Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
Pain ; 165(7): e65-e79, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38900144

ABSTRACT

ABSTRACT: Recent evidence highlights the importance of the neuroimmune interface, including periphery-to-central nervous system (CNS) neuroimmune crosstalk, in chronic pain. Although neuroinflammatory processes have been implicated in central sensitization for a long time, their potential neuroprotective and analgesic effects remain relatively elusive. We have explored the relationships between cytokine expression and symptom severity, and candidates for periphery-to-CNS crosstalk. Patients with degenerative disk disease (DDD) (nociceptive pain) or patients with lumbar disk herniation (LDH) with radiculopathy (predominantly neuropathic pain) completed questionnaires regarding pain and functional disability, underwent quantitative sensory testing, and provided blood and cerebrospinal fluid (CSF) samples. Proximity extension assay (PEA) was used to measure the levels of 92 inflammatory proteins in the CSF and serum from a total of 160 patients and controls, and CSF/serum albumin quotients was calculated for patients with DDD and patients with LDH. We found signs of neuroimmune activation, in the absence of systemic inflammation. Regarding periphery-to-CNS neuroimmune crosstalk, there were significant associations between several cytokines and albumin quotient, despite the latter being primarily at subclinical levels. The cytokines CCL11, CD5, IL8, and MMP-10 were elevated in the CSF, had positive correlations between CSF and serum levels, and associated in a nonlinear manner with back, but not leg, pain intensity in the LDH, but not the DDD, group. In conclusion, we found evidence for neuroimmune activation in the CNS of both patient groups in the absence of systemic inflammation and signs of a communication between CSF and serum. Complex and disease-specific associations were found between cytokines in CSF and back pain intensity.


Subject(s)
Chronic Pain , Cytokines , Intervertebral Disc Degeneration , Intervertebral Disc Displacement , Humans , Male , Female , Cytokines/cerebrospinal fluid , Cytokines/blood , Middle Aged , Intervertebral Disc Displacement/cerebrospinal fluid , Intervertebral Disc Displacement/complications , Intervertebral Disc Displacement/immunology , Intervertebral Disc Degeneration/cerebrospinal fluid , Intervertebral Disc Degeneration/immunology , Adult , Chronic Pain/cerebrospinal fluid , Chronic Pain/immunology , Chronic Pain/blood , Aged , Lumbar Vertebrae , Pain Measurement/methods , Neuroimmunomodulation/physiology
2.
Front Immunol ; 15: 1387142, 2024.
Article in English | MEDLINE | ID: mdl-38807587

ABSTRACT

Introduction: Chronic Pelvic Pain Syndrome or Chronic Prostatitis (CPPS/CP) is the most prevalent urologic affliction among young adult men. It is a challenging condition to treat, which significantly decreases patient quality of life, mostly because of its still uncertain aetiology. In that regard, an autoimmune origin is a prominent supported theory. Indeed, studies in patients and in rodent models of Experimental Autoimmune Prostatitis (EAP) have provided compelling evidence suggesting a key role of CD4 Th1 cells in disease pathogenesis. However, the implication of other prominent effectors of the immune system, such as CD8 T cells, has yet to be studied. Methods: We herein analyzed the induction of prostatitis and the development of chronic pelvic pain in EAP using CD8 T cell-deficient animals. Results: We found similarly elevated PA-specific immune responses, with high frequencies of specific IFNg+CD4+ and IL17+CD4+ T cells in prostate draining lymph nodes from PA-immunized either CD8 KO or wild type animals with respect to controls. Moreover, these peripheral immune responses were paralleled by the development of significant chronic pelvic pain, and accompanied by prostate histological lesions, characterized by hemorrhage, epithelial cell desquamation, marked periglandular leukocyte infiltration, and increased collagen deposition in both, PA-immunized CD8 KO and wild type animals. As expected, control animals did not develop prostate histological lesions. Discussion: Our results indicate that CD8 T cells do not play a major role in EAP pathogenesis and chronic pelvic pain development. Moreover, our results corroborate the previous notion that a CD4 Th1 associated immune response drives the induction of prostate tissue inflammation and the development of chronic pelvic pain.


