Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 324
Filter
1.
Immunity ; 52(5): 753-766, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32433948

ABSTRACT

Although the medical definition of itch has been in existence for 360 years, only in the last 20 years have we begun to understand the basic mechanisms that underlie this unique sensation. Therapeutics that specifically target chronic itch as a pathologic entity are currently still not available. Recent seminal advances in itch circuitry within the nervous system have intersected with discoveries in immunology in unexpected ways to rapidly inform emerging treatment strategies. The current review aims to introduce these basic concepts in itch biology and highlight how distinct immunologic pathways integrate with recently identified itch-sensory circuits in the nervous system to inform a major new paradigm of neuroimmunology and therapeutic development for chronic itch.


Subject(s)
Ganglia, Spinal/immunology , Pruritus/immunology , Sensory Receptor Cells/immunology , Skin/immunology , Somatosensory Cortex/immunology , Animals , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Humans , Models, Immunological , Models, Neurological , Pruritus/diagnosis , Pruritus/physiopathology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Signal Transduction/immunology , Signal Transduction/physiology , Skin/innervation , Somatosensory Cortex/physiopathology
2.
J Virol ; 98(5): e0159623, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38587378

ABSTRACT

Following acute herpes simplex virus type 2 (HSV-2) infection, the virus undergoes an asymptomatic latent infection of sensory neurons of dorsal root ganglia (DRG). Chemical and physical stress cause intermittent virus reactivation from latently infected DRG and recurrent virus shedding in the genital mucosal epithelium causing genital herpes in symptomatic patients. While T cells appear to play a role in controlling virus reactivation from DRG and reducing the severity of recurrent genital herpes, the mechanisms for recruiting these T cells into DRG and the vaginal mucosa (VM) remain to be fully elucidated. The present study investigates the effect of CXCL9, CXCL10, and CXCL11 T-cell-attracting chemokines on the frequency and function of DRG- and VM-resident CD4+ and CD8+ T cells and its effect on the frequency and severity of recurrent genital herpes in the recurrent herpes guinea pig model. HSV-2 latent-infected guinea pigs were immunized intramuscularly with the HSV-2 ribonucleotide reductase 2 (RR2) protein (Prime) and subsequently treated intravaginally with the neurotropic adeno-associated virus type 8 expressing CXCL9, CXCL10, or CXCL11 chemokines to recruit CD4+ and CD8+ T cells into the infected DRG and VM (Pull). Compared to the RR2 therapeutic vaccine alone, the RR2/CXCL11 prime/pull therapeutic vaccine significantly increased the frequencies of functional tissue-resident and effector memory CD4+ and CD8+ T cells in both DRG and VM tissues. This was associated with less virus in the healed genital mucosal epithelium and reduced frequency and severity of recurrent genital herpes. These findings confirm the role of local DRG- and VM-resident CD4+ and CD8+ T cells in reducing virus shedding at the vaginal site of infection and the severity of recurrent genital herpes and propose the novel prime-pull vaccine strategy to protect against recurrent genital herpes.IMPORTANCEThe present study investigates the novel prime/pull therapeutic vaccine strategy to protect against recurrent genital herpes using the latently infected guinea pig model. In this study, we used the strategy that involves immunization of herpes simplex virus type 2-infected guinea pigs using a recombinantly expressed herpes tegument protein-ribonucleotide reductase 2 (RR2; prime), followed by intravaginal treatment with the neurotropic adeno-associated virus type 8 expressing CXCL9, CXCL10, or CXCL11 T-cell-attracting chemokines to recruit T cells into the infected dorsal root ganglia (DRG) and vaginal mucosa (VM) (pull). We show that the RR2/CXCL11 prime-pull therapeutic vaccine strategy elicited a significant reduction in virus shedding in the vaginal mucosa and decreased the severity and frequency of recurrent genital herpes. This protection was associated with increased frequencies of functional tissue-resident (TRM cells) and effector (TEM cells) memory CD4+ and CD8+ T cells infiltrating latently infected DRG tissues and the healed regions of the vaginal mucosa. These findings shed light on the role of tissue-resident and effector memory CD4+ and CD8+ T cells in DRG tissues and the VM in protection against recurrent genital herpes and propose the prime-pull therapeutic vaccine strategy in combating genital herpes.


Subject(s)
Chemokine CXCL11 , Herpes Genitalis , Herpesvirus 2, Human , Ribonucleotide Reductases , Animals , Female , Guinea Pigs , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chemokine CXCL11/immunology , Chemokine CXCL11/metabolism , Disease Models, Animal , Ganglia, Spinal/immunology , Ganglia, Spinal/virology , Herpes Genitalis/immunology , Herpes Genitalis/prevention & control , Herpesvirus 2, Human/immunology , Memory T Cells/immunology , Ribonucleotide Reductases/metabolism , Vaccination , Vagina/virology , Vagina/immunology
3.
Clin Exp Rheumatol ; 42(6): 1141-1149, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38607678

ABSTRACT

Fibromyalgia (FM) remains a condition with a pathogenesis that is not completely understood, affecting a significant portion of the global population. This article summarises the main advances in FM during the last year. Even in 2023, research on FM was notably active. From a clinimetric perspective, studies have been conducted to evaluate the possibilities of interchanging the primary indices of disease severity, primarily for studies with substantial case numbers. Regarding FM pathogenesis, ongoing research focuses on small fiber neuropathy: some studies have documented its association with central sensitisation, while others have revealed distinct sensory profiles in patients with FM and small fiber neuropathy compared to those solely with small fiber neuropathy. Dorsal root ganglia seem to play a crucial role in the pathogenesis of FM as they host satellite glial cells, which are targeted by pain-driving immunoglobulin G. These antibodies have been identified in a subset of patients exhibiting high symptom severity. An important study conducted on animal models confirmed the role of neuroinflammation at the level of dorsal root ganglia, in this case mediated by polymorphonuclear neutrophils. Mounting evidence underscores the link between COVID-19 and the persistence of FM symptoms after recovery. In identifying potential biomarkers aiding FM diagnosis, research has also concentrated on studying the expression of specific circulating microRNAs. Recent discoveries have unveiled novel therapeutic strategies for FM, especially focused in non-pharmacological interventions. This includes a focus on non-invasive brain stimulation and exercise programs, all directed towards relieving symptoms and improving functionality in individuals affected by the condition.


