Abstract
Chronic pain is a debilitating condition that affects roughly a third to a half of the world’s population. Despite its substantial effect on society, treatment for chronic pain is modest, at best, notwithstanding its side effects. Hence, novel therapeutics are direly needed. Emerging evidence suggests that calcium plays an integral role in mediating neuronal plasticity that underlies sensitization observed in chronic pain states. The endoplasmic reticulum and the mitochondria are the largest calcium repositories in a cell. Here, we review how stressors, like accumulation of misfolded proteins and oxidative stress, influence endoplasmic reticulum and mitochondria function and contribute to chronic pain. We further examine the shuttling of calcium across the mitochondrial-associated membrane as a mechanism of cross-talk between the endoplasmic reticulum and the mitochondria. In addition, we discuss how endoplasmic reticulum stress, mitochondrial impairment, and calcium dyshomeostasis are implicated in various models of neuropathic pain. We propose a novel framework of endoplasmic reticulum–mitochondria signaling in mediating pain hypersensitivity. These observations require further investigation in order to develop novel therapies for chronic pain.
Keywords: Endoplasmic reticulum stress, mitochondria, mitochondrial-associated membrane, calcium, pain
Introduction
Pain is an adaptive response to noxious stimuli that cause tissue injury or have the potential to cause an injury.1 It is the most common symptom across a wide variety of medical conditions ranging from autoimmune disorders like rheumatoid arthritis and multiple sclerosis, to traumatic nerve injury2,3 and is a major reason for seeking consultation with a physician.4,5 When pain loses its adaptive value and becomes persistent, even after the initial injury has healed, it becomes a disease in its own right.1
Studying pain in a laboratory setting is particularly difficult due to the subjective nature of pain perception. To overcome this obstacle, researchers carefully control the extent of the injury or disease and closely monitor nocifensive behaviors in animals. Models of neuropathic pain, such as spinal nerve ligation (SNL) and spared nerve injury (SNI), typically induce traumatic damage to the nerve resulting in evoked and spontaneous pain behaviours.6,7 Other disease-oriented pain models, like diabetic neuropathy and chemotherapy-induced pain, affect the molecular integrity of nerves and typically induce a more global, complex injury response involving inflammation, immune activation, and gliosis as well as changes in the neurovasculature.8,9 The underlying pathology responsible for pain hypersensitivity across these conditions remains enigmatic. It is clear, however, that pain is a common feature of many medical conditions, regardless of their aetiologies. Whether these pain states share common or divergent mechanisms is being actively investigated.
Chronic neuropathic pain results from a lesion or disease of the somatosensory nervous system comprising of peripheral sensory neurons as well as the spinal cord and the brain.10 Enhanced excitability, or sensitization, throughout this pain axis is thought to mediate neuropathic pain. In the central nervous system (CNS), changes in the response properties of complex neuronal and glial networks, such as disinhibition, contribute to persistent evoked and spontaneous pain.11,12 In the periphery, however, specialized cells, known as nociceptors, are responsible for transducing painful stimuli. These cells typically have a high stimulation threshold allowing them to fire action potentials when exposed to noxious heat, chemicals, and mechanical stimulation.13,14 In concert with other sensory neurons, nociceptors inform the CNS about the nature, location, and intensity of the painful stimulus.15 Enhancement of the response properties of sensory neurons, known as peripheral sensitization, can arise and is response to neuropathy caused by tissue injury and inflammation or lesions to the nerve fibers.16,17 Sensitization leads to hyperalgesia (increased sensitivity to painful stimulus) and allodynia (pain from a previously non-noxious stimulus).18
Major efforts have been made to understand the mechanisms that underlie the sensitization of nociceptors.15,18,19 Some molecules that might be held responsible include reactive oxygen species (ROS), adenosine triphosphate (ATP), and calcium (Ca2+).20 Besides providing energy for many cellular processes, ATP acts as a nociceptive transmitter via the purinergic receptors, sensitizing nociceptors to further stimulation.21 ROS such as superoxide (SO) are chemically reactive molecules that are produced as a by-product of the chemical processes in the mitochondria, lysosomes, and the endoplasmic reticulum (ER). High levels of ROS during times of stress lead to oxidation of nucleic acids, lipids, and proteins in a process known as oxidative stress.22 SO and its descendant peroxynitrite have been linked to alterations in protein kinases, glutamatergic signaling, and ion channel modulation in the development of hypersensitivity.23 Ca2+ is a multifunctional ion that is involved in neuronal plasticity by regulating neurotransmitter release, action potential propagation, and changes in gene expression.24 The role of plasmalemmal Ca2+ signaling through voltage-gated Ca2+ channels and transient receptor potential (TRP) channels is well-documented in pain pathophysiology.25 Ca2+-signaling also results in increased production of both ROS and ATP by mitochondria, and thus may serve as a master regulator between these molecules and the development of nociceptor sensitization. However, the precise mechanisms that regulate intracellular Ca2+ (iCa2+) stores to mediate pain remain to be fully elucidated.
In this regard, the ER and mitochondria are two organelles that act together as an important Ca2+ signaling hub and as such, they may represent a fundamental regulator of iCa2+, excitability and neuronal plasticity.26 Indeed, emerging evidence is beginning to shed light on the critical role these two organelles play in nociceptor sensitization and chronic pain. Given their important position within the cell for Ca2+ regulation, this review will discuss the complex relationship between the ER and mitochondria and explore how dysregulation within and between these organelles can impact on nociceptor sensitization and the implications this has for chronic pain.
The endoplasmic reticulum
The ER is perhaps the largest tubular organelle in the cell.27 In neurons, the ER is continuous throughout the cell, extending from the dendritic arbors to the cell soma, across the axon and into the presynaptic terminals.28 The ER has classically been subdivided into the rough ER (containing ribosomes), the smooth ER (without ribosomes) and the nuclear envelope.27 The ER performs a variety of functions. Most notably, a host of luminal and transmembrane chaperones and foldases help synthesize and mature proteins in the ER.29 In addition, the ER is also important for lipid synthesis. As such, the smooth ER synthesizes phospholipids, glycolipids, and cholesterol which are used for various functions such as synthesizing steroid hormones from cholesterol.30 The ER also contains many detoxifying enzymes that metabolize lipid soluble molecules by converting them into polar, water-soluble compounds that are then secreted from the cell and the body.31 In addition, the ER is the cell’s largest dynamic repository of Ca2+.26 Many of the ER’s chaperones (e.g., BiP, calreticulin, calnexin) are low-affinity, high-capacity Ca2+ binding proteins (CBPs).26 Their function as chaperones also heavily depends on the luminal concentration of Ca2+.32
Since the ER extends throughout the cell, it also serves as a highway for RNAs, proteins, lipids, and ions.27 Changes in the intraluminal environment of the ER, such as increased protein synthesis and altered Ca2+ homeostasis, may lead to ER stress which is initially protective but detrimental in the long-term.33
The unfolded protein response
Stressful conditions such as amino acid deprivation, viral infection, heme deficiency, and accumulation of unfolded proteins lead to the induction of a larger translational regulation pathway known as the integrated stress response (ISR). The ISR is mediated by the stimulation of four kinases, general control nonderepressible 2 kinase (GCN2), protein kinase R (PKR), heme-regulated eIF2α kinase (HRI), and protein kinase RNA-like ER kinase (PERK), respectively.34 These kinases converge onto the phosphorylation of eukaryotic initiation factor 2 α (eIF2α) regulating its translational capacity and allowing the cell to aptly react to the stressor.34
Protein synthesis, folding, and quality control are canonical functions of the ER. Upon increased protein demand, the ER responds to an accumulation of unfolded or misfolded proteins by initiating a series of cellular processes collectively known as the unfolded protein response (UPR) (Figure 1).34 The goal of the UPR is to reduce translation of proteins, increase the expression of chaperones, and degrade misfolded proteins. This response is mediated by three downstream transducers: PERK, inositol-requiring enzyme 1α (IRElα), and activating transcription factor 6 (ATF6).34 Each of these ER resident proteins contains a single transmembrane domain and a single luminal domain.35 Under stressed conditions, binding immunoglobulin protein (BiP; also known as heat shock protein A 5, HSPA5, and glucose response protein 78, GRP78) dissociates from ATF6, PERK, and IRE1α activating their respective arm of the UPR.34 BiP, then, actively folds unfolded proteins using its allosteric ATPase activity and prevents the formation of protein aggregates by binding and restraining unfolded proteins.36
Figure 1.
The unfolded protein response of the ER. The dissociation of BiP from PERK and IRE1 induces oligomerization and autophosphorylation of these kinases, initiating the unfolded protein response. Activated IRE1 acts as an endoribonuclease and splices XBP1 mRNA (XBP1s) generating a potent transcription factor that enhances the production of ER chaperones and genes associated with ER-associated degradation. PERK activation enhances phosphorylation of eIF2α as part of the integrated stress response. This results in the suppression of global translation, and expression of stress-induced genes like ATF4, CHOP, and GADD34. In concert with protein phosphatase 1, GADD34 functions to reverse eIF2α phosphorylation and resume global translation. Many of ER’s chaperones, including BiP, require Ca2+ for proper functioning. SERCA actively pumps Ca2+ into the ER while the activation of IP3Rs and RyRs releases Ca2+ into the cytosol. Stressors, like inflammation, may modulate the activity of ER Ca2+ transporters and thus influence ER functioning.
Once BiP is sequestered from PERK’s luminal domain, PERK autophosphorylates and oligomerizes as either a dimer or a transient tetramer.37 The extent of oligomerization of PERK may represent the degree of unfolded proteins in the ER since the tetramer configuration is a more potent kinase than the dimer.34 Upon activation, PERK phosphorylates eukaryotic initiation factor 2 α (eIF2α) reducing the general translation of proteins while enhancing translation of select mRNAs. In particular, PERK-eIF2α signaling increases translation of those transcripts that encode an upstream open reading frame (uORF).38 eIF2α phosphorylation enhances the expression of activating transcription factor 4 (ATF4) which in turn increases the expression of the proapoptotic transcription factor C/EBP homologous protein (CHOP).39 ER stress and the UPR have been linked to several neuropathic pain states, including peripheral nerve injury (PNI), diabetic neuropathy, chemotherapy-induce neuropathic pain. Furthermore, inhibiting certain arms of the ER stress pathway attenuates pain hypersensitivity.40,41 Mechanisms supporting these observations will be discussed below.
Mitochondria
Mitochondria are unique organelles thought to have originated from bacteria that were endocytosed by an early eukaryote. They have their own DNA (mtDNA) that encodes for proteins which allow for efficient aerobic energy production.42 They are composed of both an outer mitochondrial membrane (OMM) and inner mitochondrial membrane (IMM). The IMM encloses the matrix, and has many folds, referred to as cristae. Cristae increase the surface area available to harbor the five transmembrane proteins involved in energy production by oxidative phosphorylation (Complex I–V).43 These transmembrane proteins pump hydrogen ions into the intermembrane space, creating a potential difference across the IMM (ΔΨm) that is used by the protein ATP synthase (Complex V) to make ATP. The ΔΨm also provides an electrochemical driving force for the movement of ions such as Ca2+ across the IMM.44 An important byproduct of mitochondria metabolism is the generation of ROS, which are important signaling molecules, but under pathological conditions can lead to oxidative stress and exacerbated damage.45
Mitochondria form dynamic and integrated tubular networks, regulated by proteins such as MFN1 and MFN2, which promote mitochondrial fusion, and by Fis1 and Drp1 which regulate fission.46,47 These networks not only allow for the coordination of ATP production spatially and temporally but also play a role in regulating mitochondria–cell communication involved in metabolism, the cell cycle, stress responses, and cell death cascades.46 Although the mechanism of how mitochondrial networks could mediate these processes is still under investigation, there are numerous hypotheses. For example, mitochondrial networks may control energy production through Ca2+ signaling. In a fused mitochondrial network, Ca2+ can diffuse throughout the network thus allowing small changes in Ca2+ to have a greater affect and reach more sites of energy production.48,49
Mitochondrial-associated membrane
Mitochondria interact with the ER through a specialized domain called the mitochondria-associated ER membrane (MAM). The MAM is composed of contact sites tethered by protein complexes, which facilitate ER–mitochondria crosstalk and juxtaposition (as reviewed by Veeresh et al.50 and Vance51). The ER–mitochondria encounter structures (ERMES) complex regulates phospholipid transfer as well as mitochondrial dynamics and mitophagy.52 Phosphofurin acidic sorting cluster 2 (PACS2) regulates communication between the ER and mitochondria, ER homeostasis, and apoptosis.53 Presenilin 2 (PS2) in the presence of mitochondrial fusion protein mitofusin (MFN2) promotes ER–mitochondrial tethering.54 The IP3R-S1R-GRP75-VDAC complex is crucial for Ca2+ transfer from the ER to mitochondria. The inositol triphosphate receptor (IP3) receptor in the ER is stabilized by the sigma1 receptor (S1R) to allow Ca2+ efflux. This complex is tethered by the chaperone glucose response protein 75 (GRP75) to the voltage-dependent anion channel (VDAC) to facilitate mitochondrial uptake of Ca2+.50,55 In addition, vesicle-associated membrane protein-associated protein B (VAPB) in the ER associates with protein tyrosine phosphatase-interacting protein 51 (PTPIP51) in the mitochondria to help facilitate Ca2+ transfer.56
In neurons, ER–mitochondrial interactions through the MAM can significantly influence many key processes relevant to nociceptor function such as mitochondrial bioenergetics, Ca2+ homeostasis, and action potential generation.55,57,58 For example, Ca2+ regulates activity-dependent signaling and neuronal excitability, and in nociceptors this regulation is important to prevent aberrant signaling and pain.59 Thus, Ca2+ modulation at the MAM can play a role in pain control through nociceptor excitability.