Subject(s)
Autoimmune Diseases , CD8-Positive T-Lymphocytes , Disease Models, Animal , Mice, Knockout , Pelvic Pain , Prostatitis , Prostatitis/immunology , Prostatitis/pathology , Male , Animals , CD8-Positive T-Lymphocytes/immunology , Pelvic Pain/immunology , Autoimmune Diseases/immunology , Mice , Chronic Pain/immunology , CD4-Positive T-Lymphocytes/immunology , Mice, Inbred C57BL , Prostate/immunology , Prostate/pathology
3.
Sci Rep ; 12(1): 215, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34997115

ABSTRACT

Disability and distress caused by chronic low back pain (LBP) lacking clear pathoanatomical explanations cause huge problems both for patients and society. A subgroup of patients has Modic changes (MC), identifiable by MRI as vertebral bone marrow lesions. The cause of such changes and their relationship to pain are not yet understood. We explored the pathobiology of these lesions using profiling of gene expression in blood, coupled with an edema-sensitive MRI technique known as short tau inversion recovery (STIR) imaging. STIR images and total RNA from blood were collected from 96 patients with chronic LBP and MC type I, the most inflammatory MC state. We found the expression of 37 genes significantly associated with STIR signal volume, ten genes with edema abundancy (a constructed combination of STIR signal volume, height, and intensity), and one gene with expression levels significantly associated with maximum STIR signal intensity. Gene sets related to interferon signaling, mitochondrial metabolism and defense response to virus were identified as significantly enriched among the upregulated genes in all three analyses. Our results point to inflammation and immunological defense as important players in MC biology in patients with chronic LBP.


Subject(s)
Bone Marrow/diagnostic imaging , Chronic Pain/diagnostic imaging , Gene Expression Profiling , Low Back Pain/diagnostic imaging , Magnetic Resonance Imaging , Spine/diagnostic imaging , Transcriptome , Adult , Bone Marrow/immunology , Chronic Pain/genetics , Chronic Pain/immunology , Female , Gene Expression Regulation , Humans , Low Back Pain/genetics , Low Back Pain/immunology , Male , Middle Aged , Predictive Value of Tests , Randomized Controlled Trials as Topic , Spine/immunology
4.
Int J Mol Sci ; 22(24)2021 Dec 19.
Article in English | MEDLINE | ID: mdl-34948407

ABSTRACT

Non-opioid single-chain variable fragment (scFv) small antibodies were generated as pain-reducing block of P2X4R receptor (P2X4R). A panel of scFvs targeting an extracellular peptide sequence of P2X4R was generated followed by cell-free ribosome display for recombinant antibody selection. After three rounds of bio-panning, a panel of recombinant antibodies was isolated and characterized by ELISA, cross-reactivity analysis, and immunoblotting/immunostaining. Generated scFv antibodies feature binding activity similar to monoclonal antibodies but with stronger affinity and increased tissue penetrability due to their ~30% smaller size. Two anti-P2X4R scFv clones (95, 12) with high specificity and affinity binding were selected for in vivo testing in male and female mice with trigeminal nerve chronic neuropathic pain (FRICT-ION model) persisting for several months in untreated BALBc mice. A single dose of P2X4R scFv (4 mg/kg, i.p.) successfully, completely, and permanently reversed chronic neuropathic pain-like measures in male mice only, providing retention of baseline behaviors indefinitely. Untreated mice retained hypersensitivity, and developed anxiety- and depression-like behaviors within 5 weeks. In vitro P2X4R scFv 95 treatment significantly increased the rheobase of larger-diameter (>25 µm) trigeminal ganglia (TG) neurons from FRICT-ION mice compared to controls. The data support use of engineered scFv antibodies as non-opioid biotherapeutic interventions for chronic pain.