Subject(s)
COVID-19 , Fibromyalgia , Fibromyalgia/diagnosis , Fibromyalgia/therapy , Fibromyalgia/physiopathology , Fibromyalgia/immunology , Humans , COVID-19/complications , COVID-19/immunology , COVID-19/diagnosis , Animals , SARS-CoV-2/immunology , Ganglia, Spinal/physiopathology , Ganglia, Spinal/immunology , Ganglia, Spinal/metabolism , Severity of Illness Index , Biomarkers/blood
4.
Inflamm Res ; 71(2): 187-190, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34940887

ABSTRACT

OBJECTIVE: We investigated whether it is possible to induce a state of "LPS-sensitization" in neurons of primary cultures from rat dorsal root ganglia by pre-treatment with ultra-low doses of LPS. METHODS: DRG primary cultures were pre-treated with low to ultra-low doses of LPS (0.001-0.1 µg/ml) for 18 h, followed by a short-term stimulation with a higher LPS-dose (10 µg/ml for 2 h). TNF-α in the supernatants was measured as a sensitive read out. Using the fura-2 340/380 nm ratio imaging technique, we further investigated the capsaicin-evoked Ca2+-signals in neurons from DRG, which were pre-treated with a wide range of LPS-doses. RESULTS: Release of TNF-α evoked by stimulation with 10 µg/ml LPS into the supernatant was not significantly modified by pre-exposure to low to ultra-low LPS-doses. Capsaicin-evoked Ca2+-signals were significantly enhanced by pre-treatment with LPS doses being above a certain threshold. CONCLUSION: Ultra-low doses of LPS, which per se do not evoke a detectable inflammatory response, are not sufficient to sensitize neurons (Ca2+-responses) and glial elements (TNF-α-responses) of the primary afferent somatosensory system.


Subject(s)
Ganglia, Spinal/drug effects , Lipopolysaccharides/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Calcium Signaling/drug effects , Capsaicin/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Ganglia, Spinal/immunology , Rats , Rats, Wistar
5.
Nature ; 533(7604): 552-6, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27225131

ABSTRACT

Circulating antibodies can access most tissues to mediate surveillance and elimination of invading pathogens. Immunoprivileged tissues such as the brain and the peripheral nervous system are shielded from plasma proteins by the blood-brain barrier and blood-nerve barrier, respectively. Yet, circulating antibodies must somehow gain access to these tissues to mediate their antimicrobial functions. Here we examine the mechanism by which antibodies gain access to neuronal tissues to control infection. Using a mouse model of genital herpes infection, we demonstrate that both antibodies and CD4 T cells are required to protect the host after immunization at a distal site. We show that memory CD4 T cells migrate to the dorsal root ganglia and spinal cord in response to infection with herpes simplex virus type 2. Once inside these neuronal tissues, CD4 T cells secrete interferon-γ and mediate local increase in vascular permeability, enabling antibody access for viral control. A similar requirement for CD4 T cells for antibody access to the brain is observed after intranasal challenge with vesicular stomatitis virus. Our results reveal a previously unappreciated role of CD4 T cells in mobilizing antibodies to the peripheral sites of infection where they help to limit viral spread.


Subject(s)
Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , Nervous System/immunology , Animals , B-Lymphocytes/immunology , Biological Transport , Blood-Brain Barrier/physiology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Capillary Permeability/immunology , Disease Models, Animal , Female , Ganglia, Spinal/immunology , Herpes Genitalis/immunology , Herpes Genitalis/virology , Herpesvirus 2, Human/immunology , Histocompatibility Antigens Class I , Immunologic Memory/immunology , Integrin alpha4/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice , Nerve Tissue/immunology , Neurons/immunology , Nose/virology , Receptors, Fc , Spinal Cord/immunology , Vesiculovirus/immunology
6.
J Allergy Clin Immunol ; 147(4): 1341-1353, 2021 04.
Article in English | MEDLINE | ID: mdl-32781002

ABSTRACT

BACKGROUND: Chronic itch is a debilitating symptom of inflammatory skin diseases, but the underlying mechanism is poorly understood. We have recently demonstrated that astrocytes in the spinal dorsal horn become reactive in models of atopic and contact dermatitis via activation of the transcription factor signal transducer and activator of transcription 3 (STAT3) and critically contribute to chronic itch. In general, STAT3 is transiently activated; however, STAT3 activation in reactive astrocytes of chronic itch model mice persistently occurs via an unknown mechanism. OBJECTIVE: We aimed to determine the mechanisms of persistent activation of astrocytic STAT3 in chronic itch conditions. METHODS: To determine the factors that are required for persistent activation of astrocytic STAT3, Western blotting and calcium imaging with cultured astrocytes or spinal cord slices were performed. Thereafter, chronic itch model mice were used for genetic and behavioral experiments to confirm the role of the factors determined to mediate persistent STAT3 activation from in vitro and ex vivo experiments in chronic itch. RESULTS: IP3 receptor type 1 (IP3R1) knockdown in astrocytes suppressed IL-6-induced persistent STAT3 activation and expression of lipocalin-2 (LCN2), an astrocytic STAT3-dependent inflammatory factor that is required for chronic itch. IP3R1-dependent astrocytic Ca2+ responses involved Ca2+ influx through the cation channel transient receptor potential canonical (TRPC), which was required for persistent STAT3 activation evoked by IL-6. IL-6 expression was upregulated in dorsal root ganglion neurons in a mouse model of chronic itch. Dorsal root ganglion neuron-specific IL-6 knockdown, spinal astrocyte-specific IP3R1 knockdown, and pharmacologic spinal TRPC inhibition attenuated LCN2 expression and chronic itch. CONCLUSION: Our findings suggest that IP3R1/TRPC channel-mediated Ca2+ signals elicited by IL-6 in astrocytes are necessary for persistent STAT3 activation, LCN2 expression, and chronic itch, and they may also provide new targets for therapeutic intervention.


