Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 787
Filter
1.
Nat Commun ; 15(1): 8585, 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39362841

ABSTRACT

Chronic pain remains a significant medical challenge with complex underlying mechanisms, and an urgent need for new treatments. Our research built and utilized the iPain single-cell atlas to study chronic pain progression in dorsal root and trigeminal ganglia. We discovered that senescence of a small subset of pain-sensing neurons may be a driver of chronic pain. This mechanism was observed in animal models after nerve injury and in human patients diagnosed with chronic pain or diabetic painful neuropathy. Notably, treatment with senolytics, drugs that remove senescent cells, reversed pain symptoms in mice post-injury. These findings highlight the role of cellular senescence in chronic pain development, demonstrate the therapeutic potential of senolytic treatments, and underscore the value of the iPain atlas for future pain research.


Subject(s)
Cellular Senescence , Chronic Pain , Ganglia, Spinal , Nociceptors , Single-Cell Analysis , Transcriptome , Animals , Humans , Nociceptors/metabolism , Nociceptors/drug effects , Chronic Pain/genetics , Chronic Pain/drug therapy , Chronic Pain/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Mice , Cellular Senescence/drug effects , Cellular Senescence/genetics , Male , Senotherapeutics/pharmacology , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/drug effects , Mice, Inbred C57BL , Disease Models, Animal , Female , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/genetics
2.
Transl Psychiatry ; 14(1): 417, 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39366962

ABSTRACT

Sleep disruption and negative affect are attendant features of many psychiatric and neurological conditions that are often co-morbid including major depressive disorder, generalized anxiety disorder and chronic pain. Whether there is a causal relationship between negative affect and sleep disruption remains unclear. We therefore asked if mechanisms promoting negative affect can disrupt sleep and whether inhibition of pathological negative affect can normalize disrupted sleep. Signaling at the kappa opioid receptor (KOR) elicits dysphoria in humans and aversive conditioning in animals. We tested the possibility that (a) increased KOR signaling in the anterior cingulate cortex (ACC), a brain region associated with negative emotions, would be sufficient to promote both aversiveness and sleep disruption and (b) inhibition of KOR signaling would normalize pathological negative affect and sleep disruption induced by chronic pain. Chemogenetic Gi-mediated inhibition of KOR-expressing ACC neurons produced conditioned place aversion (CPA) as well as sleep fragmentation in naïve mice. CRISPR/Cas9 editing of ACC KOR normalized both the negative affect and sleep disruption elicited by pathological chronic pain while maintaining the physiologically critical sensory features of pain. These findings suggest therapeutic utility of KOR antagonists for treatment of disease conditions that are associated with both negative affect and sleep disturbances.


Subject(s)
Chronic Pain , Gyrus Cinguli , Receptors, Opioid, kappa , Animals , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, kappa/genetics , Mice , Gyrus Cinguli/metabolism , Gyrus Cinguli/physiopathology , Male , Chronic Pain/metabolism , Chronic Pain/physiopathology , Sleep Wake Disorders/metabolism , Sleep Wake Disorders/physiopathology , Mice, Inbred C57BL , Affect/physiology
3.
Sci Signal ; 17(857): eadn4694, 2024 10 08.
Article in English | MEDLINE | ID: mdl-39378285

ABSTRACT

The development of new analgesics has been challenging. Candidate drugs often have limited clinical utility due to side effects that arise because many drug targets are involved in signaling pathways other than pain transduction. Here, we explored the potential of targeting protein-protein interactions (PPIs) that mediate pain signaling as an approach to developing drugs to treat chronic pain. We reviewed the approaches used to identify small molecules and peptide modulators of PPIs and their ability to decrease pain-like behaviors in rodent animal models. We analyzed data from rodent and human sensory nerve tissues to build associated signaling networks and assessed both validated and potential interactions and the structures of the interacting domains that could inform the design of synthetic peptides and small molecules. This resource identifies PPIs that could be explored for the development of new analgesics, particularly between scaffolding proteins and receptors for various growth factors and neurotransmitters, as well as ion channels and other enzymes. Targeting the adaptor function of CBL by blocking interactions between its proline-rich carboxyl-terminal domain and its SH3-domain-containing protein partners, such as GRB2, could disrupt endosomal signaling induced by pain-associated growth factors. This approach would leave intact its E3-ligase functions, which are mediated by other domains and are critical for other cellular functions. This potential of PPI modulators to be more selective may mitigate side effects and improve the clinical management of pain.