ER stress results in various functional and structural changes at the MAM.60 ER stress can lead to increased MAM contacts and strengthened Ca2+ transfer to mitochondria, which boosts energy production.61 Interestingly, some proteins known to be involved in the UPR, IRE1, and PERK perform noncanonical functions at the MAM.62,63 IRE1 localized at the MAM regulates IP3 receptors and maintains mitochondrial bioenergetics.62 Whereas PERK at the MAM regulates juxtaposition between the ER and mitochondria and promotes apoptosis by conveying ROS signals and maintaining high levels of CHOP.63
Sigma1 receptors (S1Rs) are also important in the development of neuropathic pain. In models of neuropathic pain, hypersensitivity can be reduced using S1R knockouts and pharmacological S1R antagonists.64–66 In addition to their role at the MAM, S1Rs can translocate to both the plasma membrane and nucleus to modulate voltage-gated channel activity.67 These diverse functions present multiple pathways through which S1Rs are be involved in neuropathic pain. The S1R antagonist MR309 reduced chemotherapy-induced neuropathic pain in clinical trials, demonstrating its promise as a therapeutic target in pain.68
ER Ca2+ regulation and the impact on pain
Voltage gated Ca2+ channels, TRP channels, glutamatergic channels, and purinergic receptors on the plasma membrane regulate the influx of extracellular Ca2+ in sensory neurons and are heavily involved in pain pathogenesis following disease, injury, and inflammation (for an extensive review, see Bourinet et al.25). Plasma membrane Ca2+-ATPase (PMCA) and Na+/Ca2+ exchanger (NCX) are primarily responsible for extruding cytosolic Ca2+.69 The bulk of the intracellular Ca2+ is stored in the ER.27 Since Ca2+ is a multifunctional ion involved in a variety cellular processes, the ER can directly impact gene expression, bioenergetics, ion channel physiology, and enzyme function.27 With the presence of Ca2+ transporters on the ER membrane, the ER can be excited to release or sequester Ca2+, tightly regulating the concentration of available cytosolic Ca2+. In the ER, Ca2+ is stored as bound to CBPs, many of which are chaperones, like calnexin, calreticulin, and BiP.26,27 By and large, each CBP binds to many Ca2+ molecules in a low-affinity, high-capacity manner such that the opening of a Ca2+ exporters allows for the immediate dissociation of Ca2+ from the CBPs.26 The ER also maintains a concentration of free Ca2+ unbound to CBPs. Free Ca2+ determines the driving force of Ca2+ release, while Ca2+ bound to CBPs determines the total amount of Ca2+ that can be released.70,71 Ca2+ efflux from the ER is mediated by Ca2+-sensitive ryanodine receptors (RyRs) and inositol triphosphate receptors (IP3Rs), while the influx of Ca2+ into the ER is mediated by sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) pumps in concert with store-operated Ca2+ entry (SOCE) (Figure 1).
Ryanodine receptors and pain
RyRs are named after their antagonist, ryanodine, which was originally used as an insecticide. RyRs are one of the largest membrane proteins consisting of four-identical subunits arranged in a four-leaf clover-shaped structure.72,73 There are three variants of RyRs (RyR1, RyR2, and RyR3) all of which can be activated by free Ca2+ in the cytosol in a process known as Ca2+-induced Ca2+ release or CICR.27 RyR1 is the most sensitive to cytosolic Ca2+ followed by RyR2 and then RyR374,75 RyR3s were classically known as the “brain” isoform; however, all three isoforms can be detected in the nervous system spanning across different cell types and being expressed in virtually every cellular compartment.26,76,77 RyRs are not exclusively selective for Ca2+. Instead, they also counter-conduct other cations such as Mg2+, Li+, Na+, and K+.78 This allows the channel to release Ca2+ without largely affecting the membrane potential.78 RyRs can associate with other proteins to further enhance a cellular Ca2+ response.79Voltage-gated calcium channels and Ca2+-sensitive large-conductance K+ (BK) channels are functionally coupled to RyRs.80,81 As a result, cytosolic Ca2+ increase due to neuronal activity can enhance Ca2+ release from the ER which in turn readily affects action potential dynamics via Ca2+ sensitive channels.81
A growing number of diseases and conditions are linked to impaired RyR function such as genetic RyR mutations in malignant hyperthermia, central core disease, and catecholaminergic polymorphic ventricular tachycardia.82 Aberrant RyR function has also been implicated in neurodegenerative disorders like Alzheimer’s disease.83 Recently, pain and hyperalgesic priming have been linked to RyR sensitization in rats. A single dose of a priming stimulus, like ryanodine, induces long-lasting changes in the nervous system such that a second pain-inducing stimulus, like prostaglandin E2, provokes a much more profound algesic response than would be observed without the primer. In female mice, a low dose (1 pg) of ryanodine is sufficient to induce priming compared to a much higher dose of 100 ng in males.84 This heightened sensitivity to ryanodine in females is mediated by estrogen receptor α (ERα) such that antagonizing the ERα using antisense oligonucleotides abolishes the priming phenotype. Activating ERα using an agonist, propyl pyrazole triol, promotes priming in the absence of ryanodine.84
IP3Rs and pain
IP3Rs are another class of ER Ca2+ release channel. IP3Rs respond to inositol triphosphate (IP3) which is catalyzed by phospholipase C (PLC) typically after the activation of plasmalemmal G-protein coupled receptors (GPCRs) such as metabotropic glutamate receptors (Figure 3).26 Like the RyRs, there are three types of IP3Rs: IP3R1, IP3R2, and IP3R3. The channel may form as homo- or hetero-tetramers.26 IP3Rs can be further modulated by (de)phosphorylation by various kinases, like protein kinase A, B, and C, and phosphatases like calcineurin.85 Further splice variations in the IP3R genes allow for even more functional diversity of the channel despite 60%–80% shared homology among IP3R isotypes.26 IP3Rs open when both IP3 and Ca2+ bind to the receptor.86 It is believed that IP3 primes the receptor to cytosolic Ca2+ which allows for the opening of the channel in a Ca2+-dependent manner.86 IP3R1s are maximally activated around 300–400 nM of Ca2+ beyond which they lose their sensitivity to intracellular Ca2+.87 In contrast, IP3R2s and IP3R3s are not inhibited by high Ca2+ concentration.88 Activation of the IP3Rs may lead to widespread Ca2+ waves or be confined to microdomains depending on the sensitivity of IP3Rs to IP3, the density of IP3Rs, degradation of IP3, and the cell’s buffering capacity for IP3.27 Since IP3Rs are co-activated by intracellular Ca2+, RyR-mediated CICR enhances Ca2+ release from IP3Rs (and vice versa) possibly contributing to widespread Ca2+ waves in the ER.27,86
Figure 3.
Mitochondria both regulate, and are regulated by Ca2+. Ca2+ enters the outer mitochondrial membrane through the VDAC, then subsequently is transported into the mitochondial matrix via the MCU. Ca2+ homeostasis within the mitochondrial matrix is maintained by extrusion via the NCLX, which exchanges three Na+ for one Ca2+. However, mitochondria can also buffer excess Ca2+ by precipitating it as calcium-phosphate. Ca2+ can also regulate various mitochondrial processes such as fission and fusion, ATP production, ROS production, and mPTP opening. Reducing ATP and increasing ROS can cause widespread dysfunctions within the cell. Opening of the mPTP depolarizes the mitochondrial membrane potential as it allows Ca2+ efflux, and it can also trigger apoptosis by release of cytochrome C.
The coupling of IP3 production with stimulation of GPCRs allows the cell to regulate intracellular Ca2+ release in response to the external environment (Figure 3). As such, the binding of bradykinin, an inflammatory mediator, to its B2 receptor induces IP3 production, enhances Ca2+ release from the ER, activates Ca2+-sensitive calmodulin, and contributes to neuronal sensitization.89,90 The link between bradykinin and inflammatory pain has been well-documented.90,91Fractalkine-induced inflammation and subsequent hyperalgesia was recently shown to be mediated by IP3-induced Ca2+ transients in microglia.92 Like ryanodine, a low dose (1–3 pg) of IP3 induces hyperalgesic priming in female, but not male, rats in a ERα dependent manner.93 Interestingly, both ryanodine-induced and IP3-induced hyperalgesic priming can be blocked by inhibiting either IP3Rs or RyRs, respectively.93 This observation in females implies that Ca2+ release from the ER intricately links RyRs and IP3Rs, possibly in a CICR manner that is sex specific.
SERCA pumps and pain
Cytosolic Ca2+ concentrations are tightly regulated by SERCA pumps. These channels belong to a class of P-type Ca2+ ATPases that respond to increases in intracellular Ca2+ levels by actively sequestering the ion from the cytosol into the ER (Figure 1).26 Like other ER Ca2+ transporters, there are three paralogs of SERCA: SERCA1, SERCA2, and SERCA3. SERCA2 is further differentiated by its spliced isoforms, SERCA2a and SERCA2b. Of particular interest is SERCA2b which is ubiquitously expressed in nervous tissue.27 SERCA2b activity is modulated by Ca2+-sensitive calreticulin and ER resident protein 57 (ERp57), both of which also function as chaperones.94,95 When Ca2+ decreases in the ER lumen, calreticulin binds to SERCA2b increasing its activity, while ERp57 functions as a thiol oxidoreductase, catalyzing disulfide bridges on the SERCA channel and consequently enhancing Ca2+ uptake. Hence, luminal Ca2+ concentration inversely determines the rate of Ca2+ uptake by SERCA such that channel activity peaks at low luminal Ca2+ concentrations and wanes at higher concentrations.27 With a drastic increase in cytosolic Ca2+ (e.g., during an action potential), other transporters with a greater capacity for Ca2+, such as mitochondrial calcium uniporter (MCU) and Na+/Ca2+ exchanger (NCX), can further support Ca2+ homeostasis.26
A loss of SERCA function has been linked to neuropathic pain after PNI. Reduced SERCA function in response to PNI leads to a slower recovery of Ca2+ transients in small diameter nociceptors.96 The inability of the ER to buffer increases in cytosolic Ca2+ after injury leads to Ca2+-signaling-induced hyperexcitability of these afferents and neuropathic pain.20,97 This is further exemplified in axons of sensory neurons obtained from diabetic rats where a loss of SERCA activity contributes to diminished Ca2+ levels in intracellular stores.98Thrombospondin-4, an extracellular matrix glycoprotein commonly elevated after nerve injury, was shown to suppress SERCA activity, enhance PMCA activity, drain ER Ca2+, and stimulate store-operated Ca2+-entry99 (Figure 3). The net result of this is a loss of intracellular Ca2+ levels. Similar observations have been made following axotomy.100
Store-operated Ca2+ entry and pain
During a Ca2+ transient, Ca2+ enters the cell through the plasma membrane and is also released from the ER. To replenish this loss of ER Ca2+ pool, store operated Ca2+ channels (SOCCs), particularly the Orai channels, pump extracellular Ca2+ into the cytosol and into the ER via the SERCA channels.101,102 The function of these SOCCs is dependent on the intraluminal ER Ca2+ concentration.103 Stromal interaction molecule (STIM) proteins were identified as Ca2+ sensors in the ER.104 There are two homologs of STIM proteins, STIM1 and STIM2 in vertebrates that are expressed in a variety of tissues and organs including nervous tissue.105 These single-pass transmembrane ER proteins are localized diffusely throughout the ER under resting conditions.106 Upon emptying of the Ca2+ store, STIM proteins redistribute and cluster on the ER membrane nearest to the plasma membrane where their CRAC activation domain (CAD; also known as STIM-Orai activation region or SOAR) binds and activates Orai channels on the plasma membrane.104,107 Orai channels also redistribute and cluster to ER–plasma membrane junction sites creating microdomains of store mediated Ca2+ entry (SOCE).106,108 There are three human homologs of Orai channels, Orai1-3, each of which share considerable homology (∼62% overall identity) with each other but have very little overlap with other ion channels.107 Orai channels are found ubiquitously in various tissues.109–111 Of note, high Orai1 expression in the brain implicates its role in the nervous system.112 Various mechanisms, such as glycosylation and pH sensing, and proteins, including golli proteins and septins, can further regulate SOCE.103 Through the formation of ER:plasma membrane microdomains, the ER—an intracellular Ca2+ store—becomes functionally linked to the extracellular supply of Ca2+. An extensive review of SOCE and its regulatory mechanisms can be found elsewhere.103,107
The role of SOCE in mediating disease and injury has only recently come to light primarily because the Orai channels were only identified and cloned at the turn of the century.113–115 Since then, SOCCs have been implicated in various disease states including autoimmunity,116 neurodegeneration,117 arthritis,118 and tumorigenesis.119 Only recently has the role of SOCCs been established in pain pathophysiology. It was noticed that intraplantar, intraperitoneal, and intrathecal administration of YM-58483, an inhibitor of SOCE, prevented both acute and chronic mechanical and thermal hypersensitivity in a dose-dependent manner following a SNI and CFA-administration as well as reducing both phases of the formalin response.120 Similarly, YM-58483 administration in a model of rheumatoid arthritis was found to reduce cytokine production in the periphery and ERK and CaMKIIα expression in the spinal cord, resulting in attenuated pain hypersensitivity.118 Although these reports demonstrate the analgesic effects of inhibiting SOCE, the molecular underpinning of this phenotype remained unknown until recently. In a series of experiments employing Orai1 knock-out animals, Dou et al.121 showed that Orai1 contributed to neuronal hyperexcitability in the spinal cord and pain behaviors following formalin and carrageenan administration. In the periphery, knockdown of STIM1, STIM2, Orai1, and Orai3 were found to diminish SOCE in DRG neurons.122 Thapsigargin, a SERCA blocker that depletes ER Ca2+ and thereby induces SOCE, promoted hyperexcitability of sensory neurons in vitro. This phenotype was abolished with Orai1 and Orai3 double knockdown.122 These observations clearly implicate SOCE in regulating pain pathophysiology. The sequence of events that lead to the imbalance of intracellular Ca2+, however, remains unclear. Is the ER Ca2+ dysregulated first and that is followed by increased SOCE or is the SOCE impaired that then induces ER Ca2+ release? The answer is probably dependent on the disease or injury. In either case, a vicious cycle of Ca2+ dyshomeostasis becomes detrimental to the cell.