Subject(s)
Astrocytes/immunology , Inositol 1,4,5-Trisphosphate Receptors/immunology , Interleukin-6/immunology , Pruritus/immunology , STAT3 Transcription Factor/immunology , TRPC Cation Channels/immunology , Animals , Calcium Signaling , Cells, Cultured , Chronic Disease , Ganglia, Spinal/immunology , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mice, Inbred C57BL , Mice, Knockout
7.
J Virol ; 94(14)2020 07 01.
Article in English | MEDLINE | ID: mdl-32404525

ABSTRACT

Mouse hepatitis virus (MHV) is a murine betacoronavirus (m-CoV) that causes a wide range of diseases in mice and rats, including hepatitis, enteritis, respiratory diseases, and encephalomyelitis in the central nervous system (CNS). MHV infection in mice provides an efficient cause-effect experimental model to understand the mechanisms of direct virus-induced neural-cell damage leading to demyelination and axonal loss, which are pathological features of multiple sclerosis (MS), the most common disabling neurological disease in young adults. Infiltration of T lymphocytes, activation of microglia, and their interplay are the primary pathophysiological events leading to disruption of the myelin sheath in MS. However, there is emerging evidence supporting gray matter involvement and degeneration in MS. The investigation of T cell function in the pathogenesis of deep gray matter damage is necessary. Here, we employed RSA59 (an isogenic recombinant strain of MHV-A59)-induced experimental neuroinflammation model to compare the disease in CD4-/- mice with that in CD4+/+ mice at days 5, 10, 15, and 30 postinfection (p.i.). Viral titer estimation, nucleocapsid gene amplification, and viral antinucleocapsid staining confirmed enhanced replication of the virions in the absence of functional CD4+ T cells in the brain. Histopathological analyses showed elevated susceptibility of CD4-/- mice to axonal degeneration in the CNS, with augmented progression of acute poliomyelitis and dorsal root ganglionic inflammation rarely observed in CD4+/+ mice. Depletion of CD4+ T cells showed unique pathological bulbar vacuolation in the brain parenchyma of infected mice with persistent CD11b+ microglia/macrophages in the inflamed regions on day 30 p.i. In summary, the current study suggests that CD4+ T cells are critical for controlling acute-stage poliomyelitis (gray matter inflammation), chronic axonal degeneration, and inflammatory demyelination due to loss of protective antiviral host immunity.IMPORTANCE The current trend in CNS disease biology is to attempt to understand the neural-cell-immune interaction to investigate the underlying mechanism of neuroinflammation, rather than focusing on peripheral immune activation. Most studies in MS are targeted toward understanding the involvement of CNS white matter. However, the importance of gray matter damage has become critical in understanding the long-term progressive neurological disorder. Our study highlights the importance of CD4+ T cells in safeguarding neurons against axonal blebbing and poliomyelitis from murine betacoronavirus-induced neuroinflammation. Current knowledge of the mechanisms that lead to gray matter damage in MS is limited, because the most widely used animal model, experimental autoimmune encephalomyelitis (EAE), does not present this aspect of the disease. Our results, therefore, add to the existing limited knowledge in the field. We also show that the microglia, though important for the initiation of neuroinflammation, cannot establish a protective host immune response without the help of CD4+ T cells.


Subject(s)
Axons/immunology , Axons/metabolism , CD4 Antigens/deficiency , Coronavirus Infections/immunology , Coronavirus Infections/virology , Murine hepatitis virus/physiology , Poliomyelitis/etiology , Animals , Axons/pathology , Brain/immunology , Brain/metabolism , Brain/pathology , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Coronavirus Infections/pathology , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility/immunology , Ganglia, Spinal/immunology , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Immunohistochemistry , Inflammation Mediators/metabolism , Mice
8.
FASEB J ; 34(4): 5951-5966, 2020 04.
Article in English | MEDLINE | ID: mdl-32157739

ABSTRACT

Neuron-immune interaction in the dorsal root ganglia (DRG) plays a pivotal role in the neuropathic pain development after nerve injury. Sigma-1 receptor (Sig-1R) is expressed by DRG neurons but its role in neuropathic pain is not fully understood. We investigated the effect of peripheral Sig-1R on neuroinflammation in the DRG after spared (sciatic) nerve injury (SNI) in mice. Nerve injury induced a decrease in NeuN staining along with the nuclear eccentricity and ATF3 expression in the injured DRG. Sig-1R was present in all DRG neurons examined, and after SNI this receptor translocated to the periphery of the soma and the vicinity of the nucleus, especially in injured ATF3 + neurons. In WT mice, injured DRG produced the chemokine CCL2, and this was followed by massive infiltration of macrophages/monocytes, which clustered mainly around sensory neurons with translocated Sig-1R, accompanied by robust IL-6 increase and mechanical allodynia. In contrast, Sig-1R knockout (Sig-1R-KO) mice showed reduced levels of CCL2, decreased macrophage/monocyte infiltration into DRG, and less IL-6 and neuropathic mechanical allodynia after SNI. Our findings point to an important role of peripheral Sig-1R in sensory neuron-macrophage/monocyte communication in the DRG after peripheral nerve injury; thus, these receptors may contribute to the neuropathic pain phenotype.