Subject(s)
Chronic Pain/drug therapy , Purinergic P2X Receptor Antagonists/therapeutic use , Single-Chain Antibodies/therapeutic use , Animals , Antibody Affinity , Cells, Cultured , Chronic Pain/immunology , Female , Male , Mice , Peptide Library , Purinergic P2X Receptor Antagonists/chemistry , Purinergic P2X Receptor Antagonists/immunology , Receptors, Purinergic P2X4/chemistry , Receptors, Purinergic P2X4/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology
5.
Cell Rep ; 37(12): 110152, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34936870

ABSTRACT

Chronic pain is a prevalent medical problem, and its molecular basis remains poorly understood. Here, we demonstrate the significance of the transmembrane protein (Tmem) 160 for nerve injury-induced neuropathic pain. An extensive behavioral assessment suggests a pain modality- and entity-specific phenotype in male Tmem160 global knockout (KO) mice: delayed establishment of tactile hypersensitivity and alterations in self-grooming after nerve injury. In contrast, Tmem160 seems to be dispensable for other nerve injury-induced pain modalities, such as non-evoked and movement-evoked pain, and for other pain entities. Mechanistically, we show that global KO males exhibit dampened neuroimmune signaling and diminished TRPA1-mediated activity in cultured dorsal root ganglia. Neither these changes nor altered pain-related behaviors are observed in global KO female and male peripheral sensory neuron-specific KO mice. Our findings reveal Tmem160 as a sexually dimorphic factor contributing to the establishment, but not maintenance, of discrete nerve injury-induced pain behaviors in male mice.


Subject(s)
Cytokines/metabolism , Membrane Proteins/immunology , Membrane Proteins/metabolism , Neuralgia/immunology , Neuralgia/metabolism , Peripheral Nerve Injuries/metabolism , Sensory Receptor Cells/metabolism , Animals , Behavior, Animal , Chronic Pain/immunology , Chronic Pain/metabolism , Female , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Inflammation , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroimmunomodulation , Neuropsychological Tests , Signal Transduction , TRPA1 Cation Channel/metabolism
6.
Eur J Pharmacol ; 913: 174619, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34748768

ABSTRACT

In some chronic primary pain conditions such as temporomandibular disorder (TMD) and fibromyalgia syndrome (FMS), mild or chronic stress enhances pain. TMD and FMS often occur together, but the underlying mechanisms are unclear. The purpose of this study was to investigate the role of cholecystokinin (CCK) in the spinal cord in somatic hyperalgesia induced by orofacial inflammation combined with stress. Somatic hyperalgesia was detected by the thermal withdrawal latency and mechanical withdrawal threshold. The expression of CCK1 receptors, CCK2 receptors, ERK1/2 and p-ERK1/2 in the spinal cord was examined by Western blot. After the stimulation of orofacial inflammation combined with 3 day forced swim, the expression of CCK2 receptors and p-ERK1/2 protein in the L4-L5 spinal dorsal horn increased significantly, while the expression of CCK1 receptors and ERK1/2 protein remained unchanged. Intrathecal injection of the CCK2 receptor antagonist YM-022 or mitogen-activated protein kinase (MAPK) kinase (MEK) inhibitor PD98059 blocked somatic hyperalgesia induced by orofacial inflammation combined with stress. Intrathecal administration of the MEK inhibitor blocked somatic sensitization caused by the CCK receptor agonist CCK8. The CCK2 receptor antagonist YM-022 significantly reduced the expression of p-ERK1/2. These data indicate that upregulation of CCK2 receptors through the MAPK pathway contributes to somatic hyperalgesia in this comorbid pain model. Thus, CCK2 receptors and MAPK pathway may be potential targets for the treatment of TMD comorbid with FMS.