Mitochondrial Ca2+ regulation and the impact on pain
Mitochondria were not considered to be major Ca2+ regulators under physiological conditions until fluorescent Ca2+ dyes made it possible to measure Ca2+ in organelles of live cells.123,124 Although they store less Ca2+ than the ER, mitochondria are nonetheless important buffers of intracellular Ca2+.125,126 Ca2+ influx into the mitochondrial matrix occurs through the MCU located on the IMM, which has high Ca2+ selectivity, but low affinity.127 The MAM overcomes this barrier by creating microdomains of high Ca2+ at the IP3R-S1R-GRP75-VDAC complex (Figure 2).50,128 In this complex, VDACs on the OMM of mitochondria are tethered by the chaperone GRP75 to IP3Rs in the ER. Thus, when IP3Rs open, and are stabilized by S1Rs, there can be efficient transport of Ca2+ into the mitochondrial matrix by the MCU.50,127,128 The VAPB-PTPIP51 complex also facilitates Ca2+ uptake by tethering the MAM.56,129
Figure 2.
The ER and the mitochondria are connected by Ca2+. Extracellular Ca2+ commonly enters sensory neurons via voltage-gated Ca2+ channels, store-operated Ca2+ channels (ORAI1/3), and transient receptor potential channels upon stimulation. This increase in cytosolic Ca2+ is rapidly neutralized by the SERCA pump and stored in the ER. Various CBPs including chaperones, like BiP, buffer ER Ca2+ levels. The ER and mitochondria are connected by the MAM, which facilitates Ca2+ transfer from the ER through IP3 receptors, to the mitochondria through the VDAC in a complex tethered by the chaperone GRP75. This exchange is regulated by the MAM complex VAPB-PTPIP51. Ca2+ subsequently enters the mitochondrial matrix through the MCU and is extruded by the NCLX. Ca2+ can also exit the mitochondria through the mPTP under pathological conditions. Ca2+ dyshomeostasis along this axis can not only disrupt ER and mitochondrial function but also affect iCa2+ and cell excitability, contributing to pain hypersensitivity.
Ca2+ regulates various mitochondrial processes (Figure 3) including ATP and ROS production, fission and fusion, opening of the mitochondrial permeability transition pore (mPTP), and mitochondria-mediated apoptosis.24,130 For example, Ca2+ can stimulate ATP production via activation of Krebs cycle dehydrogenases.131 ROS and Ca2+ have a complex relationship. Ca2+ signaling can stimulate of ROS-generating enzymes and upregulate ROS production during mitochondrial respiration.132 In addition, ROS can influence Ca2+-dependent processes. Oxidation of BK channels leads to a loss of Ca2+-sensing ability and thus a loss of activation.133 Since BK channels prevent repetitive firing, loss of their function can lead to nociceptor hyperexcitability and pain.134
Normally, mitochondrial Ca2+ concentration is maintained steadily by a balance between influx through MCUs and efflux through the mitochondrial sodium-calcium exchanger (NCLX).126,135,136 However, mitochondria can also buffer Ca2+ elevations in the cytosol by uptake and precipitation as Ca2+-phosphate.24,137 In DRG neurons, both the MCU and NCLX are critical to this process.138 Knocking down the NCLX can reduce both MCU Ca2+ uptake, and TRPV1 receptor-mediated Ca2+ currents in nociceptors. Thus, mitochondrial Ca2+ buffering is important for regulating intracellular Ca2+ homeostasis. This capacity to buffer intracellular Ca2+ can significantly influence Ca2+ transients in nociceptors, the overall level of nociceptor excitability, and ultimately, pain.139
Pathophysiology impacting the ER and mitochondria
ER stress in PNI
Along with various other homeostatic processes, ER stress has also been implicated in the pathophysiology of PNIs (Table 1). Damage to peripheral nerves induces a robust UPR in axons and soma of sensory neurons in the DRG (reviewed by Valenzuela et al.140) Interestingly, ER chaperones, like BiP, and transcription factors, like CHOP, are elevated at the site of injury following sciatic nerve crush and are retrogradely transported back to the soma as markers of injury.141 It is unlikely that UPR by-products from injured axons are the only source of ER stress markers observed in the DRG considering that ER stress markers are elevated in the DRG as early as one day after injury.142 Regardless, axonal ER stress is proposed to be beneficial and essential for nerve regeneration and locomotor recovery possibly by addressing the high demand of protein synthesis required for axonal outgrowth. In this respect, the upregulation of ERp57, an ER resident disulfide isomerase, was found to improve axonal regeneration and locomotion following sciatic nerve injury.143 Furthermore, regeneration and recovery after sciatic nerve crush depended on the activation of the more protective IRE1α-XBP1 axis as opposed to the more detrimental PERK-ATF4 pathway.41 Moreover, axotomy-induced Ca2+ release from the ER promotes IRE1 and PERK phosphorylation.144 Inhibiting PERK and IRE1 signaling using GSK2606414 and 4µ8c, respectively, collapses the growth cone, fragments axonal ER, and impairs axonal regeneration. Interestingly, axotomy-induced PERK activation does not increase CHOP levels in the axon, although increased phosphorylation of eIF2α is observed.144 Combined with increased splicing of XBP1 and BiP expression in the axons following axotomy, this observation further suggests that enhancing the ER folding capacity and reducing ER load after a PNI enhances the regenerative capacity of the nerve. However, which cytoprotective proteins are specifically folded following nerve injury remains to be investigated and will require high-throughput translatomic and proteomic screening.
Table 1.
Overview of ER pathologies.
Disease | Reference | Findings |
---|---|---|
Peripheral nerve injury pain | Castillo et al.143 | Overexpressing ERp57 improved nerve regeneration and locomotion after sciatic nerve crush. |
Ge et al.148 | Inhibiting PDI, a chaperone, in SDH after SNL reduced ER stress and mechanical pain. | |
Liu et al.147 | Intrathecal 4-PBA administration alleviated mechanical and thermal hypersensitivity in SNL rats. | |
Melemedjian et al.145 | SNL induced phosphorylation of eIF2α in the sciatic nerve | |
Ohtake et al.144 | Nerve regeneration following axotomy requires Ca2+-dependent activation of IRE1 and PERK signaling. | |
Onate et al.41 | Nerve regeneration and motor recovery was associated with IRE1-XBP1 signaling, but not PERK-ATF4 pathway, in a nerve crush model. | |
Yamaguchi et al.142 | ER stress markers are elevated following SNL. Salubrinal, an eIF2α phosphatase inhibitor, reduced pain hypersensitivity. | |
Ying et al.141 | Mild ER stress enhanced nerve regeneration after crush injury. UPR by-products are retrogradely transported from the nerve to the DRG. | |
Zhang et al.146 | SNL induced BiP and ATF6 in SDH neurons and PERK signaling in astrocytes. ATF6 knockdown in the SDH reduced BiP levels and pain behaviours in SNL rats. | |
Diabetic neuropathy | Barragan-Iglesias et al.165 | MGO triggers ISR in IB4+ sensory neurons and produces mechanical hypersensitivity. Treatment with ISRIB resolves eIF2α phosphorylation and pain in MGO administered and diabetic mice. MGO enhanced Ca2+ responses of sensory neurons. |
Griggs et al.163,164 | MGO sensitizes lamina II SDH neurons of db/db mice via the TRPA1-AC1-Epac pathway. Spinal neurons demonstrate enhanced Ca2+ responses upon stimulation. | |
Inceoglu et al.160 | STZ-induced diabetes robustly generates ER stress in the sciatic nerve and glabrous skin. 4-PBA treatment attenuated mechanical hypersensitivity in a time- and dose-dependent manner. | |
Lupachyk et al.159 | 4-PBA and TMAO attenuated diabetes-induced ER stress, nerve dysfunction, fiber loss, and oxidative stress in the sciatic nerve and the spinal cord. CHOP−/− mice were less susceptible to diabetic peripheral neuropathy. | |
Zherebitskaya et al.98 | Hypoalgesia associated with chronic STZ-induced diabetes is a result of reduced ER Ca2+ loading. | |
Chemotherapy-induced neuropathic pain | Andoh et al.40,185 | Ptx treatment promoted CHOP expression in the sciatic nerve, enhanced excitability of SDH neurons, and produced tactile hypersensitivity. |
Megat et al.182 | Ptx enhances excitability of sensory neurons | |
Resham and Sharma184 | Wnt signaling inhibitors normalized sciatic nerve function, reduced axonal BiP expression, and attenuated pain hypersensitivity in Ptx treated rats. | |
Tanimukai et al.186,187 | In vitro, Ptx induced CHOP and caspase 3 expression in SK-N-SH neuroblastoma cells. Inducing sigma 1 receptor and BiP reduced ER stress and was cytoprotective. |
Only recently has ER stress been linked to pain in PNI models. Melemedjian et al.145 was one of the first studies to correlate increases in eIF2α phosphorylation in injured nerves of rodents following SNL; however, no mechanistic role of the ISR in inducing mechanical pain was investigated. Another study found elevated BiP, XBP1, p-eIF2α, and ATF6 expression in the dorsal horn of the spinal cord two weeks after SNL surgery which correlated with microglial activation and mechanical hypersensitivity in these animals.146 Furthermore, knockdown of ATF6 using an intrathecal siRNA reduced neuronal BiP expression and partially reversed mechanical hypersensitivity. Intrathecal treatment of chemical chaperones, 4-PBA and tauroursodeoxycholic acid (TUDCA), was also found to partially reverse nociceptive behaviours after SNL.147,148 In the periphery, Yamaguchi et al.142 found that BiP, XBP1, and CHOP expression was elevated in the sensory neurons as early as one day post-SNL only to recover within a week. Interestingly, treatment with salubrinal, an eIF2α phosphatase inhibitor, reduced mechanical, thermal, and cold hypersensitivity142 suggesting that induction of the ISR may ameliorate pain possibly by reducing global translation, enhancing ER folding capacity, and thereby dampening ER load.
As an oxidoreductase and a chaperone, protein disulfide isomerase (PDI) reduces disulfide bonds and produces H2O2 as a by-product. Along with other chaperones, SNL induces PDI expression in the spinal cord. Inhibiting PDI using bacitracin, an antibiotic, reduces ER stress, mitochondrial SOD levels, and mechanical hypersensitivity associated with nerve injury.148 This suggests that SNL-induced pain may be a result of ER–mitochondria cross-talk.