Subject(s)
Analgesics , Signal Transduction , Humans , Animals , Analgesics/pharmacology , Analgesics/chemistry , Signal Transduction/drug effects , Protein Interaction Maps/drug effects , Chronic Pain/drug therapy , Chronic Pain/metabolism , Pain/metabolism , Pain/drug therapy
4.
Int J Mol Sci ; 25(17)2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39273183

ABSTRACT

Transient receptor potential (TRP) channels, first identified in Drosophila in 1969, are multifunctional ion channels expressed in various cell types. Structurally, TRP channels consist of six membrane segments and are classified into seven subfamilies. Transient receptor potential ankyrin 1 (TRPA1), the first member of the TRPA family, is a calcium ion affinity non-selective cation channel involved in sensory transduction and responds to odors, tastes, and chemicals. It also regulates temperature and responses to stimuli. Recent studies have linked TRPA1 to several disorders, including chronic pain, inflammatory diseases, allergies, and respiratory problems, owing to its activation by environmental toxins. Mutations in TRPA1 can affect the sensory nerves and microvasculature, potentially causing nerve pain and vascular problems. Understanding the function of TRPA1 is important for the development of treatments for these diseases. Recent developments in nanomedicines that target various ion channels, including TRPA1, have had a significant impact on disease treatment, providing innovative alternatives to traditional disease treatments by overcoming various adverse effects.


Subject(s)
TRPA1 Cation Channel , Humans , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/genetics , Animals , Nanomedicine/methods , Hypersensitivity/drug therapy , Hypersensitivity/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Chronic Pain/drug therapy , Chronic Pain/metabolism
6.
Brain Res Bull ; 217: 111081, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39277019

ABSTRACT

Chronic pain is a universal public health problem with nearly one third of global human involved, which causes significant distressing personal burden. After painful stimulus, neurobiological changes occur not only in peripheral nervous system but also in central nervous system where somatosensory cortex is important for nociception. Being an ion channel, transient receptor potential vanilloid 1 (TRPV1) act as an inflammatory detector in the brain. Thymic stromal lymphopoietin (TSLP) is a potent neuroinflammation mediator after nerve injury. Bleomycin is applied to treat dermatologic diseases, and its administration elicits local painful sensation. However, whether bleomycin administration can cause chronic pain remains unknown. In the present study, we aimed to investigate how mice develop chronic pain after receiving repeated bleomycin administration. In addition, the relevant neurobiological brain changes after noxious stimuli were clarified. C57BL/6 mice aged five- to six-weeks were randomly classified into two group, PBS (normal) group and bleomycin group which bleomycin was intradermally administered to back five times a week over a three-week period. Calibrated forceps testing was used to measure mouse pain threshold. Western blots were used to assess neuroinflammatory response; immunofluorescence assay was used to measure the status of neuron apoptosis, glial reaction, and neuro-glial communication. Bleomycin administration induced mechanical nociception and activated both TRPV1 and TSLP/TSLPR/pSTAT5 signals in mouse somatosensory cortex. Through these pathways, bleomycin not only activates glial reaction but also causes neuronal apoptosis. TRPV1 and TSLP/TSLPR/pSTAT5 signaling had co-labeled each other by immunofluorescence assay. Taken together, our study provides a new chronic pain model by repeated intradermal bleomycin injection by activating TRPV1 and glial reaction-mediated neuroinflammation via TSLP/TSLPR/pSTAT5 signals.


Subject(s)
Bleomycin , Cytokines , Neuroglia , Neuroinflammatory Diseases , Nociception , Receptors, Cytokine , Signal Transduction , TRPV Cation Channels , Thymic Stromal Lymphopoietin , Animals , Male , Mice , Bleomycin/toxicity , Chronic Pain/chemically induced , Chronic Pain/metabolism , Cytokines/metabolism , Immunoglobulins , Mice, Inbred C57BL , Neuroglia/metabolism , Neuroglia/drug effects , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/chemically induced , Nociception/drug effects , Nociception/physiology , Receptors, Cytokine/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , STAT5 Transcription Factor/metabolism , TRPV Cation Channels/metabolism
7.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273354