Subject(s)
Ganglia, Spinal/pathology , Hyperalgesia/pathology , Macrophages/pathology , Neuralgia/pathology , Neurons/pathology , Peripheral Nerve Injuries/complications , Receptors, sigma/physiology , Animals , Behavior, Animal , Disease Models, Animal , Female , Ganglia, Spinal/immunology , Ganglia, Spinal/metabolism , Hyperalgesia/etiology , Hyperalgesia/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Neuralgia/etiology , Neuralgia/metabolism , Neurons/immunology , Neurons/metabolism , Sigma-1 Receptor
9.
Int J Mol Sci ; 22(23)2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34884605

ABSTRACT

Autotomy, self-mutilation of a denervated limb, is common in animals after peripheral nerve injury (PNI) and is a reliable proxy for neuropathic pain in humans. Understanding the occurrence and treatment of autotomy remains challenging. The objective of this study was to investigate the occurrence of autotomy in nude and Wistar rats and evaluate the differences in macrophage activation and fiber sensitization contributing to the understanding of autotomy behavior. Autotomy in nude and Wistar rats was observed and evaluated 6 and 12 weeks after sciatic nerve repair surgery. The numbers of macrophages and the types of neurons in the dorsal root ganglion (DRG) between the two groups were compared by immunofluorescence studies. Immunostaining of T cells in the DRG was also assessed. Nude rats engaged in autotomy with less frequency than Wistar rats. Autotomy symptoms were also relatively less severe in nude rats. Immunofluorescence studies revealed increased macrophage accumulation and activation in the DRG of Wistar rats. The percentage of NF200+ neurons was higher at 6 and 12 weeks in Wistar rats compared to nude rats, but the percentage of CGRP+ neurons did not differ between two groups. Additionally, macrophages were concentrated around NF200-labeled A fibers. At 6 and 12 weeks following PNI, CD4+ T cells were not found in the DRG of the two groups. The accumulation and activation of macrophages in the DRG may account for the increased frequency and severity of autotomy in Wistar rats. Our results also suggest that A fiber neurons in the DRG play an important role in autotomy.


Subject(s)
Behavior, Animal , Ganglia, Spinal/immunology , Macrophage Activation/immunology , Pain, Postoperative/pathology , Peripheral Nerve Injuries/complications , Sciatic Nerve/injuries , Self Mutilation/pathology , Animals , Pain, Postoperative/etiology , Rats , Rats, Nude , Rats, Wistar , Self Mutilation/etiology
10.
Int J Mol Sci ; 22(21)2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34769297

ABSTRACT

Pulsed radiofrequency (PRF) works by delivering short bursts of radiofrequency to a target nerve, thereby affecting nerve signal transduction to reduce pain. Although preliminary clinical investigations have shown that PRF treatment can be used safely as an alternative interventional treatment in patients with refractory pain conditions, unexpected damage to a normal nerve/ganglion is still one of the possible complications of using the PRF strategy. Noxious pain may also be triggered if PRF treatment accidentally damages an intact nerve. However, few studies in the literature have described the intracellular modifications that occur in neuronal cells after PRF stimulation. Therefore, in this study, we evaluated the effects of PRF on unimpaired nerve function and investigated the potential mechanisms of PRF-induced pain. Wistar rats were stimulated with 30-60 V of PRF for 6 min, and mechanical allodynia, cold hypersensitivity, cytokine and matrix metalloproteinase (MMP) production, and mitogen-activated protein kinase activity (p38 MAPK, ERK1/2, JNK/SAPK) were analyzed. The results indicated that PRF stimulation induced a significant algesic effect and nociceptive response. In addition, the protein array and Western blotting analyses showed that the clinical application of 60 V of PRF can induce the activation of MAPKs and the production of inflammatory cytokines and MMPs in the lumbar dorsal horn, which is necessary for nerve inflammation, and it can be suppressed by MAPK antagonist treatment. These results indicate that PRF stimulation may induce inflammation of the intact nerve, which in turn causes inflammatory pain. This conclusion can also serve as a reminder for PRF treatment of refractory pain.


Subject(s)
Cryopyrin-Associated Periodic Syndromes/therapy , Ganglia, Spinal/immunology , Hyperalgesia/therapy , Pulsed Radiofrequency Treatment/adverse effects , Spinal Cord/immunology , Animals , Cryopyrin-Associated Periodic Syndromes/etiology , Cryopyrin-Associated Periodic Syndromes/metabolism , Cytokines/metabolism , Hyperalgesia/etiology , Hyperalgesia/metabolism , Male , Matrix Metalloproteinases/metabolism , Pain , Random Allocation , Rats , Rats, Wistar , Spinal Cord/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Immunology ; 159(4): 413-428, 2020 04.
Article in English | MEDLINE | ID: mdl-31919846