Mitochondria in PNI
Mitochondrial dysfunction is evident in both peripheral and central tissues in PNI models (Table 2). Mitochondria after PNI exhibit fragmentation, depolarized membrane potentials, reduced ATP synthesis, and oxidative stress.149 Mitochondrial dysfunctions can trigger apoptosis and axonal degeneration in the periphery and spinal cord, leading to pain.149–152 However, neuropathic pain in PNI is not exclusively caused by degeneration. Both peripheral and central sensitization contribute to pain maintenance.153 Elevated Ca2+ signaling underlies nociceptor hyperexcitability in other neuropathic pain conditions.97 Interestingly, mitochondrial dysfunction observed in PNI models may also contribute to iCa2+ dyshomeostasis, but the relationship is likely complicated. Mitochondria both regulate and are regulated by Ca2+,24 so, while Ca2+ elevations can overwhelm mitochondrial buffering and lead to dysfunction, mitochondrial dysfunction can also be a starting point for pathological iCa2+ elevation. This complex association presents a significant target for future study to probe into how mitochondrial dysfunction might contribute to PNI-induced neuropathic pain.
Table 2.
Overview of mitochondrial pathologies.
Disease | Study | Finding |
---|---|---|
Peripheral nerve injury | Bae et al.197 | Mitochondrial superoxide in the spinal dorsal horn of mice after PNI contributes to sensitization. |
Englezou et al.152 | DRG neurons exposed to H2O2 in vitro display mitochondrial fission and eventual mitochondria-mediated apoptosis, which may give insight into how peripheral neurons are lost after PNI. | |
Graham et al.198 | PNI in mice results in increased mitochondrial fission and mitophagic protein expression in whole muscle tissue. | |
Kim et al.151 | Inhibiting mPTP opening in PNI with cyclosporin A attenuates allodynia and spinal cord cytochrome c release. | |
Wang et al.150 | Pro-BDNF induces mitochondria-mediated apoptosis of neurons and satellite glia in vitro | |
Diabetic neuropathy | Hall et al.199 | DRG neurons from diabetic rats showed increased voltage-dependent Ca2+ currents. |
Kostyuk et al.175 | Prolonged cytosolic Ca transients in cells from diabetic mice are dependent on mitochondrial dysfunction, and may contribute to neuropathic pain. | |
Russell et al.200 | Hyperglycemia induces ROS production and mitochondrial-mediated apoptosis in vitro. | |
Srinivasan et al.168 | DRG neurons from diabetic rats in vitro have increased apoptosis due to mitochondrial dysfunction. | |
Vincent et al.170 | An imbalance in mitochondrial biogenesis and fission results from hyperglycemia in DRG neurons in vitro and may contribute to diabetic neuropathy via neuronal injury. | |
Chemotherapy-induced neuropathic pain | Duggett et al.188 | Rats treated with paclitaxel display energy dysfunction in the DRG that correlates with pain. |
Flatters and Bennett et al.176 | In rats, paclitaxel-induced mitochondrial abnormalities may be responsible for pain, as their timelines correlate. | |
Jia et al.189 | Paclitaxel treatment in rats induces mitochondrial damage and production of reaction oxygen species that may activate the NLRP3 inflammasome and contribute to pain. | |
Kidd et al.193 | Paclitaxel-induced intracellular Ca2+ elevations can be inhibited with cyclosporin A, suggesting that paclitaxel may cause pain by inducing mPTP-opening and aberrant Ca release. | |
Xiao and Bennett194 | Spontaneous discharge of sensory neurons in rats treated with paclitaxel is likely a factor in paclitaxel-induced peripheral neuropathy. |
ER stress in diabetic neuropathy
Diabetes mellitus (DM) is a metabolic disorder characterized by abnormally high blood glucose levels. The cause of DM is linked to either reduced insulin production (type 1) or a resistance to insulin (type 2).154,155 Peripheral neuropathy and subsequent neuropathic pain is a common phenomenon in DM (Table 1).156 Initially, patients with DM and preclinical rodent models of DM typically present with mechanical and thermal hypersensitivity.157 Later disease stages are associated with hyposensitivity as a result of sensory fiber retractions from the skin.158 Pain in DM follows a “glove and stocking” pattern in which distal limbs are affected first, followed by more proximal areas of the body.156 This section will highlight mechanisms involved in neuropathic pain associated with DM, rather than loss of sensation, that have been linked to impaired ER function, Ca2+ dyshomeostasis, and mitochondrial damage.
Several studies have directly investigated the role of ER stress in mediating diabetes-induced pain (Table 1). ER stress in the streptozotocin (STZ) model of type 1 DM is characterized by increased phosphorylation of PERK, eIF2α, and IRE1α, as well as the cleavage of ATF6 in the sciatic nerve and the glabrous skin of the paw.159,160 Other markers of ER stress such as BiP and spliced XBP1 are also known to be upregulated with DM.160 The spinal cord is also known to increase CHOP and BiP expression in diabetic rats.159 Alleviating UPR/ER stress using chemical chaperones, 4-phenylbutyric acid (4-PBA) and trimethylamine oxide (TMAO), reverses tactile allodynia and heat hypoalgesia in rats.159,160 Denervation associated with late-stage DM is also reversed in rodents administered with 4-PBA and TMAO.159STZ-induced DM in transgenic CHOP-deficient mice have reduced nerve fiber loss from the glabrous skin as compared to STZ-induced DM in wild-type mice.159CHOP-knockout mice experience minimal thermal hypoalgesia as compared to wild-types in response to STZ-induced diabetes. However, CHOP−/− does not confer protection against mechanical and tactile hypersensitivity.159 Collectively, these studies suggest that diabetes-induced ER stress contributes to neuropathic pain.
Methylglyoxal (MGO), a common metabolite present in high concentrations (up to 5 µM) in the plasma of diabetic patients, is known to induce heat, mechanical, and affective pain in DM by activating peripheral and spinal TRPA1 channels.161–166 Recently, the involvement of the UPR in MGO-induced pain hypersensitivity was postulated.165 Intraplantar injection of MGO induces rapid and transient mechanical hypersensitivity in rats and mice that is associated with increased phosphorylation of PERK and eIF2α, particularly in the IB4+ nociceptors.165 Preventing the phosphorylation of eIF2α using the ISR inhibitor, ISRIB, attenuated mechanical pain in both MGO-induced neuropathy and STZ-induced DM.165 Systemic 4-PBA treatment was also able to ameliorate MGO-induced mechanical hypersensitivity.165 Hence, MGO-associated ER stress and ISR in IB4+ nonpeptidergic nociceptors, may contribute to mechanical hypersensitivity observed in type 1 DM. Furthermore, acute MGO treatment in vitro enhances KCl-induced cytosolic Ca2+ transients in dissociated mouse DRG neurons.165 These studies propose that rises in MGO in diabetes can enhance intracellular Ca2+ signaling in painful diabetic neuropathy.
In contrast, axons of sensory neurons from hypoalgesic type 1 STZ-induced diabetic rats have dampened Ca2+ responses to KCl and caffeine stimulation.98 This suggests that Ca2+ stores in the ER are likely depleted in DM as a result of reduced SERCA-mediated Ca2+ uptake.98 The discrepancy between these studies may stem predominantly from the differences in their experimental paradigms (acute vs. chronic), disease model (type 1 vs. type 2 DM), and the presence of hyperalgesia. Perhaps, increased Ca2+ signaling contributes to the initial hyperalgesia in DM-induced neuropathy, while at later stages, diabetic neuropathy is characterized by reduced intracellular and intra-ER Ca2+. Both, the increase in and the loss of intracellular Ca2+ contribute to detrimental processes within the cell including excitotoxicity,69 ER stress,26 mitochondrial dysfunction (see below), and cell death.69
Mitochondria in diabetic neuropathy
Numerous studies have revealed mitochondrial dysfunction in models of DM (Table 2). This includes increased fission, ΔΨm depolarization, reduced ATP production, disrupted calcium homeostasis, and oxidative stress.97,167–170 While mitochondria dysfunction plays a role in diabetic neuropathy, both the pathways that lead to this dysfunction, and the mechanism by which it induces neuropathy are contested. The two hallmark factors of type 1 DM and its relevant models are a lack of insulin, and resultant hyperglycemia. Whether only one or both of these factors is responsible for mitochondrial dysfunction in DM is questioned in the literature.171–173 Several competing theories have been put forward to explain how mitochondrial dysfunction may be implicated in diabetic neuropathy. Hyperglycemia may increase mitochondrial fission, ROS production, and mitochondrial DNA damage, leading to neuropathy through apoptosis.171,174 Although these mechanisms would explain sensory fiber retraction, this model does not link mitochondrial dysfunction to nociceptor hyperexcitability. Alternatively, impaired mitochondrial ATP production and reduced mitochondrial membrane potential could arise in response to reduced insulin receptor stimulation. This would significantly effect Ca2+ homeostasis and trigger ER stress, leading to changes in transcriptional and translational programs of the cell.172 Considering that increased intracellular Ca2+ has been linked to nociceptor hyper-excitability and neuropathic pain in diabetic neuropathy, impaired ER–mitochondrial Ca2+ homeostasis in diabetic neuropathy is a promising potential mechanism to explain the aberrant nociceptor activity and pain associated with the disease.97,175
ER stress in chemotherapy-induced peripheral neuropathy
Neuropathy associated with chemotherapy agents is the single largest reason for reducing and halting treatment of cancer.176 Approximately 68% of patients receiving chemotherapy will develop chemotherapy-induced peripheral neuropathy (CIPN) in their first month of treatment.177 CIPN affects the nerves in a “glove and stocking” pattern where the longest axons are affected first, similar to DM-induced neuropathy.178 Unlike DM-induced neuropathy, however, CIPN symptoms appear suddenly, affect multiple areas simultaneously, and progresses rapidly.178 As a result of the CIPN, patients can experience acute and chronic sensory, motor, and autonomic dysfunction.179 Sensory impairments typically involve mechanical, thermal, and cold pain hypersensitivity as well as paresthesias and dysesthesias.179 Chronic use of chemotherapeutic agents may lead to permanent damage of the nerves, resulting in a loss of sensation.178 Platinum derivatives (e.g., oxaliplatin), vinca alkaloids (e.g., vincristine), and taxanes (e.g., paclitaxel, Ptx) are commonly used chemotherapy agents known to cause CIPN and the associated pain phenotype.178,180
Treatment with Ptx and oxaliplatin usually accompanies an acute pain syndrome three to four days after the start of the treatment as well as chronic neuropathic pain even after treatment has stopped.181 It is well established that chemotherapeutics contribute to neuronal injury and inflammation, especially in the peripheral nervous system (Table 1).181 Sensory neurons of the DRG from mice treated with paclitaxel fire spontaneously and become hyperexcitable, both of which are evidence of painful neuropathy.182 The protective and detrimental effects of ER stress and the ISR have previously been described in various cancer models (reviewed by Nam and Jeon183) However, the effects of ER dysfunction in the context of pain in CIPN are not well understood. A recent report found that intraperitoneal Ptx (2 mg/kg) administration reduced sensory nerve conduction velocity and increased pain behaviors in rats.184 Resham and Sharma184 further observed that heat, cold, and mechanical hypersensitivity was correlated with increased expression of BiP in the sciatic nerves of Ptx treated animals.184 Another study found that a single intraperitoneal injection of Ptx (5 mg/kg) was sufficient to induce CHOP expression in the sciatic nerve, increase the excitability of spinal dorsal horn neurons, and lead to mechanical allodynia in C57BL/6 NCr mice.40,185 In vitro, Ptx treatment of SK-N-SH neuroblastoma cells increased the phosphorylation of eIF2α and induced the expression of CHOP as well as the pro-apoptotic caspase 3186,187 Furthermore, pretreatment with fluvoxamine, a Sigma 1 receptor inducer, and BiP-inducer X (BIX) attenuated ER stress and protected the SK-N-SH cells from paclitaxel-associated cytotoxicity.187 However, whether these drugs can mitigate pain in CIPN remains to be investigated.
Mitochondria in chemotherapy-induced peripheral neuropathy
Mitochondrial dysfunction is a well-characterized feature of chemotherapy-induced peripheral neuropathy (Table 2). The drug paclitaxel, for example, affects both mitochondrial structure and function in sensory neurons. Some mitochondrial changes linked to Ptx include swelling, vacuolation, decreased respiration, reduced ATP production, loss of ΔΨm, mitochondrial permeability transition pore (mPTP) formation, ROS production, and Ca2+ release.176,188–190 These dysfunctions can be accounted for by the theory that Ptx interacts with the mitochondrial membrane to induce mPTP-opening.191 Opening of the mPTP allows for aberrant passage of molecules into and out of the mitochondria, which can alter oxidative phosphorylation and ATP production, as well as trigger cell death cascades.192 In addition, in a nerve-injury model, treatment with the mPTP blocker cyclosporin A not only prevented mitochondrial dysfunction, but also reduced pain.151,193 Taken together, this evidence suggests that Ptx-induced mPTP-opening and subsequent Ca2+ release may underlie nociceptor hyperexcitability and neuropathic pain.176,194
Conclusions and future directions
There is a new appreciation for the roles of the ER and mitochondria in mediating chronic pain. What is emerging from these studies is a recognition of the complex interplay between these two organelles and the impact that disturbances in this interplay may have on Ca2+ signaling and ultimately, nociceptor excitability. Although the examples discussed here generally involve some form of axonal injury or trauma, emerging research also suggests that chronic pain associated with inflammatory diseases such as Multiple Sclerosis and rheumatoid arthritis may also be linked to ER and mitochondria dysfunction.195,196 In all of these pathologies, it is clear that pain is a major overlapping symptom. A common thread linking pain to these conditions is stress at the ER and the ensuing mitochondrial dysfunction and Ca2+ dyshomeostasis. This has opened an exciting new avenue for research that is leading toward a more comprehensive understanding of the underlying mechanisms driving chronic, neuropathic pain in these states. A better understanding of how disturbed ER–mitochondrial interactions can lead to nociceptor hyperexcitability may provide insight into novel, therapeutic targets that can be manipulated to reverse the changes that have sensitized the system and thus restore the homeostatic state.