ABSTRACT

Pain is an unpleasant sensory and emotional experience. Adequate pain control is often challenging, particularly in patients with chronic pain. Despite advances in pain management, drug addiction, overtreatment, or substance use disorders are not rare. Hence the need for further studies in the field. The substantial progress made over the last decade has revealed genes, signalling pathways, molecules, and neuronal networks in pain control thus opening new clinical perspectives in pain management. In this respect, data on the epigenetic modulation of opioid and cannabinoid receptors, key actors in the modulation of pain, offered new perspectives to preserve the activity of opioid and endocannabinoid systems to increase the analgesic efficacy of opioid- and cannabinoid-based drugs. Similarly, upcoming data on cannabidiol (CBD), a non-psychoactive cannabinoid in the marijuana plant Cannabis sativa, suggests analgesic, anti-inflammatory, antioxidant, anticonvulsivant and ansiolitic effects and supports its potential application in clinical contexts such as cancer, neurodegeneration, and autoimmune diseases but also in health and fitness with potential use in athletes. Hence, in this review article, we summarize the emerging epigenetic modifications of opioid and cannabinoid receptors and focus on CBD as an emerging non-psychoactive cannabinoid in pain management in clinical practice, health, and fitness.


Subject(s)
Analgesics, Opioid , Cannabinoids , Receptors, Cannabinoid , Humans , Analgesics, Opioid/therapeutic use , Analgesics, Opioid/pharmacology , Cannabinoids/therapeutic use , Cannabinoids/pharmacology , Receptors, Cannabinoid/metabolism , Animals , Pain/drug therapy , Pain/metabolism , Cannabidiol/therapeutic use , Cannabidiol/pharmacology , Receptors, Opioid/metabolism , Epigenesis, Genetic/drug effects , Pain Management/methods , Chronic Pain/drug therapy , Chronic Pain/metabolism , Endocannabinoids/metabolism
8.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39125894

ABSTRACT

Chronic pain is a prevalent condition with a multifaceted pathogenesis, where epigenetic modifications, particularly DNA methylation, might play an important role. This review delves into the intricate mechanisms by which DNA methylation and demethylation regulate genes associated with nociception and pain perception in nociceptive pathways. We explore the dynamic nature of these epigenetic processes, mediated by DNA methyltransferases (DNMTs) and ten-eleven translocation (TET) enzymes, which modulate the expression of pro- and anti-nociceptive genes. Aberrant DNA methylation profiles have been observed in patients with various chronic pain syndromes, correlating with hypersensitivity to painful stimuli, neuronal hyperexcitability, and inflammatory responses. Genome-wide analyses shed light on differentially methylated regions and genes that could serve as potential biomarkers for chronic pain in the epigenetic landscape. The transition from acute to chronic pain is marked by rapid DNA methylation reprogramming, suggesting its potential role in pain chronicity. This review highlights the importance of understanding the temporal dynamics of DNA methylation during this transition to develop targeted therapeutic interventions. Reversing pathological DNA methylation patterns through epigenetic therapies emerges as a promising strategy for pain management.


Subject(s)
Chronic Pain , DNA Methylation , Epigenesis, Genetic , Humans , Chronic Pain/genetics , Chronic Pain/metabolism , Animals
9.
Inflamm Res ; 73(10): 1711-1726, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39095656

ABSTRACT

BACKGROUND AND OBJECTIVE: Neuropathic pain is a chronic condition characterized by aberrant signaling within the somatosensory system, affecting millions of people worldwide with limited treatment options. Herein, we aim at investigating the potential of a sigma-1 receptor (σ1R) antagonist in managing neuropathic pain. METHODS: A Chronic Constriction Injury (CCI) model was used to induce neuropathic pain. The potential of (+)-MR200 was evaluated following daily subcutaneous injections of the compound. Its mechanism of action was confirmed by administration of a well-known σ1R agonist, PRE084. RESULTS: (+)-MR200 demonstrated efficacy in protecting neurons from damage and alleviating pain hypersensitivity in CCI model. Our results suggest that (+)-MR200 reduced the activation of astrocytes and microglia, cells known to contribute to the neuroinflammatory process, suggesting that (+)-MR200 may not only address pain symptoms but also tackle the underlying cellular mechanism involved. Furthermore, (+)-MR200 treatment normalized levels of the gap junction (GJ)-forming protein connexin 43 (Cx43), suggesting a reduction in harmful intercellular communication that could fuel the chronicity of pain. CONCLUSIONS: This approach could offer a neuroprotective strategy for managing neuropathic pain, addressing both pain symptoms and cellular processes driving the condition. Understanding the dynamics of σ1R expression and function in neuropathic pain is crucial for clinical intervention.