ABSTRACT

A growing body of evidence has indicated that the release of nociceptive factors, such as interleukins and chemokines, by activated immune and glial cells has crucial significance for neuropathic pain generation and maintenance. Moreover, changes in the production of nociceptive immune factors are associated with low opioid efficacy in the treatment of neuropathy. Recently, it has been suggested that CC chemokine receptor type 1 (CCR1) signaling is important for nociception. Our study provides evidence that the development of hypersensitivity in rats following chronic constriction injury (CCI) of the sciatic nerve is associated with significant up-regulation of endogenous CCR1 ligands, namely, CCL2, CCL3, CCL4, CCL6, CCL7 and CCL9 in the spinal cord and CCL2, CCL6, CCL7 and CCL9 in dorsal root ganglia (DRG). We showed that single and repeated intrathecal administration of J113863 (an antagonist of CCR1) attenuated mechanical and thermal hypersensitivity. Moreover, repeated administration of a CCR1 antagonist enhanced the analgesic properties of morphine and buprenorphine after CCI. Simultaneously, repeated administration of J113863 reduced the protein levels of IBA-1 in the spinal cord and MPO and CD4 in the DRG and, as a consequence, the level of pronociceptive factors, such as interleukin-1ß (IL-1ß), IL-6 and IL-18. The data obtained provide evidence that CCR1 blockade reduces hypersensitivity and increases opioid-induced analgesia through the modulation of neuroimmune interactions.


Subject(s)
Analgesics/pharmacology , Buprenorphine/pharmacology , Hyperalgesia/drug therapy , Morphine/pharmacology , Neuralgia/drug therapy , Receptors, CCR1/immunology , Xanthenes/pharmacology , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/immunology , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Disease Models, Animal , Drug Synergism , Ganglia, Spinal/drug effects , Ganglia, Spinal/immunology , Ganglia, Spinal/physiopathology , Gene Expression Regulation , Hyperalgesia/genetics , Hyperalgesia/immunology , Hyperalgesia/physiopathology , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Male , Microfilament Proteins/genetics , Microfilament Proteins/immunology , Neuralgia/genetics , Neuralgia/immunology , Neuralgia/physiopathology , Nociception/drug effects , Peroxidase/genetics , Peroxidase/immunology , Protein Isoforms/genetics , Protein Isoforms/immunology , Rats , Rats, Wistar , Receptors, CCR1/antagonists & inhibitors , Receptors, CCR1/genetics , Sciatic Nerve/drug effects , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Signal Transduction
12.
Am J Physiol Lung Cell Mol Physiol ; 318(5): L953-L964, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32159971

ABSTRACT

The lungs and the immune and nervous systems functionally interact to respond to respiratory environmental exposures and infections. The lungs are innervated by vagal sensory neurons of the jugular and nodose ganglia, fused together in smaller mammals as the jugular-nodose complex (JNC). Whereas the JNC shares properties with the other sensory ganglia, the trigeminal (TG) and dorsal root ganglia (DRG), these sensory structures express differential sets of genes that reflect their unique functionalities. Here, we used RNA sequencing (RNA-seq) in mice to identify the differential transcriptomes of the three sensory ganglia types. Using a fluorescent retrograde tracer and fluorescence-activated cell sorting, we isolated a defined population of airway-innervating JNC neurons and determined their differential transcriptional map after pulmonary exposure to lipopolysaccharide (LPS), a major mediator of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) after infection with gram-negative bacteria or inhalation of organic dust. JNC neurons activated an injury response program, leading to increased expression of gene products such as the G protein-coupled receptor Cckbr, inducing functional changes in neuronal sensitivity to peptides, and Gpr151, also rapidly induced upon neuropathic nerve injury in pain models. Unique JNC-specific transcripts, present at only minimal levels in TG, DRG, and other organs, were identified. These included TMC3, encoding for a putative mechanosensor, and urotensin 2B, a hypertensive peptide. These findings highlight the unique properties of the JNC and reveal that ALI/ARDS rapidly induces a nerve injury-related state, changing vagal excitability.


Subject(s)
Nodose Ganglion/drug effects , Pneumonia/genetics , Receptor, Cholecystokinin B/genetics , Sensory Receptor Cells/drug effects , Transcriptome , Vagus Nerve Injuries/genetics , Animals , Ganglia, Spinal/drug effects , Ganglia, Spinal/immunology , Ganglia, Spinal/pathology , Gene Expression Profiling , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Lipopolysaccharides/pharmacology , Lung/drug effects , Lung/immunology , Lung/pathology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Nodose Ganglion/immunology , Nodose Ganglion/pathology , Peptide Hormones/genetics , Peptide Hormones/immunology , Pneumonia/chemically induced , Pneumonia/immunology , Pneumonia/pathology , Receptor, Cholecystokinin B/immunology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Sensory Receptor Cells/immunology , Sensory Receptor Cells/pathology , Sequence Analysis, RNA , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/immunology , Trigeminal Ganglion/pathology , Vagus Nerve Injuries/chemically induced , Vagus Nerve Injuries/immunology , Vagus Nerve Injuries/pathology
13.
Osteoarthritis Cartilage ; 28(5): 562-571, 2020 05.
Article in English | MEDLINE | ID: mdl-31862470

ABSTRACT

In this narrative review, we discuss the emerging role of innate immunity in osteoarthritis (OA) joint pain. First, we give a brief description of the pain pathway in the context of OA. Then we consider how neuro-immune signaling pathways may promote OA pain. First, activation of neuronal Pattern Recognition Receptors by mediators released in a damaged joint can result in direct excitation of nociceptors, as well as in production of chemokines and cytokines. Secondly, indirect neuro-immune signaling may occur when innate immune cells produce algogenic factors, including chemokines and cytokines, that act on the pain pathway. Neuro-immune crosstalk occurs at different levels of the pathway, starting in the joint but also in the innervating dorsal root ganglia and in the dorsal horn. Synovitis is characterized by recruitment of immune cells, including macrophages, mast cells, and CD4+ lymphocytes, which may contribute to nociceptor sensitization and OA pain through production of algogenic factors that amplify the activation of sensory neurons. We discuss examples where this scenario has been suggested by findings in human OA and in animal models. Overall, increasing evidence suggests that innate immune pathways play an initiating as well as facilitating role in pain, but information on how these pathways operate in OA remains limited. Since these innate pathways are eminently targetable, future studies in this area may provide fruitful leads towards a better management of symptomatic OA.