Declaration of Conflicting Interests
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding
The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: MSY was supported by the Alexander Graham Bell Canada Graduate Scholarship from NSERC. This work was supported by a grant from the Canadian Institutes of Health Research (CIHR): FRN-162434.
ORCID iD
Muhammad Saad Yousuf https://orcid.org/0000-0003-0829-8182
References
- 1.Nicholas M, Vlaeyen JWS, Rief W, Barke A, Aziz Q, Benoliel R, Cohen M, Evers S, Giamberardino MA, Goebel A, Korwisi B, Perrot S, Svensson P, Wang S-J, Treede R-D; IASP Taskforce for the Classification of Chronic Pain. The IASP classification of chronic pain for ICD-11: chronic primary pain. Pain 2019; 160: 28–37. [DOI] [PubMed] [Google Scholar]
- 2.Calvo M, Bennett DL. The mechanisms of microgliosis and pain following peripheral nerve injury. Exp Neurol 2012; 234: 271–282. [DOI] [PubMed] [Google Scholar]
- 3.Sarzi-Puttini P, Salaffi F, Di Franco M, Bazzichi L, Cassisi G, Casale R, Cazzola M, Stisi S, Battellino M, Atzeni F. Pain in rheumatoid arthritis: a critical review. Reumatismo 2014; 66: 18–27. [DOI] [PubMed] [Google Scholar]
- 4.Yousuf M, Ali L. A study of 22 cases of tropical splenomegaly and chloroquine therapy. J Coll Physicians Surg Pak 1997; 7. [Google Scholar]
- 5.Debono DJ, Hoeksema LJ, Hobbs RD. Caring for patients with chronic pain: pearls and pitfalls. J Am Osteopath Assoc 2013; 113: 620–627. [DOI] [PubMed] [Google Scholar]
- 6.Sewell R. Neuropathic pain models and outcome measures: a dual translational challenge. Ann Transl Med 2018; 6: S42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Gregory NS, Harris AL, Robinson CR, Dougherty PM, Fuchs PN, Sluka KA. An overview of animal models of pain: disease models and outcome measures. J Pain 2013; 14: 1255–1269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Biessels GJ, Bril V, Calcutt NA, Cameron NE, Cotter MA, Dobrowsky R, Feldman EL, Fernyhough P, Jakobsen J, Malik RA, Mizisin AP, Oates PJ, Obrosova IG, Pop-Busui R, Russell JW, Sima AA, Stevens MJ, Schmidt RE, Tesfaye S, Veves A, Vinik AI, Wright DE, Yagihashi S, Yorek MA, Ziegler D, Zochodne DW. Phenotyping animal models of diabetic neuropathy: a consensus statement of the diabetic neuropathy study group of the EASD (Neurodiab). J Peripher Nerv Syst 2014; 19: 77–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Shahid M, Subhan F, Ahmad N, Sewell R. The flavonoid 6-methoxyflavone allays cisplatin-induced neuropathic allodynia and hypoalgesia. Biomed Pharmacother 2017; 95: 1725–1733. [DOI] [PubMed] [Google Scholar]
- 10.Murnion BP. Neuropathic pain: current definition and review of drug treatment. Aust Prescr 2018; 41: 60–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Sorge RE, Mapplebeck JC, Rosen S, Beggs S, Taves S, Alexander JK, Martin LJ, Austin JS, Sotocinal SG, Chen D, Yang M, Shi XQ, Huang H, Pillon NJ, Bilan PJ, Tu Y, Klip A, Ji RR, Zhang J, Salter MW, Mogil JS. Different immune cells mediate mechanical pain hypersensitivity in male and female mice. Nat Neurosci 2015; 18: 1081–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Woolf CJ. Evidence for a central component of post-injury pain hypersensitivity. Nature 1983; 306: 686–688. [DOI] [PubMed] [Google Scholar]
- 13.Tracey WD., Jr. Nociception. Curr Biol 2017; 27: R129–R133. [DOI] [PubMed] [Google Scholar]
- 14.St John Smith E. Advances in understanding nociception and neuropathic pain. J Neurol 2018; 265: 231–238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Basbaum AI, Bautista DM, Scherrer G, Julius D. Cellular and molecular mechanisms of pain. Cell 2009; 139: 267–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Aley KO, Levine JD. Role of protein kinase A in the maintenance of inflammatory pain. J Neurosci 1999; 19: 2181–2186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Kandasamy R, Price TJ. The pharmacology of nociceptor priming. Handb Exp Pharmacol 2015; 227: 15–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Woller SA, Eddinger KA, Corr M, Yaksh TL. An overview of pathways encoding nociception. Clin Exp Rheumatol 2017; 35: 40–46. [PMC free article] [PubMed] [Google Scholar]
- 19.Woolf CJ, Ma Q. Nociceptors–noxious stimulus detectors. Neuron 2007; 55: 353–364. [DOI] [PubMed] [Google Scholar]
- 20.Hucho T, Levine JD. Signaling pathways in sensitization: toward a nociceptor cell biology. Neuron 2007; 55: 365–376. [DOI] [PubMed] [Google Scholar]
- 21.Chaban VV. Peripheral sensitization of sensory neurons. Ethn Dis 2010; 20: S1. [PMC free article] [PubMed] [Google Scholar]
- 22.Pizzino G, Irrera N, Cucinotta M, Pallio G, Mannino F, Arcoraci V, Squadrito F, Altavilla D, Bitto A. Oxidative stress: harms and benefits for human health. Oxid Med Cell Longev 2017; 2017: 8416763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Salvemini D, Little JW, Doyle T, Neumann WL. Roles of reactive oxygen and nitrogen species in pain. Free Radic Biol Med 2011; 51: 951–966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Bravo-Sagua R, Parra V, López-Crisosto C, Díaz P, Quest AFG, Lavandero S. Calcium transport and signaling in mitochondria. Compr Physiol 2017; 7: 623–634. [DOI] [PubMed] [Google Scholar]
- 25.Bourinet E, Altier C, Hildebrand ME, Trang T, Salter MW, Zamponi GW. Calcium-permeable ion channels in pain signaling. Physiol Rev 2014; 94: 81–140. [DOI] [PubMed] [Google Scholar]
- 26.Krebs J, Agellon LB, Michalak M. Ca(2+) homeostasis and endoplasmic reticulum (ER) stress: an integrated view of calcium signaling. Biochem Biophys Res Commun 2015; 460: 114–121. [DOI] [PubMed] [Google Scholar]
- 27.Verkhratsky A. Physiology and pathophysiology of the calcium store in the endoplasmic reticulum of neurons. Physiol Rev 2005; 85: 201–279. [DOI] [PubMed] [Google Scholar]
- 28.Terasaki M, Slater NT, Fein A, Schmidek A, Reese TS. Continuous network of endoplasmic reticulum in cerebellar Purkinje neurons. Proc Natl Acad Sci USA 1994; 91: 7510–7514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Hetz C, Mollereau B. Disturbance of endoplasmic reticulum proteostasis in neurodegenerative diseases. Nat Rev Neurosci 2014; 15: 233–249. [DOI] [PubMed] [Google Scholar]
- 30.Stevenson J, Huang EY, Olzmann JA. Endoplasmic reticulum-associated degradation and lipid homeostasis. Annu Rev Nutr 2016; 36: 511–542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Jacquemyn J, Cascalho A, Goodchild RE. The ins and outs of endoplasmic reticulum-controlled lipid biosynthesis. EMBO Rep 2017; 18: 1905–1921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Suzuki CK, Bonifacino JS, Lin AY, Davis MM, Klausner RD. Regulating the retention of T-cell receptor alpha chain variants within the endoplasmic reticulum: Ca(2+)-dependent association with BiP. J Cell Biol 1991; 114: 189–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hetz C, Axten JM, Patterson JB. Pharmacological targeting of the unfolded protein response for disease intervention. Nat Chem Biol 2019; 15: 764–775. [DOI] [PubMed] [Google Scholar]
- 34.Martinez G, Khatiwada S, Costa-Mattioli M, Hetz C. ER proteostasis control of neuronal physiology and synaptic function. Trends Neurosci 2018; 41: 610–624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.McQuiston A, Diehl JA. Recent insights into PERK-dependent signaling from the stressed endoplasmic reticulum. F1000Res 2017; 6: 1897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Carrara M, Prischi F, Nowak PR, Ali MM. Crystal structures reveal transient PERK luminal domain tetramerization in endoplasmic reticulum stress signaling. Embo J 2015; 34: 1589–1600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ma K, Vattem KM, Wek RC. Dimerization and release of molecular chaperone inhibition facilitate activation of eukaryotic initiation factor-2 kinase in response to endoplasmic reticulum stress. J Biol Chem 2002; 277: 18728–18735. [DOI] [PubMed] [Google Scholar]
- 38.Khoutorsky A, Price TJ. Translational control mechanisms in persistent pain. Trends Neurosci 2018; 41: 100–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Pakos-Zebrucka K, Koryga I, Mnich K, Ljujic M, Samali A, Gorman AM. The integrated stress response. EMBO Rep 2016; 17: 1374–1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Andoh T, Uta D, Kato M, Toume K, Komatsu K, Kuraishi Y. Prophylactic administration of aucubin inhibits paclitaxel-induced mechanical allodynia via the inhibition of endoplasmic reticulum stress in peripheral Schwann cells. Biol Pharm Bull 2017; 40: 473–478. [DOI] [PubMed] [Google Scholar]
- 41.Onate M, Catenaccio A, Martinez G, Armentano D, Parsons G, Kerr B, Hetz C, Court FA. Activation of the unfolded protein response promotes axonal regeneration after peripheral nerve injury. Sci Rep 2016; 6: 21709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Gray MW. Mitochondrial evolution. Cold Spring Harb Perspect Biol 2012; 4: a011403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Friedman JR, Nunnari J. Mitochondrial form and function. Nature 2014; 505: 335–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Zorova LD, Popkov VA, Plotnikov EY, Silachev DN, Pevzner IB, Jankauskas SS, Babenko VA, Zorov SD, Balakireva AV, Juhaszova M, Sollott SJ, Zorov DB. Mitochondrial membrane potential. Anal Biochem 2018; 552: 50–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Brieger K, Schiavone S, Miller FJ, Jr, Krause KH. Reactive oxygen species: from health to disease. Swiss Med Wkly 2012; 142: w13659. [DOI] [PubMed] [Google Scholar]
- 46.McBride HM, Neuspiel M, Wasiak S. Mitochondria: more than just a powerhouse. Curr Biol 2006; 16: R551–R560. [DOI] [PubMed] [Google Scholar]
- 47.Disatnik MH, Hwang S, Ferreira JC, Mochly-Rosen D. New therapeutics to modulate mitochondrial dynamics and mitophagy in cardiac diseases. J Mol Med (Med ) 2015; 93: 279–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Hoitzing H, Johnston IG, Jones NS. What is the function of mitochondrial networks? A theoretical assessment of hypotheses and proposal for future research. Bioessays 2015; 37: 687–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Szabadkai G, Simoni AM, Chami M, Wieckowski MR, Youle RJ, Rizzuto R. Drp-1-dependent division of the mitochondrial network blocks intraorganellar Ca2+ waves and protects against Ca2+-mediated apoptosis. Mol Cell 2004; 16: 59–68. [DOI] [PubMed] [Google Scholar]
- 50.Veeresh P, Kaur H, Sarmah D, Mounica L, Verma G, Kotian V, Kesharwani R, Kalia K, Borah A, Wang X, Dave KR, Rodriguez AM, Yavagal DR, Bhattacharya P. Endoplasmic reticulum-mitochondria crosstalk: from junction to function across neurological disorders. Ann N Y Acad Sci 2019; 1457: 41–60. [DOI] [PubMed] [Google Scholar]
- 51.Vance JE. MAM (mitochondria-associated membranes) in mammalian cells: lipids and beyond. Biochim Biophys Acta 2014; 1841: 595–609. [DOI] [PubMed] [Google Scholar]
- 52.Kojima R, Endo T, Tamura Y. A phospholipid transfer function of ER-mitochondria encounter structure revealed in vitro. Sci Rep 2016; 6: 30777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Simmen T, Aslan JE, Blagoveshchenskaya AD, Thomas L, Wan L, Xiang Y, Feliciangeli SF, Hung CH, Crump CM, Thomas G. PACS-2 controls endoplasmic reticulum-mitochondria communication and Bid-mediated apoptosis. Embo J 2005; 24: 717–729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Filadi R, Greotti E, Turacchio G, Luini A, Pozzan T, Pizzo P. Presenilin 2 modulates endoplasmic reticulum-mitochondria coupling by tuning the antagonistic effect of mitofusin 2. Cell Rep 2016; 15: 2226–2238. [DOI] [PubMed] [Google Scholar]