Subject(s)
Connexin 43 , Neuralgia , Receptors, sigma , Sigma-1 Receptor , Receptors, sigma/metabolism , Receptors, sigma/agonists , Neuralgia/drug therapy , Neuralgia/metabolism , Animals , Male , Connexin 43/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Cell Communication/drug effects , Chronic Pain/drug therapy , Chronic Pain/metabolism , Microglia/drug effects , Microglia/metabolism , Rats, Sprague-Dawley , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Neurons/drug effects , Neurons/metabolism , Sciatic Nerve/injuries , Morpholines/pharmacology , Morpholines/therapeutic use
10.
Int J Mol Sci ; 25(16)2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39201323

ABSTRACT

Pain perception is the consequence of a complex interplay between activation and inhibition. Noradrenergic pain modulation inhibits nociceptive transmission and pain perception. The main source of norepinephrine (NE) in the central nervous system is the Locus Coeruleus (LC), a small but complex cluster of cells in the pons. The aim of this study is to review the literature on the LC-NE inhibitory system, its influence on chronic pain pathways and its frequent comorbidities. The literature research showed that pain perception is the consequence of nociceptive and environmental processing and is modulated by the LC-NE system. If perpetuated in time, nociceptive inputs can generate neuroplastic changes in the central nervous system that reduce the inhibitory effects of the LC-NE complex and facilitate the development of chronic pain and frequent comorbidities, such as anxiety, depression or sleeping disturbances. The exact mechanisms involved in the LC functional shift remain unknown, but there is some evidence that they occur through plastic changes in the medial and lateral pathways and their brain projections. Additionally, there are other influencing factors, like developmental issues, neuroinflammatory glial changes, NE receptor affinity and changes in LC neuronal firing rates.


Subject(s)
Chronic Pain , Locus Coeruleus , Norepinephrine , Locus Coeruleus/metabolism , Humans , Chronic Pain/physiopathology , Chronic Pain/metabolism , Animals , Norepinephrine/metabolism , Neuronal Plasticity , Neurons/metabolism , Neurons/physiology , Pain Perception/physiology
11.
Mol Pain ; 20: 17448069241275097, 2024.
Article in English | MEDLINE | ID: mdl-39093627

ABSTRACT

Chronic pain is a debilitating symptom with a significant negative impact on the quality of life and socioeconomic status, particularly among adults and the elderly. Major Depressive Disorder (MDD) stands out as one of the most important comorbid disorders accompanying chronic pain. The kynurenine pathway serves as the primary route for tryptophan degradation and holds critical significance in various biological processes, including the regulation of neurotransmitters, immune responses, cancer development, metabolism, and inflammation. This review encompasses key research studies related to the kynurenine pathway in the context of headache, neuropathic pain, gastrointestinal disorders, fibromyalgia, chronic fatigue syndrome, and MDD. Various metabolites produced in the kynurenine pathway, such as kynurenic acid and quinolinic acid, exhibit neuroprotective and neurotoxic effects, respectively. Recent studies have highlighted the significant involvement of kynurenine and its metabolites in the pathophysiology of pain. Moreover, pharmacological interventions targeting the regulation of the kynurenine pathway have shown therapeutic promise in pain management. Understanding the underlying mechanisms of this pathway presents an opportunity for developing personalized, innovative, and non-opioid approaches to pain treatment. Therefore, this narrative review explores the role of the kynurenine pathway in various chronic pain disorders and its association with depression and chronic pain.


Subject(s)
Chronic Pain , Kynurenine , Kynurenine/metabolism , Humans , Chronic Pain/metabolism , Animals , Signal Transduction
12.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39125713

ABSTRACT

Endometriosis, often associated with chronic pelvic pain, can lead to anxiety and depression. This study investigates the role and mechanism of Glycine receptor alpha 3 (Glrα3) in the central sensitization of pain in endometriosis, aiming to identify new therapeutic targets. Using a Glrα3 knockout mouse model of endometriosis, we employed behavioral tests, qPCR, immunofluorescence, Nissl staining, MRI, and Western blot to assess the involvement of Glrα3 in central pain sensitization. Our results indicate that endometriosis-induced hyperalgesia and anxiety-depressive-like behaviors are linked to increased Glrα3 expression. Chronic pain in endometriosis leads to gray matter changes in the sensory and insular cortices, with Glrα3 playing a significant role. The inhibition of Glrα3 alleviates pain, reduces neuronal abnormalities, and decreases glial cell activation. The absence of Glrα3 effectively regulates the central sensitization of pain in endometriosis by inhibiting glial cell activation and maintaining neuronal stability. This study offers new therapeutic avenues for the clinical treatment of endometriosis-related pain.


Subject(s)
Endometriosis , Mice, Knockout , Animals , Female , Mice , Anxiety , Chronic Pain/metabolism , Chronic Pain/etiology , Chronic Pain/pathology , Chronic Pain/genetics , Disease Models, Animal , Endometriosis/metabolism , Endometriosis/pathology , Endometriosis/complications , Endometriosis/genetics , Hyperalgesia/metabolism , Hyperalgesia/etiology , Mice, Inbred C57BL , Pelvic Pain/etiology , Pelvic Pain/metabolism , Receptors, Glycine/genetics , Receptors, Glycine/metabolism
13.
ACS Chem Biol ; 19(7): 1626-1637, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39026469

ABSTRACT

Chronic pain is a prevalent problem that plagues modern society, and better understanding its mechanisms is critical for developing effective therapeutics. Nerve growth factor (NGF) and its primary receptor, Tropomyosin receptor kinase A (TrkA), are known to be potent mediators of chronic pain, but there is a lack of established methods for precisely perturbing the NGF/TrkA signaling pathway in the study of pain and nociception. Optobiological tools that leverage light-induced protein-protein interactions allow for precise spatial and temporal control of receptor signaling. Previously, our lab reported a blue light-activated version of TrkA generated using light-induced dimerization of the intracellular TrkA domain, opto-iTrkA. In this work, we show that opto-iTrkA activation is able to activate endogenous ERK and Akt signaling pathways and causes the retrograde transduction of phospho-ERK signals in dorsal root ganglion (DRG) neurons. Opto-iTrkA activation also sensitizes the transient receptor potential vanilloid 1 (TRPV1) channel in cellular models, further corroborating the physiological relevance of the optobiological stimulus. Finally, we show that opto-iTrkA enables light-inducible potentiation of mechanical sensitization in mice. Light illumination enables nontraumatic and reversible (<2 days) sensitization of mechanical pain in mice transduced with opto-iTrkA, which provides a platform for dissecting TrkA pathways for nociception in vitro and in vivo.


Subject(s)
Chronic Pain , Ganglia, Spinal , Light , Receptor, trkA , Animals , Receptor, trkA/metabolism , Chronic Pain/metabolism , Mice , Ganglia, Spinal/metabolism , TRPV Cation Channels/metabolism , Humans , Signal Transduction , Mice, Inbred C57BL , Male , Nerve Growth Factor/metabolism , Neurons/metabolism
14.
Sci Signal ; 17(847): eadn8936, 2024 07 30.
Article in English | MEDLINE | ID: mdl-39078919

ABSTRACT

Obstructive sleep apnea (OSA) is a prevalent sleep disorder that is associated with increased incidence of chronic musculoskeletal pain. We investigated the mechanism of this association in a mouse model of chronic intermittent hypoxia (CIH) that mimics the repetitive hypoxemias of OSA. After 14 days of CIH, both male and female mice exhibited behaviors indicative of persistent pain, with biochemical markers in the spinal cord dorsal horn and sensory neurons of the dorsal root ganglia consistent with hyperalgesic priming. CIH, but not sleep fragmentation alone, induced an increase in macrophage recruitment to peripheral sensory tissues (sciatic nerve and dorsal root ganglia), an increase in inflammatory cytokines in the circulation, and nociceptor sensitization. Peripheral macrophage ablation blocked CIH-induced hyperalgesic priming. The findings suggest that correcting the hypoxia or targeting macrophage signaling might suppress persistent pain in patients with OSA.