Subject(s)
Arthralgia/immunology , Immunity, Innate/immunology , Nociceptors/metabolism , Osteoarthritis/immunology , Synovitis/immunology , Animals , Arthralgia/metabolism , Cytokines/immunology , Cytokines/metabolism , Ganglia, Spinal/immunology , Ganglia, Spinal/metabolism , Humans , Neuroimmunomodulation/immunology , Osteoarthritis/metabolism , Receptors, Pattern Recognition/metabolism , Spinal Cord Dorsal Horn/immunology , Spinal Cord Dorsal Horn/metabolism , Synovitis/metabolism
14.
FASEB J ; 33(1): 668-682, 2019 01.
Article in English | MEDLINE | ID: mdl-30024789

ABSTRACT

Nogo-A is a key inhibitory molecule of axon regeneration in oligodendrocytes. However, little is known about its role in adult neurons. In this study, we showed an important function of Nogo-A on regulation of inflammatory pain in dorsal root ganglion (DRG) neurons. In adult rats with complete Freund's adjuvant (CFA) hind paw inflammation, DRG neurons showed a significant increase in Nogo-A expression. Disruption of Nogo-A signaling with Nogo-66 receptor antagonist peptide, Nogo-A blocking antibody, Nogo-A short hairpin RNA, or Nogo-A gene knockout attenuated CFA-induced inflammatory heat hyperalgesia. Moreover, disruption of Nogo-A signaling suppressed the function and expression in DRG neurons of the transient receptor potential vanilloid subfamily member (TRPV)-1 channel, which is known to be the endogenous transducer of noxious heat during inflammation. These effects were accompanied with a reduction in LIM domain kinase (LIMK)/cofilin phosphorylation and actin polymerization. Similar disruption of actin filament architecture by direct action of Latrunculin A reduced the TRPV-1 activity and up-regulation of TRPV-1 protein caused by CFA. We conclude that Nogo-A plays an essential role in the development of inflammatory heat hyperalgesia, partly through maintaining TRPV-1 function via activation of the LIMK/cofilin pathway, which regulates actin filament dynamics. These findings support a therapeutic potential of modulating Nogo-A signaling in pain management.-Hu, F., Liu, H.-C., Su, D.-Q., Chen, H.-J., Chan, S.-O., Wang, Y., Wang, J. Nogo-A promotes inflammatory heat hyperalgesia by maintaining TRPV-1 function in the rat dorsal root ganglion neuron.


Subject(s)
Ganglia, Spinal/pathology , Hot Temperature/adverse effects , Hyperalgesia/etiology , Neurogenic Inflammation/complications , Neurons/pathology , Nogo Proteins/metabolism , TRPV Cation Channels/metabolism , Animals , Cofilin 1/metabolism , Ganglia, Spinal/immunology , Ganglia, Spinal/metabolism , Hyperalgesia/metabolism , Hyperalgesia/pathology , Lim Kinases/metabolism , Male , Neurogenic Inflammation/metabolism , Neurogenic Inflammation/pathology , Neurons/immunology , Neurons/metabolism , Rats , Rats, Sprague-Dawley
15.
Glia ; 66(3): 538-561, 2018 03.
Article in English | MEDLINE | ID: mdl-29148104

ABSTRACT

Oligodendroglial cell death and demyelination are hallmarks of neurotrauma and multiple sclerosis that cause axonal damage and functional impairments. Remyelination remains a challenge as the ability of endogenous precursor cells for oligodendrocyte replacement is hindered in the unfavorable milieu of demyelinating conditions. Here, in a rat model of lysolecithin lysophosphatidyl-choline (LPC)-induced focal demyelination, we report that Neuregulin-1 (Nrg-1), an important factor for oligodendrocytes and myelination, is dysregulated in demyelinating lesions and its bio-availability can promote oligodendrogenesis and remyelination. We delivered recombinant human Nrg-1ß1 (rhNrg-1ß1) intraspinally in the vicinity of LPC demyelinating lesion in a sustained manner using poly lactic-co-glycolic acid microcarriers. Availability of Nrg-1 promoted generation and maturation of new oligodendrocytes, and accelerated endogenous remyelination by both oligodendrocyte and Schwann cell populations in demyelinating foci. Importantly, Nrg-1 enhanced myelin thickness in newly remyelinated spinal cord axons. Our complementary in vitro studies also provided direct evidence that Nrg-1 significantly promotes maturation of new oligodendrocytes and facilitates their transition to a myelinating phenotype. Nrg-1 therapy remarkably attenuated the upregulated expression chondroitin sulfate proteoglycans (CSPGs) specific glycosaminoglycans in the extracellular matrix of demyelinating foci and promoted interleukin-10 (IL-10) production by immune cells. CSPGs and IL-10 are known to negatively and positively regulate remyelination, respectively. We found that Nrg-1 effects are mediated through ErbB2 and ErbB4 receptor activation. Our work provides novel evidence that dysregulated levels of Nrg-1 in demyelinating lesions of the spinal cord pose a challenge to endogenous remyelination, and appear to be an underlying cause of myelin thinning in newly remyelinated axons.