- 55.Raturi A, Simmen T. Where the endoplasmic reticulum and the mitochondrion tie the knot: the mitochondria-associated membrane (MAM). Biochim Biophys Acta 2013; 1833: 213–224. [DOI] [PubMed] [Google Scholar]
- 56.De Vos KJ, Morotz GM, Stoica R, Tudor EL, Lau KF, Ackerley S, Warley A, Shaw CE, Miller CC. VAPB interacts with the mitochondrial protein PTPIP51 to regulate calcium homeostasis. Hum Mol Genet 2012; 21: 1299–1311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Pinton P. Mitochondria-associated membranes (MAMs) and pathologies. Cell Death Dis 2018; 9: 413–018/03/20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Missiroli S, Patergnani S, Caroccia N, Pedriali G, Perrone M, Previati M, Wieckowski MR, Giorgi C. Mitochondria-associated membranes (MAMs) and inflammation. Cell Death Dis 2018; 9: 329–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Gleichmann M, Mattson MP. Neuronal calcium homeostasis and dysregulation. Antioxid Redox Signal 2011; 14: 1261–1273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.van Vliet AR, Agostinis P. Mitochondria-associated membranes and ER stress. Curr Top Microbiol Immunol 2018; 414: 73–102. [DOI] [PubMed] [Google Scholar]
- 61.Bravo R, Vicencio JM, Parra V, Troncoso R, Munoz JP, Bui M, Quiroga C, Rodriguez AE, Verdejo HE, Ferreira J, Iglewski M, Chiong M, Simmen T, Zorzano A, Hill JA, Rothermel BA, Szabadkai G, Lavandero S. Increased ER-mitochondrial coupling promotes mitochondrial respiration and bioenergetics during early phases of ER stress. J Cell Sci 2011; 124: 2143–2152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Carreras-Sureda A, Jaña F, Urra H, Durand S, Mortenson DE, Sagredo A, Bustos G, Hazari Y, Ramos-Fernández E, Sassano ML, Pihán P, van Vliet AR, González-Quiroz M, Torres AK, Tapia-Rojas C, Kerkhofs M, Vicente R, Kaufman RJ, Inestrosa NC, Gonzalez-Billault C, Wiseman RL, Agostinis P, Bultynck G, Court FA, Kroemer G, Cárdenas JC, Hetz C. Non-canonical function of IRE1alpha determines mitochondria-associated endoplasmic reticulum composition to control calcium transfer and bioenergetics. Nat Cell Biol 2019; 21: 755–767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Verfaillie T, Rubio N, Garg AD, Bultynck G, Rizzuto R, Decuypere JP, Piette J, Linehan C, Gupta S, Samali A, Agostinis P. PERK is required at the ER-mitochondrial contact sites to convey apoptosis after ROS-based ER stress. Cell Death Differ 2012; 19: 1880–1891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.de la Puente B, Nadal X, Portillo-Salido E, Sánchez-Arroyos R, Ovalle S, Palacios G, Muro A, Romero L, Entrena JM, Baeyens JM, López-García JA, Maldonado R, Zamanillo D, Vela JM. Sigma-1 receptors regulate activity-induced spinal sensitization and neuropathic pain after peripheral nerve injury. Pain 2009; 145: 294–303. [DOI] [PubMed] [Google Scholar]
- 65.Romero L, Zamanillo D, Nadal X, Sánchez-Arroyos R, Rivera-Arconada I, Dordal A, Montero A, Muro A, Bura A, Segalés C, Laloya M, Hernández E, Portillo-Salido E, Escriche M, Codony X, Encina G, Burgueño J, Merlos M, Baeyens JM, Giraldo J, López-García JA, Maldonado R, Plata-Salamán CR, Vela JM. Pharmacological properties of S1RA, a new sigma-1 receptor antagonist that inhibits neuropathic pain and activity-induced spinal sensitization. Br J Pharmacol 2012; 166: 2289–2306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Romero L, Merlos M, Vela JM. Antinociception by sigma-1 receptor antagonists: central and peripheral effects. Adv Pharmacol 2016; 75: 179–215. [DOI] [PubMed] [Google Scholar]
- 67.Bruna J, Velasco R. Sigma-1 receptor: a new player in neuroprotection against chemotherapy-induced peripheral neuropathy. Neural Regen Res 2018; 13: 775–778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Bruna J, Videla S, Argyriou AA, Velasco R, Villoria J, Santos C, Nadal C, Cavaletti G, Alberti P, Briani C, Kalofonos HP, Cortinovis D, Sust M, Vaqué A, Klein T, Plata-Salamán C. Efficacy of a novel sigma-1 receptor antagonist for oxaliplatin-induced neuropathy: a randomized, double-blind, placebo-controlled phase IIa clinical trial. Neurotherapeutics 2018; 15: 178–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Bano D, Ankarcrona M. Beyond the critical point: an overview of excitotoxicity, calcium overload and the downstream consequences. Neurosci Lett 2018; 663: 79–85. [DOI] [PubMed] [Google Scholar]
- 70.Nakamura K, Zuppini A, Arnaudeau S, Lynch J, Ahsan I, Krause R, Papp S, De Smedt H, Parys JB, Müller-Esterl W, Lew DP, Krause K-H, Demaurex N, Opas M, Michalak M. Functional specialization of calreticulin domains. J Cell Biol 2001; 154: 961–972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Solovyova N, Verkhratsky A. Monitoring of free calcium in the neuronal endoplasmic reticulum: an overview of modern approaches. J Neurosci Methods 2002; 122: 1–12. [DOI] [PubMed] [Google Scholar]
- 72.Inui M, Saito A, Fleischer S. Purification of the ryanodine receptor and identity with feet structures of junctional terminal cisternae of sarcoplasmic reticulum from fast skeletal muscle. J Biol Chem 1987; 262: 1740–1747. [PubMed] [Google Scholar]
- 73.Zalk R, Clarke OB, Des Georges A, Grassucci RA, Reiken S, Mancia F, Hendrickson WA, Frank J, Marks AR. Structure of a mammalian ryanodine receptor. Nature 2015; 517: 44–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Chen SR, Li X, Ebisawa K, Zhang L. Functional characterization of the recombinant type 3 Ca2+ release channel (ryanodine receptor) expressed in HEK293 cells. J Biol Chem 1997; 272: 24234–24246. [DOI] [PubMed] [Google Scholar]
- 75.Murayama T, Ogawa Y. Characterization of type 3 ryanodine receptor (RyR3) of sarcoplasmic reticulum from rabbit skeletal muscles. J Biol Chem 1997; 272: 24030–24037. [DOI] [PubMed] [Google Scholar]
- 76.Murayama T, Ogawa Y. Properties of Ryr3 ryanodine receptor isoform in mammalian brain. J Biol Chem 1996; 271: 5079–5084. [DOI] [PubMed] [Google Scholar]
- 77.Rossi D, Simeoni I, Micheli M, Bootman M, Lipp P, Allen PD, Sorrentino V. RyR1 and RyR3 isoforms provide distinct intracellular Ca2+ signals in HEK 293 cells. J Cell Sci 2002; 115: 2497–2504. [DOI] [PubMed] [Google Scholar]
- 78.Gillespie D, Chen H, Fill M. Is ryanodine receptor a calcium or magnesium channel? Roles of K+ and Mg2+ during Ca2+ release. Cell Calcium 2012; 51: 427–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Van Petegem F. Ryanodine receptors: allosteric ion channel giants. J Mol Biol 2015; 427: 31–53. [DOI] [PubMed] [Google Scholar]
- 80.Tully K, Treistman SN. Distinct intracellular calcium profiles following influx through N- versus L-type calcium channels: role of Ca2+-induced Ca2+ release. J Neurophysiol 2004; 92: 135–143. [DOI] [PubMed] [Google Scholar]
- 81.Irie T, Trussell LO. Double-nanodomain coupling of calcium channels, ryanodine receptors, and BK channels controls the generation of burst firing. Neuron 2017; 96: 856–870 e854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Rosenberg H, Davis M, James D, Pollock N, Stowell K. Malignant hyperthermia. Orphanet J Rare Dis 2007; 2: 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Del Prete D, Checler F, Chami M. Ryanodine receptors: physiological function and deregulation in Alzheimer disease. Mol Neurodegener 2014; 9: 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Ferrari LF, Khomula EV, Araldi D, Levine JD. Marked sexual dimorphism in the role of the ryanodine receptor in a model of pain chronification in the rat. Sci Rep 2016; 6: 31221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Vanderheyden V, Devogelaere B, Missiaen L, De Smedt H, Bultynck G, Parys JB. Regulation of inositol 1,4,5-trisphosphate-induced Ca2+ release by reversible phosphorylation and dephosphorylation. Biochim Biophys Acta 2009; 1793: 959–970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Taylor CW, Tovey SC. IP(3) receptors: toward understanding their activation. Cold Spring Harb Perspect Biol 2010; 2: a004010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Bezprozvanny I, Watras J, Ehrlich BE. Bell-shaped calcium-response curves of Ins(1,4,5)P3- and calcium-gated channels from endoplasmic reticulum of cerebellum. Nature 1991; 351: 751–754. [DOI] [PubMed] [Google Scholar]
- 88.Hagar RE, Burgstahler AD, Nathanson MH, Ehrlich BE. Type III InsP3 receptor channel stays open in the presence of increased calcium. Nature 1998; 396: 81–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Bouhadfane M, Kaszas A, Rozsa B, Harris-Warrick RM, Vinay L, Brocard F. Sensitization of neonatal rat lumbar motoneuron by the inflammatory pain mediator bradykinin. Elife 2015; 4: e06195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Choi SI, Hwang SW. Depolarizing effectors of bradykinin signaling in nociceptor excitation in pain perception. Biomol Ther (Seoul) 2018; 26: 255–267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Dray A, Perkins M. Bradykinin and inflammatory pain. Trends Neurosci 1993; 16: 99–104. [DOI] [PubMed] [Google Scholar]
- 92.Wang A, Yang T, Zhang L, Jia L, Wu Q, Yao S, Xu J, Yang H. IP3-mediated calcium signaling is involved in the mechanism of fractalkine-induced hyperalgesia response. Med Sci Monit 2018; 24: 8804–8811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Khomula EV, Ferrari LF, Araldi D, Levine JD. Sexual dimorphism in a reciprocal interaction of ryanodine and IP3 receptors in the induction of hyperalgesic priming. J Neurosci 2017; 37: 2032–2044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.John LM, Lechleiter JD, Camacho P. Differential modulation of SERCA2 isoforms by calreticulin. J Cell Biol 1998; 142: 963–973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Li Y, Camacho P. Ca2+-dependent redox modulation of SERCA 2b by ERp57. J Cell Biol 2004; 164: 35–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Duncan C, Mueller S, Simon E, Renger JJ, Uebele VN, Hogan QH, Wu HE. Painful nerve injury decreases sarco-endoplasmic reticulum Ca(2)(+)-ATPase activity in axotomized sensory neurons. Neuroscience 2013; 231: 247–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Fernyhough P, Calcutt NA. Abnormal calcium homeostasis in peripheral neuropathies. Cell Calcium 2010; 47: 130–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Zherebitskaya E, Schapansky J, Akude E, Smith DR, Van der Ploeg R, Solovyova N, Verkhratsky A, Fernyhough P. Sensory neurons derived from diabetic rats have diminished internal Ca2+ stores linked to impaired re-uptake by the endoplasmic reticulum. ASN Neuro 2012; 4: AN20110038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Guo Y, Zhang Z, Wu HE, Luo ZD, Hogan QH, Pan B. Increased thrombospondin-4 after nerve injury mediates disruption of intracellular calcium signaling in primary sensory neurons. Neuropharmacology 2017; 117: 292–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Rigaud M, Gemes G, Weyker PD, Cruikshank JM, Kawano T, Wu HE, Hogan QH. Axotomy depletes intracellular calcium stores in primary sensory neurons. Anesthesiology 2009; 111: 381–392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Manjarrés IM, Alonso MT, García-Sancho J. Calcium entry-calcium refilling (CECR) coupling between store-operated Ca2+ entry and sarco/endoplasmic reticulum Ca2+-ATPase. Cell Calcium 2011; 49: 153–161. [DOI] [PubMed] [Google Scholar]
- 102.Manjarrés IM, Rodríguez-García A, Alonso MT, García-Sancho J. The sarco/endoplasmic reticulum Ca2+ ATPase (SERCA) is the third element in capacitative calcium entry. Cell Calcium 2010; 47: 412–418. [DOI] [PubMed] [Google Scholar]
- 103.Kappel S, Borgström A, Stokłosa P, Dörr K, Peinelt C. Store-operated calcium entry in disease: beyond STIM/Orai expression levels. Semin Cell Dev Biol 2019; 94: 66–73. [DOI] [PubMed] [Google Scholar]
- 104.Liou J, Kim ML, Do Heo W, Jones JT, Myers JW, Ferrell JE, Meyer T. STIM is a Ca2+ sensor essential for Ca2+-store-depletion-triggered Ca2+ influx. Curr Biol 2005; 15: 1235–1241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Williams RT, Manji SS, Parker NJ, Hancock MS, Van Stekelenburg L, Eid JP, Senior PV, Kazenwadel JS, Shandala T, Saint R, Smith PJ, Dziadek MA. Identification and characterization of the STIM (stromal interaction molecule) gene family: coding for a novel class of transmembrane proteins. Biochem J 2001; 357: 673–685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Luik RM, Wu MM, Buchanan J, Lewis RS. The elementary unit of store-operated Ca2+ entry: local activation of CRAC channels by STIM1 at ER–plasma membrane junctions. J Cell Biol 2006; 174: 815–825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Prakriya M, Lewis RS. Store-operated calcium channels. Physiol Rev 2015; 95: 1383–1436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Wu MM, Buchanan J, Luik RM, Lewis RS. Ca2+ store depletion causes STIM1 to accumulate in ER regions closely associated with the plasma membrane. J Cell Biol 2006; 174: 803–813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Wissenbach U, Philipp SE, Gross SA, Cavalié A, Flockerzi V. Primary structure, chromosomal localization and expression in immune cells of the murine ORAI and STIM genes. Cell Calcium 2007; 42: 439–446. [DOI] [PubMed] [Google Scholar]
- 110.Gross SA, Wissenbach U, Philipp SE, Freichel M, Cavalié A, Flockerzi V. Murine ORAI2 splice variants form functional Ca2+ release-activated Ca2+ (CRAC) channels. J Biol Chem 2007; 282: 19375–19384. [DOI] [PubMed] [Google Scholar]
- 111.Gwack Y, Srikanth S, Feske S, Cruz-Guilloty F, Oh-Hora M, Neems DS, Hogan PG, Rao A. Biochemical and functional characterization of ORAI proteins. J Biol Chem 2007; 282: 16232–16243. [DOI] [PubMed] [Google Scholar]
- 112.Guzman R, Valente EG, Pretorius J, Pacheco E, Qi M, Bennett BD, Fong DH, Lin FF, Bi V, McBride HJ. Expression of ORAI1, a plasma membrane resident subunit of the CRAC channel, in rodent and non-rodent species. J Histochem Cytochem 2014; 62: 864–878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Zhang SL, Yeromin AV, Zhang XHF, Yu Y, Safrina O, Penna A, Roos J, Stauderman KA, Cahalan MD. Genome-wide RNAi screen of Ca2+ influx identifies genes that regulate Ca2+ release-activated Ca2+ channel activity. Proc Natl Acad Sci USA 2006; 103: 9357–9362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Vig M, Peinelt C, Beck A, Koomoa DL, Rabah D, Koblan-Huberson M, Kraft S, Turner H, Fleig A, Penner R, Kinet JP. CRACM1 is a plasma membrane protein essential for store-operated Ca2+ entry. Science 2006; 312: 1220–1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Feske S, Gwack Y, Prakriya M, Srikanth S, Puppel SH, Tanasa B, Hogan PG, Lewis RS, Daly M, Rao A. A mutation in Orai1 causes immune deficiency by abrogating CRAC channel function. Nature 2006; 441: 179–185. [DOI] [PubMed] [Google Scholar]
- 116.Lin FF, Elliott R, Colombero A, Gaida K, Kelley L, Moksa A, Ho SY, Bykova E, Wong M, Rathanaswami P, Hu S, Sullivan JK, Nguyen HQ, McBride HJ. Generation and characterization of fully human monoclonal antibodies against human Orai1 for autoimmune disease. J Pharmacol Exp Ther 2013; 345: 225–238. [DOI] [PubMed] [Google Scholar]
- 117.Wegierski T, Kuznicki J. Neuronal calcium signaling via store-operated channels in health and disease. Cell Calcium 2018; 74: 102–111. [DOI] [PubMed] [Google Scholar]
- 118.Gao XH, Gao R, Tian YZ, McGonigle P, Barrett JE, Dai Y, Hu H. A store-operated calcium channel inhibitor attenuates collagen-induced arthritis. Br J Pharmacol 2015; 172: 2991–3002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Wang L, Hao J, Zhang Y, Yang Z, Cao Y, Lu W, Shu Y, Jiang L, Hu Y, Lv W, Liu Y, Dong P. Orai1 mediates tumor-promoting store-operated Ca2+ entry in human gastrointestinal stromal tumors via c-KIT and the extracellular signal–regulated kinase pathway. Tumour Biol 2017; 39: 1010428317691426. [DOI] [PubMed] [Google Scholar]
- 120.Gao R, Gao X, Xia J, Tian Y, Barrett JE, Dai Y, Hu H. Potent analgesic effects of a store-operated calcium channel inhibitor. Pain 2013; 154: 2034–2044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Dou Y, Xia J, Gao R, Gao X, Munoz FM, Wei D, Tian Y, Barrett JE, Ajit S, Meucci O, Putney JW, Jr, Dai Y, Hu H. Orai1 plays a crucial role in central sensitization by modulating neuronal excitability. J Neurosci 2018; 38: 887–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Wei D, Mei Y, Xia J, Hu H. Orai1 and Orai3 mediate store-operated calcium entry contributing to neuronal excitability in dorsal root ganglion neurons. Front Cell Neurosci 2017; 11: 400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Hajnoczky G, Robb-Gaspers LD, Seitz MB, Thomas AP. Decoding of cytosolic calcium oscillations in the mitochondria. Cell 1995; 82: 415–424. [DOI] [PubMed] [Google Scholar]
- 124.Contreras L, Drago I, Zampese E, Pozzan T. Mitochondria: the calcium connection. Biochim Biophys Acta 2010; 1797: 607–618. [DOI] [PubMed] [Google Scholar]
- 125.Prins D, Michalak M. Organellar calcium buffers. Cold Spring Harb Perspect Biol 2011; 3: 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Williams GS, Boyman L, Chikando AC, Khairallah RJ, Lederer WJ. Mitochondrial calcium uptake. Proc Natl Acad Sci USA 2013; 110: 10479–10486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Marchi S, Pinton P. The mitochondrial calcium uniporter complex: molecular components, structure and physiopathological implications. J Physiol (Lond) 2014; 592: 829–839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Filadi R, Theurey P, Pizzo P. The endoplasmic reticulum-mitochondria coupling in health and disease: Molecules, functions and significance. Cell Calcium 2017; 62: 1–15. [DOI] [PubMed] [Google Scholar]
- 129.Gomez-Suaga P, Paillusson S, Stoica R, Noble W, Hanger DP, Miller C. The ER-mitochondria tethering complex VAPB-PTPIP51 regulates autophagy. Curr Biol 2017; 27: 371–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Rizzuto R, Bernardi P, Pozzan T. Mitochondria as all-round players of the calcium game. J Physiol (Lond) 2000; 529 Pt 1: 37–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Ivannikov MV, Macleod GT. Mitochondrial free Ca(2)(+) levels and their effects on energy metabolism in Drosophila motor nerve terminals. Biophys J 2013; 104: 2353–2361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Gordeeva AV, Zvyagilskaya RA, Labas YA. Cross-talk between reactive oxygen species and calcium in living cells. Biochemistry Mosc 2003; 68: 1077–1080. [DOI] [PubMed] [Google Scholar]
- 133.Sesti F, Liu S, Cai SQ. Oxidation of potassium channels by ROS: a general mechanism of aging and neurodegeneration? Trends Cell Biol 2010; 20: 45–51. [DOI] [PubMed] [Google Scholar]
- 134.N'Gouemo P. BKCa channel dysfunction in neurological diseases. Front Physiol 2014; 5: 373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Boyman L, Williams GS, Khananshvili D, Sekler I, Lederer WJ. NCLX: the mitochondrial sodium calcium exchanger. J Mol Cell Cardiol 2013; 59: 205–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Boyman L, Karbowski M, Lederer WJ. Regulation of mitochondrial ATP production: Ca(2+) signaling and quality control. Trends Mol Med 2020; 26: 21–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Finkel T, Menazza S, Holmstrom KM, Parks RJ, Liu J, Sun J, Liu J, Pan X, Murphy E. The ins and outs of mitochondrial calcium. Circ Res 2015; 116: 1810–1819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Shutov LP, Kim MS, Houlihan PR, Medvedeva YV, Usachev YM. Mitochondria and plasma membrane Ca2+-ATPase control presynaptic Ca2+ clearance in capsaicin-sensitive rat sensory neurons. J Physiol (Lond) 2013; 591: 2443–2462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Nita II, Caspi Y, Gudes S, Fishman D, Lev S, Hersfinkel M, Sekler I, Binshtok AM. Privileged crosstalk between TRPV1 channels and mitochondrial calcium shuttling machinery controls nociception. Biochim Biophys Acta 2016; 1863: 2868–2880. [DOI] [PubMed] [Google Scholar]
- 140.Valenzuela V, Onate M, Hetz C, Court FA. Injury to the nervous system: a look into the ER. Brain Res 2016; 1648: 617–625. [DOI] [PubMed] [Google Scholar]
- 141.Ying Z, Zhai R, McLean NA, Johnston JM, Misra V, Verge VM. The unfolded protein response and cholesterol biosynthesis link luman/CREB3 to regenerative axon growth in sensory neurons. J Neurosci 2015; 35: 14557–14570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Yamaguchi Y, Oh-Hashi K, Matsuoka Y, Takemura H, Yamakita S, Matsuda M, Sawa T, Amaya F. Endoplasmic reticulum stress in the dorsal root ganglion contributes to the development of pain hypersensitivity after nerve injury. Neuroscience 2018; 394: 288–299. [DOI] [PubMed] [Google Scholar]
- 143.Castillo V, Onate M, Woehlbier U, Rozas P, Andreu C, Medinas D, Valdes P, Osorio F, Mercado G, Vidal RL, Kerr B, Court FA, Hetz C. Functional role of the disulfide isomerase ERp57 in axonal regeneration. PLoS One 2015; 10: e0136620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Ohtake Y, Matsuhisa K, Kaneko M, Kanemoto S, Asada R, Imaizumi K, Saito A. Axonal activation of the unfolded protein response promotes axonal regeneration following peripheral nerve injury. Neuroscience 2018; 375: 34–48. [DOI] [PubMed] [Google Scholar]
- 145.Melemedjian OK, Asiedu MN, Tillu DV, Sanoja R, Yan J, Lark A, Khoutorsky A, Johnson J, Peebles KA, Lepow T, Sonenberg N, Dussor G, Price TJ. Targeting adenosine monophosphate-activated protein kinase (AMPK) in preclinical models reveals a potential mechanism for the treatment of neuropathic pain. Mol Pain 2011; 7: 70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Zhang E, Yi MH, Shin N, Baek H, Kim S, Kim E, Kwon K, Lee S, Kim HW, Chul Bae Y, Kim Y, Kwon OY, Lee WH, Kim DW. Endoplasmic reticulum stress impairment in the spinal dorsal horn of a neuropathic pain model. Sci Rep 2015; 5: 11555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Liu Y, Wang S, Wang Z, Ding M, Li X, Guo J, Han G, Zhao P. Dexmedetomidine alleviated endoplasmic reticulum stress via inducing ER-phagy in the spinal cord of neuropathic pain model. Front Neurosci 2020; 14: 90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Ge Y, Jiao Y, Li P, Xiang Z, Li Z, Wang L, Li W, Gao H, Shao J, Wen D, Yu W. Coregulation of endoplasmic reticulum stress and oxidative stress in neuropathic pain and disinhibition of the spinal nociceptive circuitry. Pain 2018; 159: 894–906. [DOI] [PubMed] [Google Scholar]
- 149.Persson AK, Hoeijmakers JGJ, Estacion M, Black JA, Waxman SG. Sodium channels, mitochondria, and axonal degeneration in peripheral neuropathy. Trends Mol Med 2016; 22: 377–390. [DOI] [PubMed] [Google Scholar]
- 150.Wang X, Ma W, Wang T, Yang J, Wu Z, Liu K, Dai Y, Zang C, Liu W, Liu J, Liang Y, Guo J, Li L. BDNF-TrkB and proBDNF-p75NTR/sortilin signaling pathways are involved in mitochondria-mediated neuronal apoptosis in dorsal root ganglia after sciatic nerve transection. CNS Neurol Disord Drug Targets 2020; 19: 66–82. [DOI] [PubMed] [Google Scholar]
- 151.Kim WS, Park JY, Kim TK, Baik SW. The changes of mitochondrial cytochrome c and GABAergic neuron in neuropathic pain model. Korean J Anesthesiol 2012; 62: 365–370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Englezou PC, Esposti MD, Wiberg M, Reid AJ, Terenghi G. Mitochondrial involvement in sensory neuronal cell death and survival. Exp Brain Res 2012; 221: 357–367. [DOI] [PubMed] [Google Scholar]
- 153.Campbell JN, Meyer RA. Mechanisms of neuropathic pain. Neuron 2006; 52: 77–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Pearson ER. Type 2 diabetes: a multifaceted disease. Diabetologia 2019; 62: 1107–1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Li W, Huang E, Gao S. Type 1 diabetes mellitus and cognitive impairments: a systematic review. J Alzheimers Dis 2017; 57: 29–36. [DOI] [PubMed] [Google Scholar]
- 156.Iqbal Z, Azmi S, Yadav R, Ferdousi M, Kumar M, Cuthbertson DJ, Lim J, Malik RA, Alam U. Diabetic peripheral neuropathy: epidemiology, diagnosis, and pharmacotherapy. Clin Ther 2018; 40: 828–849. [DOI] [PubMed] [Google Scholar]
- 157.Singh J, Yousuf MS, Jones KE, Shelemey PTM, Joy T, Macandili H, Kerr BJ, Zochodne DW, Sauve Y, Ballanyi K, Webber CA. Characterization of the Nile Grass rat as a unique model for type 2 diabetic polyneuropathy. J Neuropathol Exp Neurol 2018; 77: 469–478. [DOI] [PubMed] [Google Scholar]
- 158.Obrosova IG. Diabetic painful and insensate neuropathy: pathogenesis and potential treatments. Neurotherapeutics 2009; 6: 638–647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Lupachyk S, Watcho P, Stavniichuk R, Shevalye H, Obrosova IG. Endoplasmic reticulum stress plays a key role in the pathogenesis of diabetic peripheral neuropathy. Diabetes 2013; 62: 944–952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Inceoglu B, Bettaieb A, Trindade da Silva CA, Lee KS, Haj FG, Hammock BD. Endoplasmic reticulum stress in the peripheral nervous system is a significant driver of neuropathic pain. Proc Natl Acad Sci USA 2015; 112: 9082–9087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Eberhardt MJ, Filipovic MR, Leffler A, de la Roche J, Kistner K, Fischer MJ, Fleming T, Zimmermann K, Ivanovic-Burmazovic I, Nawroth PP, Bierhaus A, Reeh PW, Sauer SK. Methylglyoxal activates nociceptors through transient receptor potential channel A1 (TRPA1): a possible mechanism of metabolic neuropathies. J Biol Chem 2012; 287: 28291–28306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Andersson DA, Gentry C, Light E, Vastani N, Vallortigara J, Bierhaus A, Fleming T, Bevan S. Methylglyoxal evokes pain by stimulating TRPA1. PLoS One 2013; 8: e77986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Griggs RB, Laird DE, Donahue RR, Fu W, Taylor BK. Methylglyoxal requires AC1 and TRPA1 to produce pain and spinal neuron activation. Front Neurosci 2017; 11: 679–612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Griggs RB, Santos DF, Laird DE, Doolen S, Donahue RR, Wessel CR, Fu W, Sinha GP, Wang P, Zhou J, Brings S, Fleming T, Nawroth PP, Susuki K, Taylor BK. Methylglyoxal and a spinal TRPA1-AC1-Epac cascade facilitate pain in the db/db mouse model of type 2 diabetes. Neurobiol Dis 2019; 127: 76–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Barragan-Iglesias P, Kuhn J, Vidal-Cantu GC, Salinas-Abarca AB, Granados-Soto V, Dussor GO, Campbell ZT, Price TJ. Activation of the integrated stress response in nociceptors drives methylglyoxal-induced pain. Pain 2019; 160: 160–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Dull MM, Riegel K, Tappenbeck J, Ries V, Strupf M, Fleming T, Sauer SK, Namer B. Methylglyoxal causes pain and hyperalgesia in human through C-fiber activation. Pain 2019; 160: 2497–2507. [DOI] [PubMed] [Google Scholar]
- 167.Rumora AE, Savelieff MG, Sakowski SA, Feldman EL. Disorders of mitochondrial dynamics in peripheral neuropathy: clues from hereditary neuropathy and diabetes. Int Rev Neurobiol 2019; 145: 127–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Srinivasan S, Stevens M, Wiley JW. Diabetic peripheral neuropathy: evidence for apoptosis and associated mitochondrial dysfunction. Diabetes 2000; 49: 1932–1938. [DOI] [PubMed] [Google Scholar]
- 169.Vincent AM, Russell JW, Low P, Feldman EL. Oxidative stress in the pathogenesis of diabetic neuropathy. Endocr Rev 2004; 25: 612–628. [DOI] [PubMed] [Google Scholar]
- 170.Vincent AM, Edwards JL, McLean LL, Hong Y, Cerri F, Lopez I, Quattrini A, Feldman EL. Mitochondrial biogenesis and fission in axons in cell culture and animal models of diabetic neuropathy. Acta Neuropathol 2010; 120: 477–489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Sifuentes-Franco S, Pacheco-Moisés FP, Rodríguez-Carrizalez AD, Miranda-Díaz AG. The role of oxidative stress, mitochondrial function, and autophagy in diabetic polyneuropathy. J Diabetes Res 2017; 2017: 1673081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Verkhratsky A, Fernyhough P. Mitochondrial malfunction and Ca2+ dyshomeostasis drive neuronal pathology in diabetes. Cell Calcium 2008; 44: 112–122. [DOI] [PubMed] [Google Scholar]
- 173.Hernández-Beltrán N, Moreno CB, Gutiérrez-Álvarez AM. Contribution of mitochondria to pain in diabetic neuropathy. Endocrinol Nutr 2013; 60: 25–32. [DOI] [PubMed] [Google Scholar]
- 174.Bennett GJ, Doyle T, Salvemini D. Mitotoxicity in distal symmetrical sensory peripheral neuropathies. Nat Rev Neurol 2014; 10: 326–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Kostyuk E, Svichar N, Shishkin V, Kostyuk P. Role of mitochondrial dysfunction in calcium signalling alterations in dorsal root ganglion neurons of mice with experimentally-induced diabetes. Neuroscience 1999; 90: 535–541. [DOI] [PubMed] [Google Scholar]
- 176.Flatters SJ, Bennett GJ. Studies of peripheral sensory nerves in paclitaxel-induced painful peripheral neuropathy: evidence for mitochondrial dysfunction. Pain 2006; 122: 245–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Seretny M, Currie GL, Sena ES, Ramnarine S, Grant R, MacLeod MR, Colvin LA, Fallon M. Incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy: a systematic review and meta-analysis. Pain 2014; 155: 2461–2470. [DOI] [PubMed] [Google Scholar]
- 178.Zajączkowska R, Kocot-Kępska M, Leppert W, Wrzosek A, Mika J, Wordliczek J. Mechanisms of chemotherapy-induced peripheral neuropathy. IJMS 2019; 20: 1451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.De Iuliis F, Taglieri L, Salerno G, Lanza R, Scarpa S. Taxane induced neuropathy in patients affected by breast cancer: literature review. Crit Rev Oncol Hematol 2015; 96: 34–45. [DOI] [PubMed] [Google Scholar]
- 180.Ewertz M, Qvortrup C, Eckhoff L. Chemotherapy-induced peripheral neuropathy in patients treated with taxanes and platinum derivatives. Acta Oncol 2015; 54: 587–591. [DOI] [PubMed] [Google Scholar]
- 181.Sisignano M, Baron R, Scholich K, Geisslinger G. Mechanism-based treatment for chemotherapy-induced peripheral neuropathic pain. Nat Rev Neurol 2014; 10: 694–707. [DOI] [PubMed] [Google Scholar]
- 182.Megat S, Ray PR, Moy JK, Lou TF, Barragan-Iglesias P, Li Y, Pradhan G, Wanghzou A, Ahmad A, Burton MD, North RY, Dougherty PM, Khoutorsky A, Sonenberg N, Webster KR, Dussor G, Campbell ZT, Price TJ. Nociceptor translational profiling reveals the Ragulator-Rag GTPase complex as a critical generator of neuropathic pain. J Neurosci 2019; 39: 393–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Nam SM, Jeon YJ. Proteostasis in the endoplasmic reticulum: road to cure. Cancers (Basel) 2019; 11: 1793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Resham K, Sharma SS. Pharmacological interventions targeting Wnt/beta-catenin signaling pathway attenuate paclitaxel-induced peripheral neuropathy. Eur J Pharmacol 2019; 864: 172714–172710. [DOI] [PubMed] [Google Scholar]
- 185.Andoh T, Kobayashi N, Uta D, Kuraishi Y. Prophylactic topical paeoniflorin prevents mechanical allodynia caused by paclitaxel in mice through adenosine A1 receptors. Phytomedicine 2017; 25: 1–7. [DOI] [PubMed] [Google Scholar]
- 186.Tanimukai H, Kanayama D, Omi T, Takeda M, Kudo T. Paclitaxel induces neurotoxicity through endoplasmic reticulum stress. Biochem Biophys Res Commun 2013; 437: 151–155. [DOI] [PubMed] [Google Scholar]
- 187.Tanimukai H, Kudo T. Fluvoxamine alleviates paclitaxel-induced neurotoxicity. Biochem Biophys Rep 2015; 4: 202–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Duggett NA, Griffiths LA, Flatters S. Paclitaxel-induced painful neuropathy is associated with changes in mitochondrial bioenergetics, glycolysis, and an energy deficit in dorsal root ganglia neurons. Pain 2017; 158: 1499–1508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Jia M, Wu C, Gao F, Xiang H, Sun N, Peng P, Li J, Yuan X, Li H, Meng X, Tian B, Shi J, Li M. Activation of NLRP3 inflammasome in peripheral nerve contributes to paclitaxel-induced neuropathic pain. Mol Pain 2017; 13: 1744806917719804–1744806917719807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Canta A, Pozzi E, Carozzi VA. Mitochondrial dysfunction in chemotherapy-induced peripheral neuropathy (CIPN). Toxics 2015; 3: 198–223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Waseem M, Kaushik P, Tabassum H, Parvez S. Role of mitochondrial mechanism in chemotherapy-induced peripheral neuropathy. Curr Drug Metab 2018; 19: 47–54. [DOI] [PubMed] [Google Scholar]
- 192.Halestrap AP. What is the mitochondrial permeability transition pore? J Mol Cell Cardiol 2009; 46: 821–831. [DOI] [PubMed] [Google Scholar]
- 193.Kidd JF, Pilkington MF, Schell MJ, Fogarty KE, Skepper JN, Taylor CW, Thorn P. Paclitaxel affects cytosolic calcium signals by opening the mitochondrial permeability transition pore. J Biol Chem 2002; 277: 6504–6510. [DOI] [PubMed] [Google Scholar]
- 194.Xiao WH, Bennett GJ. Chemotherapy-evoked neuropathic pain: abnormal spontaneous discharge in A-fiber and C-fiber primary afferent neurons and its suppression by acetyl-L-carnitine. Pain 2008; 135: 262–270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Clayton BLL, Popko B. Endoplasmic reticulum stress and the unfolded protein response in disorders of myelinating glia. Brain Res 2016; 1648: 594–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Rahmati M, Moosavi MA, McDermott MF. ER stress: a therapeutic target in rheumatoid arthritis? Trends Pharmacol Sci 2018; 39: 610–623. [DOI] [PubMed] [Google Scholar]
- 197.Bae C, Wang J, Shim HS, Tang SJ, Chung JM and La JH. Mitochondrial superoxide increases excitatory synaptic strength in spinal dorsal horn neurons of neuropathic mice. Mol Pain 2018; 14: 1--9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Graham ZA, Harlow L, Bauman WA and Cardozo CP. Alterations in mitochondrial fission, fusion, and mitophagic protein expression in the gastrocnemius of mice after a sciatic nerve transection. Muscle Nerve 2018; 58: 592-599. [DOI] [PubMed] [Google Scholar]
- 199.Hall KE, Sima AA and Wiley JW. Voltage-dependent calcium currents are enhanced in dorsal root ganglion neurones from the Bio Bred/Worchester diabetic rat. J Physiol 1995; 486 ( Pt 2): 313--322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Russell JW, Golovoy D, Vincent AM, Mahendru P, Olzmann JA, Mentzer A and Feldman EL. High glucose-induced oxidative stress and mitochondrial dysfunction in neurons. FASEB J 2002; 16: 1738--1748. [DOI] [PubMed] [Google Scholar]