Subject(s)
Cholecystokinin/metabolism , Chronic Pain/immunology , Facial Pain/immunology , Hyperalgesia/immunology , Stress, Psychological/complications , Animals , Chronic Pain/pathology , Disease Models, Animal , Facial Pain/pathology , Female , Humans , Hyperalgesia/pathology , Inflammation/immunology , Inflammation/pathology , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin B/metabolism , Spinal Cord Dorsal Horn/immunology , Spinal Cord Dorsal Horn/pathology , Stress, Psychological/immunology , Stress, Psychological/psychology
7.
Front Immunol ; 12: 783725, 2021.
Article in English | MEDLINE | ID: mdl-34804074

ABSTRACT

Interferons (IFNs) are cytokines that possess antiviral, antiproliferative, and immunomodulatory actions. IFN-α and IFN-ß are two major family members of type-I IFNs and are used to treat diseases, including hepatitis and multiple sclerosis. Emerging evidence suggests that type-I IFN receptors (IFNARs) are also expressed by microglia, astrocytes, and neurons in the central and peripheral nervous systems. Apart from canonical transcriptional regulations, IFN-α and IFN-ß can rapidly suppress neuronal activity and synaptic transmission via non-genomic regulation, leading to potent analgesia. IFN-γ is the only member of the type-II IFN family and induces central sensitization and microglia activation in persistent pain. We discuss how type-I and type-II IFNs regulate pain and infection via neuro-immune modulations, with special focus on neuroinflammation and neuro-glial interactions. We also highlight distinct roles of type-I IFNs in the peripheral and central nervous system. Insights into IFN signaling in nociceptors and their distinct actions in physiological vs. pathological and acute vs. chronic conditions will improve our treatments of pain after surgeries, traumas, and infections.


Subject(s)
Acute Pain/immunology , Chronic Pain/immunology , Interferon Type I/metabolism , Interferon-gamma/metabolism , Neuroinflammatory Diseases/immunology , Acute Pain/pathology , Animals , Chronic Pain/pathology , Disease Models, Animal , Humans , Neuroglia/cytology , Neuroglia/immunology , Neuroglia/pathology , Neuroinflammatory Diseases/pathology , Nociceptors/immunology , Nociceptors/metabolism , Receptors, Interferon/metabolism , Signal Transduction/immunology , Spinal Cord/cytology , Spinal Cord/immunology , Spinal Cord/pathology
8.
Neurosci Lett ; 764: 136205, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34478818

ABSTRACT

Lactate transport is an important means of communication between astrocytes and neurons and is implicated in a variety of neurobiological processes. However, the connection between astrocyte-neuron lactate transport and nociceptive modulation has not been well established. Here, we found that Complete Freund's adjuvant (CFA)-induced inflammation pain leads to a significant increase in extracellular lactate levels in the anterior cingulate cortex (ACC). Inhibition of glycogenolysis and lactate release in the ACC disrupted the persistent, but not acute, inflammation pain induced by CFA, and this effect was reversed by exogenous L-lactate administration. Knocking down the expression of lactate transporters (MCT1, MCT4, or MCT2) also disrupted the long lasting inflammation pain induced by CFA. Moreover, glycogenolysis in the ACC is critical for the induction of molecular changes related to neuronal plasticity, including the induction of phospho- (p-) ERK, p-CREB, and Fos. Taken together, our findings indicate that astrocyte-neuron lactate transport in the ACC is critical for the occurrence of persistent inflammation pain, suggesting a novel mechanism underlying chronic pain.


Subject(s)
Arabinose/pharmacology , Cell Communication/immunology , Chronic Pain/immunology , Gyrus Cinguli/pathology , Imino Furanoses/pharmacology , Lactic Acid/metabolism , Sugar Alcohols/pharmacology , Animals , Arabinose/therapeutic use , Astrocytes/metabolism , Cell Communication/drug effects , Chronic Pain/drug therapy , Chronic Pain/pathology , Disease Models, Animal , Freund's Adjuvant/administration & dosage , Freund's Adjuvant/immunology , Glycogenolysis/drug effects , Glycogenolysis/immunology , Gyrus Cinguli/cytology , Gyrus Cinguli/drug effects , Gyrus Cinguli/immunology , Humans , Imino Furanoses/therapeutic use , Male , Mice , Neuronal Plasticity/drug effects , Neuronal Plasticity/immunology , Neurons/metabolism , Sugar Alcohols/therapeutic use
9.
Int Immunopharmacol ; 98: 107882, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34182245

ABSTRACT

Daphnetin (7, 8-dihydroxycoumarin, DAPH), a coumarin derivative isolated from Daphne odora var., recently draws much more attention as a promising drug candidate to treat neuroinflammatory diseases due to its protective effects against neuroinflammation. However, itscontribution to chronic inflammatory pain is largely unknown. In the current work, we investigated the effects of DAPH in a murine model of inflammatory pain induced by complete Freund's adjuvant (CFA) and its possible underlying mechanisms. Our results showed that DAPH treatment significantly attenuated mechanical allodynia provoked by CFA. A profound inhibition of spinal glial activation, followed by attenuated expression levels of spinal pro-inflammatory cytokines, was observed in DAPH-treated inflammatory pain mice. Further study demonstrated that DAPH mediated negative regulation of spinal NF-κB pathway, as well as its preferential activation of Nrf2/HO-1 signaling pathway in inflammatory pain mice. This study, for the first time, indicated that DAPH might preventthe development of mechanical allodynia in mice with inflammatory pain. And more importantly, these data provide evidence for the potential application of DAPH in the treatment of chronic inflammatory pain.


Subject(s)
Chronic Pain/drug therapy , Hyperalgesia/drug therapy , Pain/drug therapy , Umbelliferones/pharmacology , Animals , Chronic Pain/immunology , Chronic Pain/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Freund's Adjuvant/administration & dosage , Freund's Adjuvant/immunology , Heme Oxygenase-1/metabolism , Humans , Hyperalgesia/immunology , Hyperalgesia/pathology , Male , Membrane Proteins/metabolism , Mice , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Neuroglia/drug effects , Neuroglia/immunology , Neuroglia/pathology , Oxidative Stress/drug effects , Oxidative Stress/immunology , Pain/immunology , Pain/pathology , Signal Transduction/drug effects , Signal Transduction/immunology , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/pathology , Umbelliferones/therapeutic use
12.
J Pain ; 22(10): 1146-1179, 2021 10.
Article in English | MEDLINE | ID: mdl-33892151

ABSTRACT

During persistent pain, the dorsal spinal cord responds to painful inputs from the site of injury, but the molecular modulatory processes have not been comprehensively examined. Using transcriptomics and multiplex in situ hybridization, we identified the most highly regulated receptors and signaling molecules in rat dorsal spinal cord in peripheral inflammatory and post-surgical incisional pain models. We examined a time course of the response including acute (2 hours) and longer term (2 day) time points after peripheral injury representing the early onset and instantiation of hyperalgesic processes. From this analysis, we identify a key population of superficial dorsal spinal cord neurons marked by somatotopic upregulation of the opioid neuropeptide precursor prodynorphin, and 2 receptors: the neurokinin 1 receptor, and anaplastic lymphoma kinase. These alterations occur specifically in the glutamatergic subpopulation of superficial dynorphinergic neurons. In addition to specific neuronal gene regulation, both models showed induction of broad transcriptional signatures for tissue remodeling, synaptic rearrangement, and immune signaling defined by complement and interferon induction. These signatures were predominantly induced ipsilateral to tissue injury, implying linkage to primary afferent drive. We present a comprehensive set of gene regulatory events across 2 models that can be targeted for the development of non-opioid analgesics. PERSPECTIVE: The deadly impact of the opioid crisis and the need to replace morphine and other opioids in clinical practice is well recognized. Embedded within this research is an overarching goal of obtaining foundational knowledge from transcriptomics to search for non-opioid analgesic targets. Developing such analgesics would address unmet clinical needs.


Subject(s)
Anaplastic Lymphoma Kinase/metabolism , Chronic Pain/metabolism , Hyperalgesia/metabolism , Neuroinflammatory Diseases/metabolism , Peripheral Nerve Injuries/metabolism , Posterior Horn Cells/metabolism , Transcriptome/physiology , Animals , Chronic Pain/immunology , Disease Models, Animal , Hyperalgesia/immunology , Neuroinflammatory Diseases/immunology , Peripheral Nerve Injuries/immunology , Posterior Horn Cells/immunology , Rats , Sequence Analysis, RNA
13.
Front Immunol ; 12: 641588, 2021.
Article in English | MEDLINE | ID: mdl-33692810

ABSTRACT

Chronic neuropathic pain (CNP) is caused by a lesion or disease of the somatosensory nervous system. It affects ~8% of the general population and negatively impacts a person's level of functioning and quality of life. Its resistance to available pain therapies makes CNP a major unmet medical need. Immune cells have been shown to play a role for development, maintenance and recovery of CNP and therefore are attractive targets for novel pain therapies. In particular, in neuropathic mice and humans, microglia are activated in the dorsal horn and peripheral immune cells infiltrate the nervous system to promote chronic neuroinflammation and contribute to the initiation and progression of CNP. Importantly, immunity not only controls pain development and maintenance, but is also essential for pain resolution. In particular, regulatory T cells, a subpopulation of T lymphocytes with immune regulatory function, and macrophages were shown to be important contributors to pain recovery. In this review we summarize the interactions of the peripheral immune system with the nervous system and outline their contribution to the development and recovery of pain.


Subject(s)
Chronic Pain/immunology , Immunologic Memory , Macrophages/immunology , Neuralgia/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Chronic Pain/pathology , Chronic Pain/therapy , Humans , Macrophages/pathology , Neuralgia/pathology , Neuralgia/therapy , T-Lymphocytes, Regulatory/pathology
14.
Int J Mol Sci ; 22(3)2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33535595

ABSTRACT

Pain can be induced by tissue injuries, diseases and infections. The interactions between the peripheral nervous system (PNS) and immune system are primary actions in pain sensitizations. In response to stimuli, nociceptors release various mediators from their terminals that potently activate and recruit immune cells, whereas infiltrated immune cells further promote sensitization of nociceptors and the transition from acute to chronic pain by producing cytokines, chemokines, lipid mediators and growth factors. Immune cells not only play roles in pain production but also contribute to PNS repair and pain resolution by secreting anti-inflammatory or analgesic effectors. Here, we discuss the distinct roles of four major types of immune cells (monocyte/macrophage, neutrophil, mast cell, and T cell) acting on the PNS during pain process. Integration of this current knowledge will enhance our understanding of cellular changes and molecular mechanisms underlying pain pathogenies, providing insights for developing new therapeutic strategies.


Subject(s)
Chronic Pain/blood , Chronic Pain/immunology , Immune System , Neuroimmunomodulation/physiology , Peripheral Nervous System/immunology , Animals , Humans , Inflammation , Macrophages/cytology , Macrophages/metabolism , Mast Cells/cytology , Mice , Monocytes/cytology , Neutrophils/cytology , Neutrophils/metabolism , Nociception , Pain Management , Peripheral Nervous System/metabolism , Signal Transduction , T-Lymphocytes/cytology
15.
Cytokine ; 141: 155440, 2021 05.
Article in English | MEDLINE | ID: mdl-33550164

ABSTRACT

BACKGROUND: As one of the most common conditions in urological outpatients, chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) puzzles many individuals because of its unclear etiology and lack of effective treatment. Recently, immunological alterations underpinning CP/CPPS have been extensively investigated. METHODS: The PubMed, Web of Science, Cochrane library, and EMBASE databases were used to search original articles on immune mediators in patients with CP/CPPS and in experimental autoimmune prostatitis (EAP) models through April 10, 2020. Standardized mean differences (SMD) were calculated to summarize the differences in immune mediator levels between groups. Funnel plot, Begg's funnel plot, Egger's regression test, and the sensitivity analysis were applied to determine and visualize the stability of our findings. RESULTS: A total of 34 original studies were included in the meta-analysis, including 24 studies on patients with CP/CPPS and 10 studies on EAP models. We found that TNF-α, IL-1ß, IL-6, and IL-8 were the four immune mediators that elevated in most of the samples derived from patients with CP/CPPS and the EAP models. The adjusted publication bias analysis indicated that publication bias was not existed, and the sensitivity analyses showed that the results were stable. CONCLUSIONS: Immune responses play significant roles during the pathogenesis of CP/CPPS by promoting intraprostatic inflammation. Our findings provide potential diagnostic and therapeutic targets for CP/CPPS patients.


Subject(s)
Chronic Pain/immunology , Cytokines/immunology , Pelvic Pain/immunology , Prostatitis/immunology , Animals , Chronic Disease , Chronic Pain/pathology , Disease Models, Animal , Humans , Male , Pelvic Pain/pathology , Prostatitis/pathology
16.
Trends Mol Med ; 27(4): 302-313, 2021 04.
Article in English | MEDLINE | ID: mdl-33431239

ABSTRACT

Despite successful research efforts aimed at understanding pain mechanisms, there is still no adequate treatment for many patients suffering from chronic pain. The contribution of neuroinflammation to chronic pain is widely acknowledged. Here, we summarize findings indicating that T cells play a key role in the suppression of pain. An active contribution of the immune system to resolution of pain may explain why immunosuppressive drugs are often not sufficient to control pain. This would also imply that dysregulation of certain immune functions promote transition to chronic pain. Conversely, stimulating the endogenous immune-mediated resolution pathways may provide a potent approach to treat chronic pain.


Subject(s)
Pain , T-Lymphocytes/immunology , Chronic Pain/immunology , Chronic Pain/physiopathology , Chronic Pain/therapy , Humans , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/physiopathology , Pain/immunology , Pain/physiopathology
17.
J Urol ; 205(4): 1133-1138, 2021 04.
Article in English | MEDLINE | ID: mdl-33347771

ABSTRACT

PURPOSE: We sought to determine whether pollen triggers urological chronic pelvic pain syndrome flares. MATERIALS AND METHODS: We assessed flare status every 2 weeks for 1 year as part of the Multidisciplinary Approach to the Study of Chronic Pelvic Pain case-crossover analysis of flare triggers (NCT01098279). Flare symptoms, flare start date and exposures in the 3 days before a flare were queried for the first 3 flares and at 3 randomly selected nonflare times. These data were linked to daily pollen count by date and the first 3 digits of participants' zip codes. Pollen count in the 3 days before and day of a flare, as well as pollen rises past established thresholds, were compared to nonflare values by conditional logistic regression. Poisson regression was used to estimate flare rates in the 3 weeks following pollen rises past established thresholds in the full longitudinal study. Analyses were performed in all participants and separately in those who reported allergies or respiratory tract disorders. RESULTS: Although no associations were observed for daily pollen count and flare onset, positive associations were observed for pollen count rises past medium or higher thresholds in participants with allergies or respiratory tract disorders in the case-crossover (OR 1.31, 95% CI 1.04-1.66) and full longitudinal (RR 1.23, 95% CI 1.03-1.46) samples. CONCLUSIONS: We found some evidence to suggest that rising pollen count may trigger flares of urological chronic pelvic pain syndrome. If confirmed in future studies, these findings may help to inform flare pathophysiology, prevention and treatment, and control over the unpredictability of flares.


Subject(s)
Chronic Pain/immunology , Cystitis, Interstitial/immunology , Pelvic Pain/immunology , Pollen/immunology , Prostatitis/immunology , Symptom Flare Up , Adult , Aged , Aged, 80 and over , Cross-Over Studies , Female , Humans , Longitudinal Studies , Male , Middle Aged , Syndrome , United States
18.
Neuroscientist ; 27(2): 113-128, 2021 04.
Article in English | MEDLINE | ID: mdl-32441204

ABSTRACT

Mounting evidence indicates that disruptions in bidirectional communication pathways between the central nervous system (CNS) and peripheral immune system underlie the etiology of pathologic pain conditions. The purpose of this review is to focus on the cross-talk between these two systems in mediating nociceptive circuitry under various conditions, including nervous system disorders. Elevated and prolonged proinflammatory signaling in the CNS is argued to play a role in psychiatric illnesses and chronic pain states. Here we review current research on the dynamic interplay between altered nociceptive mechanisms, both peripheral and central, and physiological and behavioral changes associated with CNS disorders.


Subject(s)
Chronic Pain/immunology , Chronic Pain/psychology , Inflammation Mediators/immunology , Neuroimmunomodulation/physiology , Stress, Psychological/immunology , Stress, Psychological/psychology , Animals , Brain/immunology , Brain/metabolism , Chronic Pain/metabolism , Humans , Inflammation Mediators/metabolism , Spinal Cord/immunology , Spinal Cord/metabolism , Stress, Psychological/metabolism
19.
J Neurosci ; 41(5): 855-865, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33239404

ABSTRACT

Chronic pain, encompassing conditions, such as low back pain, arthritis, persistent post-surgical pain, fibromyalgia, and neuropathic pain disorders, is highly prevalent but remains poorly treated. The vast majority of therapeutics are directed solely at neurons, despite the fact that signaling between immune cells, glia, and neurons is now recognized as indispensable for the initiation and maintenance of chronic pain. This review highlights recent advances in understanding fundamental neuroimmune signaling mechanisms and novel therapeutic targets in rodent models of chronic pain. We further discuss new technological developments to study, diagnose, and quantify neuroimmune contributions to chronic pain in patient populations.


Subject(s)
Autoantibodies/immunology , Chronic Pain/immunology , Disease Models, Animal , Neuroglia/immunology , Neuroimmunomodulation/physiology , Neurons/immunology , Animals , Autoantibodies/metabolism , Chronic Pain/metabolism , Humans , NF-E2-Related Factor 2/immunology , NF-E2-Related Factor 2/metabolism , Neuroglia/metabolism , Neurons/metabolism , Rodentia
20.
Biol Sex Differ ; 11(1): 62, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33183347

ABSTRACT

Chronic pain occurs with greater frequency in women, with a parallel sexually dimorphic trend reported in sufferers of many autoimmune diseases. There is a need to continue examining neuro-immune-endocrine crosstalk in the context of sexual dimorphisms in chronic pain. Several phenomena in particular need to be further explored. In patients, autoantibodies to neural antigens have been associated with sensory pathway hyper-excitability, and the role of self-antigens released by damaged nerves remains to be defined. In addition, specific immune cells release pro-nociceptive cytokines that directly influence neural firing, while T lymphocytes activated by specific antigens secrete factors that either support nerve repair or exacerbate the damage. Modulating specific immune cell populations could therefore be a means to promote nerve recovery, with sex-specific outcomes. Understanding biological sex differences that maintain, or fail to maintain, neuroimmune homeostasis may inform the selection of sex-specific treatment regimens, improving chronic pain management by rebalancing neuroimmune feedback. Given the significance of interactions between nerves and immune cells in the generation and maintenance of neuropathic pain, this review focuses on sex differences and possible links with persistent autoimmune activity using sciatica as an example.


Subject(s)
Autoimmunity , Chronic Pain/immunology , Neuroimmunomodulation , Sciatica/immunology , Sex Characteristics , Animals , Chronic Pain/genetics , Gonadal Steroid Hormones/physiology , Humans , Sciatica/genetics , Sex Chromosomes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...