Subject(s)
Hypoxia , Macrophages , Nociceptors , Animals , Hypoxia/metabolism , Macrophages/metabolism , Male , Female , Mice , Nociceptors/metabolism , Ganglia, Spinal/metabolism , Sleep Apnea, Obstructive/metabolism , Mice, Inbred C57BL , Disease Models, Animal , Hyperalgesia/metabolism , Cytokines/metabolism , Chronic Pain/metabolism , Chronic Pain/immunology
15.
CNS Neurosci Ther ; 30(7): e14831, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38961317

ABSTRACT

AIMS: Comorbid anxiodepressive-like symptoms (CADS) in chronic pain are closely related to the overactivation of the lateral habenula (LHb). Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels have been implicated to play a key role in regulating neuronal excitability. However, the role of HCN channels in the LHb during CADS has not yet been characterized. This study aimed to investigate the effect of HCN channels in the LHb on CADS during chronic pain. METHODS: After chronic neuropathic pain induction by spared nerve injury (SNI), mice underwent a sucrose preference test, forced swimming test, tail suspension test, open-field test, and elevated plus maze test to evaluate their anxiodepressive-like behaviors. Electrophysiological recordings, immunohistochemistry, Western blotting, pharmacological experiments, and virus knockdown strategies were used to investigate the underlying mechanisms. RESULTS: Evident anxiodepressive-like behaviors were observed 6w after the SNI surgery, accompanied by increased neuronal excitability, enhanced HCN channel function, and increased expression of HCN2 isoforms in the LHb. Either pharmacological inhibition or virus knockdown of HCN2 channels significantly reduced LHb neuronal excitability and ameliorated both pain and depressive-like behaviors. CONCLUSION: Our results indicated that the LHb neurons were hyperactive under CADS in chronic pain, and this hyperactivation possibly resulted from the enhanced function of HCN channels and up-regulation of HCN2 isoforms.


Subject(s)
Depression , Habenula , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Animals , Habenula/metabolism , Habenula/drug effects , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Mice , Male , Depression/metabolism , Neuralgia/metabolism , Neuralgia/psychology , Mice, Inbred C57BL , Chronic Pain/metabolism , Chronic Pain/psychology , Potassium Channels
16.
Neuropharmacology ; 258: 110067, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-38992792

ABSTRACT

Chronic primary pain (CPP) occurs in the absence of tissue injury and includes temporomandibular disorders (TMD), fibromyalgia syndrome (FMS) and irritable bowel syndrome (IBS). CPP is commonly considered a stress-related chronic pain and often presents as wide-spread pain or comorbid pain conditions in different regions of the body. However, whether prolonged stress can directly result in the development of CPP comorbidity remains unclear. In the present study, we adapted a 21 day heterotypic stress paradigm in mice and examined whether chronic stress induced wide-spread hyperalgesia, modeling comorbid CPP in the clinic. We found that chronic stress induced anxiety- and depression-like behaviors, and resulted in long-lasting wide-spread hyperalgesia over several body regions such as the orofacial area, hindpaw, thigh, upper back and abdomen in female mice. We further found that the expression of cholecystokinin (CCK)1 receptors was significantly increased in the L4-L5 spinal dorsal horn of the female mice after 14 and 21 day heterotypic stress compared with the control animals. Intrathecal injection of the CCK1 receptor antagonist CR-1505 blocked pain hypersensitivity in the subcervical body including the upper back, thigh, hindpaw and abdomen. These findings suggest that the upregulation of spinal CCK1 receptors after chronic stress contributes to the central mechanisms underlying the development of wide-spread hyperalgesia, and may provide a potential and novel central target for clinical treatment of CPP.


Subject(s)
Hyperalgesia , Receptors, Cholecystokinin , Stress, Psychological , Animals , Female , Mice , Anxiety/metabolism , Chronic Pain/metabolism , Disease Models, Animal , Hyperalgesia/metabolism , Mice, Inbred C57BL , Receptors, Cholecystokinin/metabolism , Receptors, Cholecystokinin/antagonists & inhibitors , Spinal Cord/metabolism , Spinal Cord/drug effects , Stress, Psychological/metabolism
17.
Biomed Pharmacother ; 176: 116856, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38852510

