, 2007.

[29] Ebenezer, G. J., et al. “Impaired Neurovascular Repair in Subjects with Diabetes Following Experimental Intracutaneous Axotomy.” Brain, vol. 134, no. 6, 2011, pp. 1853-1863.

[30] Fontanillas, P. et al. “Genome-wide association study of pain sensitivity assessed by questionnaire and the cold pressor test.”Pain, 2022.

[31] Kays, J. S., et al. “Expression of Sphingosine 1-Phosphate Receptors in the Rat Dorsal Root Ganglia and Defined Single Isolated Sensory Neurons.”Physiological Genomics, vol. 44, no. 18, 2012, pp. 889-901.

[32] Old, Elizabeth A., et al. “Monocytes Expressing CX3CR1 Orchestrate the Development of Vincristine-Induced Pain.”Journal of Clinical Investigation, vol. 124, no. 5, 2014, pp. 2023-2036.

[33] Quarta, Silvia, et al. “Sphingosine-1-Phosphate and the S1P3 Receptor Initiate Neuronal Retraction via RhoA/ROCK Associated with CRMP2 Phosphorylation.”Frontiers in Molecular Neuroscience, vol. 10, 2017, p. 13.

[34] Ripke, S., et al. “Biological insights from 108 schizophrenia-associated genetic loci.”Nature, vol. 511, no. 7510, 2014, pp. 421–427.

[35] Turley, P., et al. “Multi-trait analysis of genome-wide association summary statistics using MTAG.” Nat Genet, vol. 50, no. 2, 2018, pp. 290–295.