Subject(s)
Demyelinating Diseases/therapy , Immunomodulation , Neuregulin-1/administration & dosage , Neuroprotective Agents/administration & dosage , Remyelination/physiology , Spinal Cord/immunology , Animals , Cells, Cultured , Chondroitin Sulfate Proteoglycans/metabolism , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Disease Models, Animal , Drug Carriers , Extracellular Matrix/immunology , Extracellular Matrix/pathology , Female , Ganglia, Spinal/immunology , Ganglia, Spinal/pathology , Humans , Lactic Acid , Male , Neural Stem Cells/immunology , Neural Stem Cells/pathology , Oligodendroglia/immunology , Oligodendroglia/pathology , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Spinal Cord/pathology , Spinal Cord Diseases/immunology , Spinal Cord Diseases/pathology , Spinal Cord Diseases/therapy
16.
Mol Pain ; 14: 1744806918817975, 2018.
Article in English | MEDLINE | ID: mdl-30453826

ABSTRACT

Diabetic peripheral neuropathy is a major debilitating late complication of diabetes, which significantly reduces the quality of life in patients. Diabetic peripheral neuropathy is associated with a wide spectrum of sensory abnormalities, where in loss of sensation or hypoalgesia to applied external stimuli is paradoxically accompanied by debilitating tonic spontaneous pain. In numerous studies on animal models of diabetic peripheral neuropathy, behavioural measurements have been largely confined to analysis of evoked withdrawal to mechanical and thermal stimuli applied to dermatomes, whereas spontaneous, on-going pain has not been widely studied. In the Streptozotocin model of type 1 diabetes, we employed the Conditioned Place Preference test to assess tonic pain. Our results indicate that both phases, that is, early evoked hypersensitivity (i.e. 5-7 weeks post-Streptozotocin) as well as late stage hypoalgesia (i.e. 17-20 weeks post-Streptozotocin) are accompanied by significant tonic pain in mice with diabetic peripheral neuropathy. We also report on the temporal relation between on-going pain and neuropathological changes in the dorsal root ganglia of mice with diabetic peripheral neuropathy up to 6 months post-Streptozotocin. Neither early hypersensitivity nor late hypoalgesia were associated with markers of cellular stress in the dorsal root ganglia. Whereas significant neutrophil infiltration was observed in the dorsal root ganglia over both early and late stages post-Streptozotocin, T-cell infiltration in the dorsal root ganglia was prominent at late stages post-Streptozotocin. Thus, longitudinal analyses reveal that similar to patients with chronic diabetic peripheral neuropathy, mice show tonic pain despite sensory loss after several months in the Streptozotocin model, which is accompanied by neuroimmune interactions in the dorsal root ganglia.


Subject(s)
Diabetic Neuropathies/complications , Diabetic Neuropathies/immunology , Ganglia, Spinal/immunology , Ganglia, Spinal/pathology , Hyperalgesia/complications , Neuralgia/complications , Activating Transcription Factor 3/metabolism , Animals , CD3 Complex/metabolism , Diabetic Neuropathies/pathology , Hyperalgesia/pathology , Male , Mice, Inbred C57BL , Neuralgia/pathology
17.
Rheumatology (Oxford) ; 57(3): 429-440, 2018 03 01.
Article in English | MEDLINE | ID: mdl-28968842

ABSTRACT

Chronic pain is a major debilitating condition that is difficult to treat. Although chronic pain may appear to be a disorder of the nervous system, crucial roles for immune cells and their mediators have been identified as important contributors in various types of pain. This review focuses on how the immune system regulates pain and discusses the emerging roles of immune cells in the initiation or maintenance of chronic pain. We highlight which immune cells infiltrate damaged nerves, the dorsal root ganglia, spinal cord and tissues around free nerve endings and discuss through which mechanisms they control pain. Finally we discuss emerging roles of the immune system in resolving pain and how the immune system contributes to the transition from acute to chronic pain. We propose that targeting some of these immune processes may provide novel therapeutic opportunities for the treatment of chronic pain.


Subject(s)
Acute Pain/immunology , Chronic Pain/immunology , Immune System/physiology , Immunity, Cellular , Ganglia, Spinal/immunology , Humans , Spinal Cord/immunology
18.
Gut ; 66(12): 2121-2131, 2017 12.
Article in English | MEDLINE | ID: mdl-27590998

ABSTRACT

AIMS AND BACKGROUND: Psychological stress accompanies chronic inflammatory diseases such as IBD, and stress hormones can exacerbate pain signalling. In contrast, the endogenous opioid system has an important analgesic action during chronic inflammation. This study examined the interaction of these pathways. METHODS: Mouse nociceptive dorsal root ganglia (DRG) neurons were incubated with supernatants from segments of inflamed colon collected from patients with chronic UC and mice with dextran sodium sulfate (cDSS)-induced chronic colitis. Stress effects were studied by adding stress hormones (epinephrine and corticosterone) to dissociated neurons or by exposing cDSS mice to water avoidance stress. Changes in excitability of colonic DRG nociceptors were measured using patch clamp and Ca2+ imaging techniques. RESULTS: Supernatants from patients with chronic UC and from colons of mice with chronic colitis caused a naloxone-sensitive inhibition of neuronal excitability and capsaicin-evoked Ca2+ responses. Stress hormones decreased signalling induced by human and mouse supernatants. This effect resulted from stress hormones signalling directly to DRG neurons and indirectly through signalling to the immune system, leading to decreased opioid levels and increased acute inflammation. The net effect of stress was a change endogenous opioid signalling in DRG neurons from an inhibitory to an excitatory effect. This switch was associated with a change in G protein-coupled receptor excitatory signalling to a pathway sensitive to inhibitors of protein kinase A-protein, phospholipase C-protein and G protein ßϒ subunits. CONCLUSIONS: Stress hormones block the inhibitory actions of endogenous opioids and can change the effect of opioid signalling in DRG neurons to excitation. Targeting these pathways may prevent heavy opioid use in IBD.