ABSTRACT

Fibromyalgia is characterised by widespread chronic pain and is often accompanied by comorbidities such as sleep disorders, anxiety, and depression. Because it is often accompanied by many adverse symptoms and lack of effective treatment, it is important to search for the pathogenesis and treatment of fibromyalgia. Astaxanthin, a carotenoid pigment known for its anti-inflammatory and antioxidant properties, has demonstrated effective analgesic effects in neuropathic pain. However, its impact on fibromyalgia remains unclear. Therefore, in this study, we constructed a mouse model of fibromyalgia and investigated the effect of astaxanthin on chronic pain and associated symptoms through multiple intragastrical injections. We conducted behavioural assessments to detect pain and depression-like states in mice, recorded electroencephalograms to monitor sleep stages, examined c-Fos activation in the anterior cingulate cortex, measured activation of spinal glial cells, and assessed levels of inflammatory factors in the brain and spinal cord, including interleukin (IL)-1ß, IL-6, and tumour necrosis factor- α(TNF-α).Additionally, we analysed the expression levels of IL-6, IL-10, NOD-like receptor thermal protein domain associated protein 3 (NLRP3), Apoptosis-associated speck-like protein containing CARD, and Caspase-1 proteins. The findings revealed that astaxanthin significantly ameliorated mechanical and thermal pain in mice with fibromyalgia and mitigated sleep disorders and depressive-like symptoms induced by pain. A potential mechanism underlying these effects is the anti-inflammatory action of astaxanthin, likely mediated through the inhibition of the NLRP3 inflammasome, which could be one of the pathways through which astaxanthin alleviates fibromyalgia. In conclusion, our study suggests that astaxanthin holds promise as a potential analgesic medication for managing fibromyalgia and its associated symptoms.


Subject(s)
Depression , Fibromyalgia , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Xanthophylls , Animals , Xanthophylls/pharmacology , Fibromyalgia/drug therapy , Fibromyalgia/complications , Fibromyalgia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Inflammasomes/metabolism , Inflammasomes/antagonists & inhibitors , Depression/drug therapy , Depression/metabolism , Mice , Male , Mice, Inbred C57BL , Disease Models, Animal , Analgesics/pharmacology , Anti-Inflammatory Agents/pharmacology , Chronic Pain/drug therapy , Chronic Pain/metabolism , Cytokines/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism , Behavior, Animal/drug effects
18.
Sci Rep ; 14(1): 14715, 2024 06 26.
Article in English | MEDLINE | ID: mdl-38926482

ABSTRACT

Opioids are the gold standard for the treatment of chronic pain but are limited by adverse side effects. In our earlier work, we showed that Heat shock protein 90 (Hsp90) has a crucial role in regulating opioid signaling in spinal cord; Hsp90 inhibition in spinal cord enhances opioid anti-nociception. Building on these findings, we injected the non-selective Hsp90 inhibitor KU-32 by the intrathecal route into male and female CD-1 mice, showing that morphine anti-nociceptive potency was boosted by 1.9-3.5-fold in acute and chronic pain models. At the same time, tolerance was reduced from 21-fold to 2.9 fold and established tolerance was rescued, while the potency of constipation and reward was unchanged. These results demonstrate that spinal Hsp90 inhibition can improve the therapeutic index of morphine. However, we also found that systemic non-selective Hsp90 inhibition blocked opioid pain relief. To avoid this effect, we used selective small molecule inhibitors and CRISPR gene editing to identify 3 Hsp90 isoforms active in spinal cord (Hsp90α, Hsp90ß, and Grp94) while only Hsp90α was active in brain. We thus hypothesized that a systemically delivered selective inhibitor to Hsp90ß or Grp94 could selectively inhibit spinal cord Hsp90 activity, resulting in enhanced opioid therapy. We tested this hypothesis using intravenous delivery of KUNB106 (Hsp90ß) and KUNG65 (Grp94), showing that both drugs enhanced morphine anti-nociceptive potency while rescuing tolerance. Together, these results suggest that selective inhibition of spinal cord Hsp90 isoforms is a novel, translationally feasible strategy to improve the therapeutic index of opioids.


Subject(s)
Analgesics, Opioid , HSP90 Heat-Shock Proteins , Morphine , Spinal Cord , Animals , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Spinal Cord/metabolism , Spinal Cord/drug effects , Mice , Analgesics, Opioid/pharmacology , Male , Female , Morphine/pharmacology , Protein Isoforms/metabolism , Drug Tolerance , Chronic Pain/drug therapy , Chronic Pain/metabolism , Disease Models, Animal , Injections, Spinal
19.
J Pain ; 25(10): 104610, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38909833

ABSTRACT

Numerous genome-wide association studies have identified risk genes for chronic pain, yet the mechanisms by which genetic variants modify susceptibility have remained elusive. We sought to identify key genes modulating chronic pain risk by regulating brain protein expression. We integrated brain proteomic data with the largest genome-wide dataset for multisite chronic pain (N = 387,649) in a proteome-wide association study (PWAS) using discovery and confirmatory proteomic datasets (N = 376 and 152) from the dorsolateral prefrontal cortex. Leveraging summary data-based Mendelian randomization and Bayesian colocalization analysis, we pinpointed potential causal genes, while a transcriptome-wide association study integrating 452 human brain transcriptomes investigated whether cis-effects on protein abundance extended to the transcriptome. Single-cell RNA-sequencing data and single-nucleus transcriptomic data revealed cell-type-specific expression patterns for identified causal genes in the dorsolateral prefrontal cortex and dorsal root ganglia (DRG), complemented by RNA microarray analysis of expression profiles in other pain-related brain regions. Of the 22 genes cis-regulating protein abundance identified by the discovery PWAS, 18 (82%) were deemed causal by summary data-based Mendelian randomization or Bayesian colocalization analysis analyses, with 7 of these 18 genes (39%) replicating in the confirmatory PWAS, including guanosine diphosphate-mannose pyrophosphorylase B, which also associated at the transcriptome level. Several causal genes exhibited selective expression in excitatory and inhibitory neurons, oligodendrocytes, and astrocytes, while most identified genes were expressed across additional pain-related brain regions. This integrative proteogenomic approach identified 18 high-confidence causal genes for chronic pain, regulated by cis-effects on brain protein levels, suggesting promising avenues for treatment research and indicating a contributory role for the DRG. PERSPECTIVE: The current post genome-wide association study analyses identified 18 high-confidence causal genes regulating chronic pain risk via cis-modulation of brain protein abundance, suggesting promising avenues for future chronic pain therapies. Additionally, the significant expression of these genes in the DRG indicated a potential contributory role, warranting further investigation.


Subject(s)
Brain , Chronic Pain , Genome-Wide Association Study , Proteome , Humans , Chronic Pain/genetics , Chronic Pain/metabolism , Brain/metabolism , Proteome/metabolism , Transcriptome , Proteomics
20.
J Neural Transm (Vienna) ; 131(9): 1079-1094, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38896135

ABSTRACT

Proton magnetic resonance spectroscopy (1H-MRS) has shown inconsistent alterations in the brain metabolites of individuals with chronic pain. We used 3T 1H-MRS to investigate the brain metabolites in the anterior cingulate cortex and thalamus of 22 patients with chronic mild pain and no gait disturbance and 22 healthy controls. The chronic-pain group included patients with chronic low back pain and/or osteoarthritis but none suffering from hypersensitivity. There were no significant between group-differences in glutamate, glutamate plus glutamine (Glx), N-acetylaspartate, glycerophosphorylcholine (GPC), glutamine, creatine plus phosphocreatine, or myo-inositol in the anterior cingulate cortex, but the patients showed a significant decrease in GPC, but not other metabolites, in the thalamus compared to the controls. The GPC values in the patients' thalamus were significantly correlated with pain components on the Short-Form McGill Pain Questionnaire (SF-MPQ-2) and affective empathy components on the Questionnaire of Cognitive and Affective Empathy (QCAE). The GPC in the patients' anterior cingulate cortex showed significant correlations with cognitive empathy components on the QCAE. Myo-inositol in the controls' anterior cingulate cortex and Glx in the patients' thalamus each showed significant relationships with peripheral responsivity on the QCAE. These significances were not significant after Bonferroni corrections. These preliminary findings indicate important roles of GPC, myo-inositol, and Glx in the brain of patients with chronic mild pain.


Subject(s)
Chronic Pain , Empathy , Gyrus Cinguli , Thalamus , Humans , Gyrus Cinguli/metabolism , Gyrus Cinguli/diagnostic imaging , Female , Male , Chronic Pain/metabolism , Chronic Pain/diagnostic imaging , Middle Aged , Thalamus/metabolism , Thalamus/diagnostic imaging , Empathy/physiology , Adult , Inositol/metabolism , Proton Magnetic Resonance Spectroscopy , Aged , Glutamic Acid/metabolism , Glutamine/metabolism , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Pain Measurement
SELECTION OF CITATIONS
SEARCH DETAIL