Subject(s)
Colitis/metabolism , Colon/innervation , Ganglia, Spinal/metabolism , Stress, Psychological/physiopathology , beta-Endorphin/metabolism , Adult , Aged , Animals , Biopsy , Chronic Disease , Colitis/immunology , Cytokines/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/immunology , Humans , Mice , Mice, Inbred C57BL , Middle Aged , Naloxone/pharmacology , Nociceptors/physiology , Patch-Clamp Techniques , Signal Transduction
19.
Biochem Biophys Res Commun ; 485(4): 707-712, 2017 Apr 15.
Article in English | MEDLINE | ID: mdl-28237704

ABSTRACT

Atopic dermatitis (AD), a chronic inflammatory skin disease, manifests as intractable itch, but its underlying mechanisms are poorly understood. This study assessed the relationship between immunoglobulin G (IgG) and dorsal root ganglia (DRG) in NC/Nga mice, a model of AD that manifests AD-like symptoms including itch. Immunohistochemical analysis showed large amounts of IgG in DRG extracts of NC/Nga mice with AD-like dermatitis, with a large fraction of the IgG distributed in satellite glial cells of the DRG. Proteomic analysis showed that this IgG was reactive against tropomyosin of Dermatophagoides farinae. These findings indicate that the accumulation of anti-tropomyosin IgG in DRG of atopic NC/Nga mice may be associated with the pathogenesis of AD-like symptoms, including itch.


Subject(s)
Arthropod Proteins/immunology , Dermatitis, Atopic/immunology , Dermatophagoides farinae/immunology , Ganglia, Spinal/immunology , Immunoglobulin G/immunology , Tropomyosin/immunology , Amino Acid Sequence , Animals , Antigens, Dermatophagoides/immunology , Blotting, Western , Dermatitis, Atopic/metabolism , Disease Models, Animal , Ganglia, Spinal/metabolism , Humans , Immunoglobulin G/metabolism , Immunohistochemistry , Male , Mice , Neuroglia/immunology , Neuroglia/metabolism , Proteome/immunology , Proteome/metabolism , Proteomics/methods , Skin/immunology , Skin/metabolism , Skin/pathology
20.
J Autoimmun ; 77: 11-38, 2017 02.
Article in English | MEDLINE | ID: mdl-27939129

ABSTRACT

Campylobacter jejuni is a leading cause of bacterial gastroenteritis linked to several serious autoimmune sequelae such as the peripheral neuropathies Guillain Barré syndrome (GBS) and Miller Fisher syndrome (MFS). We hypothesized that GBS and MFS can result in NOD wild type (WT) mice or their congenic interleukin (IL)-10 or B7-2 knockouts secondary to C. jejuni infection. Mice were gavaged orally with C. jejuni strains HB93-13 and 260.94 from patients with GBS or CF93-6 from a patient with MFS and assessed for clinical neurological signs and phenotypes, anti-ganglioside antibodies, and cellular infiltrates and lesions in gut and peripheral nerve tissues. Significant increases in autoantibodies against single gangliosides (GM1, GQ1b, GD1a) occurred in infected NOD mice of all genotypes, although the isotypes varied (NOD WT had IgG1, IgG3; NOD B7-2-/- had IgG3; NOD IL-10-/- had IgG1, IgG3, IgG2a). Infected NOD WT and NOD IL-10-/- mice also produced anti-ganglioside antibodies of the IgG1 isotype directed against a mixture of GM1/GQ1b gangliosides. Phenotypic tests showed significant differences between treatment groups of all mouse genotypes. Peripheral nerve lesions with macrophage infiltrates were significantly increased in infected mice of NOD WT and IL-10-/- genotypes compared to sham-inoculated controls, while lesions with T cell infiltrates were significantly increased in infected mice of the NOD B7-2-/- genotype compared to sham-inoculated controls. In both infected and sham inoculated NOD IL-10-/- mice, antibiotic treatment exacerbated neurological signs, lesions and the amount and number of different isotypes of antiganglioside autoantibodies produced. Thus, inducible mouse models of post-C. jejuni GBS are feasible and can be characterized based on evaluation of three factors-onset of GBS clinical signs/phenotypes, anti-ganglioside autoantibodies and nerve lesions. Based on these factors we characterized 1) NOD B-7-/- mice as an acute inflammatory demyelinating polyneuropathy (AIDP)-like model, 2) NOD IL-10-/- mice as an acute motor axonal neuropathy (AMAN)-like model best employed over a limited time frame, and 3) NOD WT mice as an AMAN model with mild clinical signs and lesions. Taken together these data demonstrate that C. jejuni strain genotype, host genotype and antibiotic treatment affect GBS disease outcomes in mice and that many disease phenotypes are possible.


Subject(s)
Anti-Bacterial Agents/adverse effects , Campylobacter Infections/complications , Campylobacter Infections/microbiology , Campylobacter jejuni , Guillain-Barre Syndrome/etiology , Guillain-Barre Syndrome/pathology , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Bacterial/immunology , Autoantibodies/immunology , Campylobacter Infections/drug therapy , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Ganglia, Spinal/immunology , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Guillain-Barre Syndrome/physiopathology , Immunoglobulin G/immunology , Mice , Mice, Inbred NOD , Mice, Knockout , Peripheral Nerves/metabolism , Peripheral Nerves/pathology , Peripheral Nerves/physiopathology , Peripheral Nerves/virology , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL