Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20.480
Filter
1.
J Vis Exp ; (210)2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39221953

ABSTRACT

The dorsal root ganglia (DRG), housing primary sensory neurons, transmit somatosensory and visceral afferent inputs to the dorsal horn of the spinal cord. They play a pivotal role in both physiological and pathological states, including neuropathic and visceral pain. In vivo calcium imaging of DRG enables real-time observation of calcium transients in single units or neuron ensembles. Accumulating evidence indicates that DRG neuronal activities induced by somatic stimulation significantly affect autonomic and visceral functions. While lumbar DRG calcium imaging has been extensively studied, thoracic segment DRG calcium imaging has been less explored due to surgical exposure and stereotaxic fixation challenges. Here, we utilized in vivo calcium imaging at the thoracic1 dorsal root ganglion (T1-DRG) to investigate changes in neuronal activity resulting from somatic stimulations of the forelimb. This approach is crucial for understanding the somato-cardiac reflex triggered by peripheral nerve stimulations (PENS), such as acupuncture. Notably, synchronization of cardiac function was observed and measured by electrocardiogram (ECG), with T-DRG neuronal activities, potentially establishing a novel paradigm for somato-visceral reflex in the thoracic segments.


Subject(s)
Calcium , Electrocardiography , Ganglia, Spinal , Animals , Ganglia, Spinal/physiology , Calcium/metabolism , Calcium/analysis , Electrocardiography/methods , Mice , Peripheral Nerves/physiology , Forelimb/innervation , Forelimb/physiology
2.
PLoS One ; 19(9): e0308389, 2024.
Article in English | MEDLINE | ID: mdl-39236043

ABSTRACT

Micro and nanoscale patterning of surface features and biochemical cues have emerged as tools to precisely direct neurite growth into close proximity with next generation neural prosthesis electrodes. Biophysical cues can exert greater influence on neurite pathfinding compared to the more well studied biochemical cues; yet the signaling events underlying the ability of growth cones to respond to these microfeatures remain obscure. Intracellular Ca2+ signaling plays a critical role in how a growth cone senses and grows in response to various cues (biophysical features, repulsive peptides, chemo-attractive gradients). Here, we investigate the role of inositol triphosphate (IP3) and ryanodine-sensitive receptor (RyR) signaling as sensory neurons (spiral ganglion neurons, SGNs, and dorsal root ganglion neurons, DRGNs) pathfind in response to micropatterned substrates of varied geometries. We find that IP3 and RyR signaling act in the growth cone as they navigate biophysical cues and enable proper guidance to biophysical, chemo-permissive, and chemo-repulsive micropatterns. In response to complex micropatterned geometries, RyR signaling appears to halt growth in response to both topographical features and chemo-repulsive cues. IP3 signaling appears to play a more complex role, as growth cones appear to sense the microfeatures in the presence of xestospongin C but are unable to coordinate turning in response to them. Overall, key Ca2+ signaling elements, IP3 and RyR, are found to be essential for SGNs to pathfind in response to engineered biophysical and biochemical cues. These findings inform efforts to precisely guide neurite regeneration for improved neural prosthesis function, including cochlear implants.


Subject(s)
Neurites , Ryanodine Receptor Calcium Release Channel , Signal Transduction , Ryanodine Receptor Calcium Release Channel/metabolism , Neurites/metabolism , Animals , Ganglia, Spinal/metabolism , Ganglia, Spinal/cytology , Growth Cones/metabolism , Growth Cones/drug effects , Calcium Signaling , Rats , Surface Properties , Cells, Cultured , Oxazoles , Macrocyclic Compounds
3.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39125611

ABSTRACT

Sexual dimorphism among mammals includes variations in the pain threshold. These differences are influenced by hormonal fluctuations in females during the estrous and menstrual cycles of rodents and humans, respectively. These physiological conditions display various phases, including proestrus and diestrus in rodents and follicular and luteal phases in humans, distinctly characterized by varying estrogen levels. In this study, we evaluated the capsaicin responses in male and female mice at different estrous cycle phases, using two murine acute pain models. Our findings indicate that the capsaicin-induced pain threshold was lower in the proestrus phase than in the other three phases in both pain assays. We also found that male mice exhibited a higher pain threshold than females in the proestrus phase, although it was similar to females in the other cycle phases. We also assessed the mRNA and protein levels of TRPV1 in the dorsal root and trigeminal ganglia of mice. Our results showed higher TRPV1 protein levels during proestrus compared to diestrus and male mice. Unexpectedly, we observed that the diestrus phase was associated with higher TRPV1 mRNA levels than those in both proestrus and male mice. These results underscore the hormonal influence on TRPV1 expression regulation and highlight the role of sex steroids in capsaicin-induced pain.


Subject(s)
Capsaicin , Pain , TRPV Cation Channels , Animals , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Capsaicin/pharmacology , Male , Female , Mice , Pain/metabolism , Pain/genetics , Gonadal Steroid Hormones/metabolism , Estrous Cycle/drug effects , Pain Threshold/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/drug effects , Gene Expression Regulation/drug effects , Sex Characteristics , RNA, Messenger/metabolism , RNA, Messenger/genetics
4.
Biomed Mater ; 19(5)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39094612

ABSTRACT

The therapy of large defects in peripheral nerve injury (PNI) suffers from several drawbacks, especially the lack of autologous nerve donors. Nerve conduits are considered as a solution for nerve injury treatment, but biocompatibility improvements is still required for conduits prepared with synthetic materials. Cell-derived extracellular matrix (ECM) has drawn attention due to its lower risk of immunogenic response and independence from donor availability. The goal of this study is to coat bone mesenchymal stem cell-derived ECMs on poly(lactic-co-glycolic) acid (PLGA) conduits to enhance their ability to support neural growth and neurite extensions. The ECM-coated conduits have better hydrophilic properties than the pure PLGA conduits. A marked increase on PC12 and RSC96 cells' viability, proliferation and dorsal root ganglion neurite extension was observed. Quantitative PCR analysis exhibited a significant increase in markers for cell proliferation (GAP43), neurite extension (NF-H, MAP2, andßIII-tubulin) and neural function (TREK-1). These results show the potential of ECM-coated PLGA conduits in PNI therapy.


Subject(s)
Cell Proliferation , Cell Survival , Extracellular Matrix , Mesenchymal Stem Cells , Nerve Regeneration , Neurites , Polylactic Acid-Polyglycolic Acid Copolymer , Animals , Rats , Neurites/metabolism , PC12 Cells , Extracellular Matrix/metabolism , Mesenchymal Stem Cells/cytology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Nerve Regeneration/drug effects , Tissue Scaffolds/chemistry , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Ganglia, Spinal , Peripheral Nerve Injuries/therapy , Tissue Engineering/methods , Polymers/chemistry , Materials Testing
5.
J Med Virol ; 96(8): e29821, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39175267

ABSTRACT

Herpes zoster (HZ), resulting from the reactivation of the varicella-zoster virus, is a significant disease. This study aimed to explore the factors influencing sensory neuron involvement in HZ at different locations and its association with postherpetic neuralgia (PHN). A total of 3143 cases were retrieved from an electronic medical record system, including 2676 cases of HZ and 467 cases of PHN. Gender, age, site of onset, past surgical history, and comorbidities were analyzed using a multifactorial logistic regression model. The results revealed correlations between age, gender, comorbidities (diabetes, coronary heart disease, percutaneous coronary intervention [PCI]), and sensory neuron involvement in HZ. Specifically, older age, female gender, and comorbid conditions such as diabetes/coronary heart disease were associated with sacral dorsal root ganglion (DRG) involvement, while PCI history was associated with lumbar DRG involvement. Additionally, sensory neuron involvement at different locations by HZ was linked to PHN. Furthermore, independent risk factors for PHN included thoracic DRG involvement, older age, and comorbidities (diabetes, surgical history, malignancy). It is crucial to prevent damage to the DRG, especially in individuals with comorbidities, through activities avoidance and active treatment, to minimize the occurrence of PHN.


Subject(s)
Herpes Zoster , Neuralgia, Postherpetic , Humans , Herpes Zoster/epidemiology , Herpes Zoster/complications , Male , Female , Aged , Middle Aged , Retrospective Studies , Neuralgia, Postherpetic/epidemiology , Risk Factors , Aged, 80 and over , Adult , Comorbidity , Ganglia, Sensory/virology , Herpesvirus 3, Human , Age Factors , Ganglia, Spinal/virology , Young Adult , Sex Factors
6.
BMC Pharmacol Toxicol ; 25(1): 53, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169383

ABSTRACT

BACKGROUND: Econazole is a widely used imidazole derivative antifungal for treating skin infections. The molecular targets for its frequent adverse effects of skin irritation symptoms, such as pruritus, burning sensation, and pain, have not been clarified. Transient receptor potential (TRP) channels, non-selective cation channels, are mainly expressed in peripheral sensory neurons and serve as sensors for various irritants. METHODS: We investigated the effect of econazole on TRP channel activation by measuring intracellular calcium concentration ([Ca2+]i) through fluorescent ratio imaging in mouse dorsal root ganglion (DRG) neurons isolated from wild-type, TRPA1(-/-) and TRPV1(-/-) mice, as well as in heterologously TRP channel-expressed cells. A cheek injection model was employed to assess econazole-induced itch and pain in vivo. RESULTS: Econazole evoked an increase in [Ca2+]i, which was abolished by the removal of extracellular Ca2+ in mouse DRG neurons. The [Ca2+]i responses to econazole were suppressed by a TRPA1 blocker but not by a TRPV1 blocker. Attenuation of the econazole-induced [Ca2+]i responses was observed in the TRPA1(-/-) mouse DRG neurons but was not significant in the TRPV1(-/-) neurons. Econazole increased the [Ca2+]i in HEK293 cells expressing TRPA1 (TRPA1-HEK) but not in those expressing TRPV1, although at higher concentrations, it induced Ca2+ mobilization from intracellular stores in untransfected naïve HEK293 cells. Miconazole, which is a structural analog of econazole, also increased the [Ca2+]i in mouse DRG neurons and TRPA1-HEK, and its nonspecific action was larger than econazole. Fluconazole, a triazole drug failed to activate TRPA1 and TRPV1 in mouse DRG neurons and TRPA1-HEK. Econazole induced itch and pain in wild-type mice, with reduced responses in TRPA1(-/-) mice. CONCLUSIONS: These findings suggested that the imidazole derivatives econazole and miconazole may induce skin irritation by activating nociceptive TRPA1 in the sensory neurons. Suppression of TRPA1 activation may mitigate the adverse effects of econazole.


Subject(s)
Antifungal Agents , Calcium , Econazole , Ganglia, Spinal , Sensory Receptor Cells , TRPA1 Cation Channel , TRPV Cation Channels , Transient Receptor Potential Channels , Animals , Econazole/pharmacology , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/genetics , Antifungal Agents/toxicity , Antifungal Agents/pharmacology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/cytology , Humans , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/genetics , HEK293 Cells , Calcium/metabolism , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Mice , Male , Mice, Knockout , Mice, Inbred C57BL , Pruritus/chemically induced , Pain/drug therapy
7.
Science ; 385(6708): eadk1679, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39088603

ABSTRACT

Neuroimmune cross-talk participates in intestinal tissue homeostasis and host defense. However, the matrix of interactions between arrays of molecularly defined neuron subsets and of immunocyte lineages remains unclear. We used a chemogenetic approach to activate eight distinct neuronal subsets, assessing effects by deep immunophenotyping, microbiome profiling, and immunocyte transcriptomics in intestinal organs. Distinct immune perturbations followed neuronal activation: Nitrergic neurons regulated T helper 17 (TH17)-like cells, and cholinergic neurons regulated neutrophils. Nociceptor neurons, expressing Trpv1, elicited the broadest immunomodulation, inducing changes in innate lymphocytes, macrophages, and RORγ+ regulatory T (Treg) cells. Neuroanatomical, genetic, and pharmacological follow-up showed that Trpv1+ neurons in dorsal root ganglia decreased Treg cell numbers via the neuropeptide calcitonin gene-related peptide (CGRP). Given the role of these neurons in nociception, these data potentially link pain signaling with gut Treg cell function.


Subject(s)
Calcitonin Gene-Related Peptide , Ganglia, Spinal , Neuroimmunomodulation , Nociceptors , T-Lymphocytes, Regulatory , TRPV Cation Channels , Th17 Cells , Animals , Mice , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/genetics , Cholinergic Neurons/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/cytology , Gastrointestinal Microbiome , Intestines/immunology , Intestines/cytology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Nociception , Nociceptors/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/immunology , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics
8.
Pain Res Manag ; 2024: 2437396, 2024.
Article in English | MEDLINE | ID: mdl-39104725

ABSTRACT

Tuina is a treatment method in traditional Chinese medicine which has analgesic effects and effectively alleviates the symptoms of neuropathic pain (NP). Transient receptor potential vanilloid type 1 (TRPV1) and transient receptor potential ankyrin type 1 (TRPA1) play major roles in transmitting nociceptive sensory signals in the nociceptive primary sensory dorsal root ganglion (DRG) nerve. The nitric oxide (NO)/cyclic guanosine 3',5'-monophosphate(cGMP) pathway exerts both nociceptive and antinociceptive effects in various chronic pain models. TRPV1 and TRPA1 mediate the influx of calcium, which stimulates the generation of NO. Subsequently, NO activates the NO/cGMP/protein kinase G (PKG) signaling pathway, thereby improving hyperalgesia. In the present study, oa rat model of NP with minor chronic constriction injury (CCI) of the right sciatic nerve of NP was established. The results of behavioral testing showed that, after a one-time tuina intervention, the mechanical withdrawal threshold (MWT) and thermal withdrawal latency (TWL) were prolonged to varying degrees in the tuina group compared with the model group. Similarly, the expression of TRPV1, TRPA1, NO, soluble guanylate cyclase ß (sGCß), cGMP, and PKG1 was significantly decreased in the DRG of the tuina and tuina + TRPV1/TRPA1 antagonist group was significantly decreased. These findings suggest that the tuina intervention can effectively improve the symptoms of thermal and mechanical allodynia caused by peripheral nerve injuries. Tuina exerts immediate analgesic effects through the TRPV1/TRPA1-NO-cGMP-PKG signaling pathway.


Subject(s)
Cyclic GMP , Disease Models, Animal , Ganglia, Spinal , Rats, Sprague-Dawley , Signal Transduction , TRPA1 Cation Channel , TRPV Cation Channels , Animals , Ganglia, Spinal/metabolism , TRPV Cation Channels/metabolism , Male , Cyclic GMP/metabolism , TRPA1 Cation Channel/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Rats , Neuralgia/metabolism , Neuralgia/drug therapy , Analgesics/pharmacology , Analgesics/therapeutic use , Hyperalgesia/metabolism , Hyperalgesia/drug therapy , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Medicine, Chinese Traditional
10.
Cell Rep Methods ; 4(8): 100835, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39116883

ABSTRACT

We developed a rat dorsal root ganglion (DRG)-derived sensory nerve organotypic model by culturing DRG explants on an organoid culture device. With this method, a large number of organotypic cultures can be produced simultaneously with high reproducibility simply by seeding DRG explants derived from rat embryos. Unlike previous DRG explant models, this organotypic model consists of a ganglion and an axon bundle with myelinated A fibers, unmyelinated C fibers, and stereo-myelin-forming nodes of Ranvier. The model also exhibits Ca2+ signaling in cell bodies in response to application of chemical stimuli to nerve terminals. Further, axonal transection increases the activating transcription factor 3 mRNA level in ganglia. Axons and myelin are shown to regenerate 14 days following transection. Our sensory organotypic model enables analysis of neuronal excitability in response to pain stimuli and tracking of morphological changes in the axon bundle over weeks.


Subject(s)
Axons , Ganglia, Spinal , Microphysiological Systems , Animals , Rats , Activating Transcription Factor 3 , Axons/physiology , Axons/metabolism , Calcium Signaling , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Myelin Sheath/physiology , Myelin Sheath/metabolism , Organoids/metabolism , Peripheral Nerves/metabolism , Rats, Sprague-Dawley , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology
11.
Neuropharmacology ; 259: 110120, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39159835

ABSTRACT

A large portion of neuropathic pain suffering patients may also concurrently experience neuropathic itch, with a negative impact on the quality of life. The limited understanding of neuropathic itch and the low efficacy of current anti-itch therapies dictate the urgent need of a better comprehension of molecular mechanisms involved and development of relevant animal models. This study was aimed to characterize the itching phenotype in a model of trauma-induced peripheral neuropathy, the spared nerve injury (SNI), and the molecular events underlying the overlap with the nociceptive behavior. SNI mice developed hyperknesis and spontaneous itch 7-14 days after surgery that was prevented by gabapentin treatment. Itch was associated with pain hypersensitivity, loss of intraepidermal nerve fiber (IENF) density and increased epidermal thickness. In coincidence with the peak of scratching behavior, SNI mice showed a spinal overexpression of IBA1 and GFAP, microglia and astrocyte markers respectively. An increase of the itch neuropeptide B-type natriuretic peptide (BNP) in NeuN+ cells, of its downstream effector interleukin 17 (IL17) along with increased pERK1/2 levels occurred in the spinal cord dorsal horn and DRG. A raise in BNP and IL17 was also detected at skin level. Stimulation of HaCat cells with conditioned medium from BV2-stimulated SH-SY5Y cells produced a dramatic reduction of HaCat cell viability. This study showed that SNI mice might represent a model for neuropathic itch and pain. Collectively, our finding suggest that neuropathic itch might initiate at spinal level, then affecting skin epidermis events, through a glia-mediated neuroinflammation-evoked BNP/IL17 mechanism.


Subject(s)
Disease Models, Animal , Neuralgia , Neuroinflammatory Diseases , Pruritus , Animals , Pruritus/metabolism , Pruritus/pathology , Neuralgia/metabolism , Neuralgia/etiology , Mice , Male , Neuroinflammatory Diseases/metabolism , Humans , Gabapentin/pharmacology , Interleukin-17/metabolism , Mice, Inbred C57BL , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/pathology , HaCaT Cells , Microglia/metabolism , Microglia/drug effects , Hyperalgesia/metabolism , Microfilament Proteins/metabolism , Spinal Cord Dorsal Horn/metabolism , Spinal Cord Dorsal Horn/drug effects , Calcium-Binding Proteins
12.
Sci Rep ; 14(1): 19016, 2024 08 16.
Article in English | MEDLINE | ID: mdl-39152157

ABSTRACT

Peripheral nerve injury (PNI) often leads to retrograde cell death in the spinal cord and dorsal root ganglia (DRG), hindering nerve regeneration and functional recovery. Repetitive magnetic stimulation (rMS) promotes nerve regeneration following PNI. Therefore, this study aimed to investigate the effects of rMS on post-injury neuronal death and nerve regeneration. Seventy-two rats underwent autologous sciatic nerve grafting and were divided into two groups: the rMS group, which received rMS and the control (CON) group, which received no treatment. Motor neuron, DRG neuron, and caspase-3 positive DRG neuron counts, as well as DRG mRNA expression analyses, were conducted at 1-, 4-, and 8-weeks post-injury. Functional and axon regeneration analyses were performed at 8-weeks post-injury. The CON group demonstrated a decreased DRG neuron count starting from 1 week post-injury, whereas the rMS group exhibited significantly higher DRG neuron counts at 1- and 4-weeks post-injury. At 8-weeks post-injury, the rMS group demonstrated a significantly greater myelinated nerve fiber density in autografted nerves. Furthermore, functional analysis showed significant improvements in latency and toe angle in the rMS group. Overall, these results suggest that rMS can prevent DRG neuron death and enhance nerve regeneration and motor function recovery after PNI.


Subject(s)
Cell Death , Disease Models, Animal , Ganglia, Spinal , Nerve Regeneration , Peripheral Nerve Injuries , Sciatic Nerve , Animals , Ganglia, Spinal/metabolism , Rats , Sciatic Nerve/injuries , Peripheral Nerve Injuries/therapy , Male , Rats, Sprague-Dawley , Neurons/metabolism , Magnetic Field Therapy/methods , Recovery of Function , Motor Neurons/metabolism , Motor Neurons/physiology
13.
Sci Rep ; 14(1): 18077, 2024 08 05.
Article in English | MEDLINE | ID: mdl-39103432

ABSTRACT

Insulin has been shown to modulate neuronal processes through insulin receptors. The ion channels located on neurons may be important targets for insulin/insulin receptor signaling. Both insulin receptors and acid-sensing ion channels (ASICs) are expressed in dorsal root ganglia (DRG) neurons. However, it is still unclear whether there is an interaction between them. Therefore, the purpose of this investigation was to determine the effects of insulin on the functional activity of ASICs. A 5 min application of insulin rapidly enhanced acid-evoked ASIC currents in rat DRG neurons in a concentration-dependent manner. Insulin shifted the concentration-response plot for ASIC currents upward, with an increase of 46.2 ± 7.6% in the maximal current response. The insulin-induced increase in ASIC currents was eliminated by the insulin receptor antagonist GSK1838705, the tyrosine kinase inhibitor lavendustin A, and the phosphatidylinositol-3 kinase antagonist wortmannin. Moreover, insulin increased the number of acid-triggered action potentials by activating insulin receptors. Finally, local administration of insulin exacerbated the spontaneous nociceptive behaviors induced by intraplantar acid injection and the mechanical hyperalgesia induced by intramuscular acid injections through peripheral insulin receptors. These results suggested that insulin/insulin receptor signaling enhanced the functional activity of ASICs via tyrosine kinase and phosphatidylinositol-3 kinase pathways. Our findings revealed that ASICs were targets in primary sensory neurons for insulin receptor signaling, which may underlie insulin modulation of pain.


Subject(s)
Acid Sensing Ion Channels , Ganglia, Spinal , Insulin , Receptor, Insulin , Sensory Receptor Cells , Animals , Acid Sensing Ion Channels/metabolism , Insulin/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/cytology , Rats , Receptor, Insulin/metabolism , Male , Signal Transduction/drug effects , Action Potentials/drug effects , Rats, Sprague-Dawley , Hyperalgesia/metabolism , Cells, Cultured
14.
Turk J Gastroenterol ; 35(6): 453-464, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-39114901

ABSTRACT

The pathogenesis mechanism of acute gastric mucosal lesions (AGML) is still unclear; further exploration is urgently needed to find a new therapeutic target. This study aimed to investigate whether morphine might regulate the expression and function of transient receptor potential ankyrin 1 (TRPA1) through a cyclic adenosine monophosphate/protein kinase A (cAMP/PKA)-dependent pathway, thereby alleviating gastric mucosal lesions caused by water-immersion restraint stress (WIRS). Rats were administered with intrathecal morphine, TRPA1 antagonist (HC-030031), µ-opioid receptor antagonist, or protein kinase A inhibitor (H-89), respectively, before WIRS. After 6 hours of WIRS, microscopic lesions, hematoxylin and eosin staining, and transmission electron microscopy were applied to assess the damage of the gastric mucosa. Real-time polymerase chain reaction, Western blot, and enzyme-linked immunosorbent assay were conducted to detect the levels of TRPA1 and substance P (SP) in the dorsal root ganglia (DRG) and gastric tissues. In addition, immunofluorescence was used to explore the possible co-expression of TRPA1 and µ-opioid receptors in the DRG. The results indicated that WIRS upregulated TRPA1 and SP in gastric mucosa, and HC-030031 or H-89 could alleviate gastric mucosal lesions caused by WIRS (P < .0001). Morphine was found to suppress both WIRS-induced gastric mucosal lesions (P < .0001) and the upregulation of TRPA1 (P = .0086) and SP (P = .0013). Both TRPA1 and SP play important roles in the pathogenesis of WIRS-induced AGML. Exogenous gastroprotective strategies reduce elevated levels of TRPA1 via the cAMP/PKA-dependent pathway. Inhibition of TRPA1 upregulation in the DRG is critical for intrathecal morphine preconditioning-induced gastric protection.


Subject(s)
Ganglia, Spinal , Gastric Mucosa , Isoquinolines , Morphine , Rats, Sprague-Dawley , Restraint, Physical , TRPA1 Cation Channel , Up-Regulation , Animals , Morphine/pharmacology , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Up-Regulation/drug effects , TRPA1 Cation Channel/metabolism , Male , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Restraint, Physical/adverse effects , Rats , Isoquinolines/pharmacology , Acetanilides/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Purines/pharmacology , Stress, Psychological/complications , Immersion , Receptors, Opioid, mu/metabolism , Cyclic AMP/metabolism , Sulfonamides
15.
J Neuroimmune Pharmacol ; 19(1): 46, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39162886

ABSTRACT

The mechanisms for neuropathic pain amelioration by sigma-1 receptor inhibition are not fully understood. We studied genome-wide transcriptomic changes (RNAseq) in the dorsal root ganglia (DRG) from wild-type and sigma-1 receptor knockout mice prior to and following Spared Nerve Injury (SNI). In wildtype mice, most of the transcriptomic changes following SNI are related to the immune function or neurotransmission. Immune function transcripts contain cytokines and markers for immune cells, including macrophages/monocytes and CD4 + T cells. Many of these immune transcripts were attenuated by sigma-1 knockout in response to SNI. Consistent with this we found, using flow cytometry, that sigma-1 knockout mice showed a reduction in macrophage/monocyte recruitment as well as an absence of CD4 + T cell recruitment in the DRG after nerve injury. Sigma-1 knockout mice showed a reduction of neuropathic (mechanical and cold) allodynia and spontaneous pain-like responses (licking of the injured paw) which accompany the decreased peripheral neuroinflammatory response after nerve injury. Treatment with maraviroc (a CCR5 antagonist which preferentially inhibits CD4 + T cells in the periphery) of neuropathic wild-type mice only partially replicated the sigma-1 knockout phenotype, as it did not alter cold allodynia but attenuated spontaneous pain-like responses and mechanical hypersensitivity. Therefore, modulation of peripheral CD4 + T cell activity might contribute to the amelioration of spontaneous pain and neuropathic tactile allodynia seen in the sigma-1 receptor knockout mice, but not to the effect on cold allodynia. We conclude that sigma-1 receptor inhibition decreases DRG neuroinflammation which might partially explain its anti-neuropathic effect.


Subject(s)
Mice, Inbred C57BL , Mice, Knockout , Neuralgia , Receptors, sigma , Sigma-1 Receptor , Transcriptome , Animals , Receptors, sigma/genetics , Receptors, sigma/metabolism , Receptors, sigma/antagonists & inhibitors , Neuralgia/metabolism , Mice , Female , Neuroinflammatory Diseases/metabolism , Ganglia, Spinal/metabolism , Peripheral Nerve Injuries/metabolism
16.
Nat Commun ; 15(1): 7065, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39152112

ABSTRACT

The sympathetic nervous system controls bodily functions including vascular tone, cardiac rhythm, and the "fight-or-flight response". Sympathetic chain ganglia develop in parallel with preganglionic motor nerves extending from the neural tube, raising the question of whether axon targeting contributes to sympathetic chain formation. Using nerve-selective genetic ablations and lineage tracing in mouse, we reveal that motor nerve-associated Schwann cell precursors (SCPs) contribute sympathetic neurons and satellite glia after the initial seeding of sympathetic ganglia by neural crest. Motor nerve ablation causes mispositioning of SCP-derived sympathoblasts as well as sympathetic chain hypoplasia and fragmentation. Sympathetic neurons in motor-ablated embryos project precociously and abnormally towards dorsal root ganglia, eventually resulting in fusion of sympathetic and sensory ganglia. Cell interaction analysis identifies semaphorins as potential motor nerve-derived signaling molecules regulating sympathoblast positioning and outgrowth. Overall, central innervation functions both as infrastructure and regulatory niche to ensure the integrity of peripheral ganglia morphogenesis.


Subject(s)
Ganglia, Sympathetic , Motor Neurons , Neural Crest , Schwann Cells , Sympathetic Nervous System , Animals , Sympathetic Nervous System/embryology , Mice , Motor Neurons/physiology , Schwann Cells/metabolism , Neural Crest/cytology , Neural Crest/metabolism , Ganglia, Sympathetic/cytology , Ganglia, Spinal , Semaphorins/metabolism , Semaphorins/genetics , Mice, Transgenic , Neuroglia/metabolism , Female
17.
CNS Neurosci Ther ; 30(9): e70012, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39215404

ABSTRACT

AIMS: Poly (ADP-ribose) polymerase (PARP) has been extensively investigated in human cancers. Recent studies verified that current available PARP inhibitors (Olaparib or Veliparib) provided clinical palliation of clinical patients suffering from paclitaxel-induced neuropathic pain (PINP). However, the underlying mechanism of PARP overactivation in the development of PINP remains to be investigated. METHODS AND RESULTS: We reported induction of DNA oxidative damage, PARP-1 overactivation, and subsequent nicotinamide adenine dinucleotide (NAD+) depletion as crucial events in the pathogenesis of PINP. Therefore, we developed an Olaparib PROTAC to achieve the efficient degradation of PARP. Continuous intrathecal injection of Olaparib PROTAC protected against PINP by inhibiting the activity of PARP-1 in rats. PARP-1, but not PARP-2, was shown to be a crucial enzyme in the development of PINP. Specific inhibition of PARP-1 enhanced mitochondrial redox metabolism partly by upregulating the expression and deacetylase activity of sirtuin-3 (SIRT3) in the dorsal root ganglions and spinal cord in the PINP rats. Moreover, an increase in the NAD+ level was found to be a crucial mechanism by which PARP-1 inhibition enhanced SIRT3 activity. CONCLUSION: The findings provide a novel insight into the mechanism of DNA oxidative damage in the development of PINP and implicate PARP-1 as a possible therapeutic target for clinical PINP treatment.


Subject(s)
DNA Damage , Mitochondria , Neuralgia , Paclitaxel , Poly (ADP-Ribose) Polymerase-1 , Animals , Male , Rats , Disease Models, Animal , DNA Damage/drug effects , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , NAD/metabolism , Neuralgia/chemically induced , Neuralgia/metabolism , Neuralgia/drug therapy , Oxidative Stress/drug effects , Paclitaxel/toxicity , Phthalazines/pharmacology , Piperazines/pharmacology , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/metabolism
18.
Cell Commun Signal ; 22(1): 416, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39192337

ABSTRACT

Bone cancer pain (BCP) represents a prevalent symptom among cancer patients with bone metastases, yet its underlying mechanisms remain elusive. This study investigated the transcriptional regulation mechanism of Kv7(KCNQ)/M potassium channels in DRG neurons and its involvement in the development of BCP in rats. We show that HDAC2-mediated transcriptional repression of kcnq2/kcnq3 genes, which encode Kv7(KCNQ)/M potassium channels in dorsal root ganglion (DRG), contributes to the sensitization of DRG neurons and the pathogenesis of BCP in rats. Also, HDAC2 requires the formation of a corepressor complex with MeCP2 and Sin3A to execute transcriptional regulation of kcnq2/kcnq3 genes. Moreover, EREG is identified as an upstream signal molecule for HDAC2-mediated kcnq2/kcnq3 genes transcription repression. Activation of EREG/EGFR-ERK-Runx1 signaling, followed by the induction of HDAC2-mediated transcriptional repression of kcnq2/kcnq3 genes in DRG neurons, leads to neuronal hyperexcitability and pain hypersensitivity in tumor-bearing rats. Consequently, the activation of EREG/EGFR-ERK-Runx1 signaling, along with the subsequent transcriptional repression of kcnq2/kcnq3 genes by HDAC2 in DRG neurons, underlies the sensitization of DRG neurons and the pathogenesis of BCP in rats. These findings uncover a potentially targetable mechanism contributing to bone metastasis-associated pain in cancer patients.


Subject(s)
Bone Neoplasms , Cancer Pain , ErbB Receptors , Ganglia, Spinal , Histone Deacetylase 2 , KCNQ2 Potassium Channel , Animals , Histone Deacetylase 2/metabolism , Histone Deacetylase 2/genetics , KCNQ2 Potassium Channel/genetics , KCNQ2 Potassium Channel/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Bone Neoplasms/pathology , Rats , Cancer Pain/genetics , Cancer Pain/metabolism , Cancer Pain/pathology , ErbB Receptors/metabolism , ErbB Receptors/genetics , KCNQ3 Potassium Channel/genetics , KCNQ3 Potassium Channel/metabolism , Transcription, Genetic , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Sin3 Histone Deacetylase and Corepressor Complex/genetics , Signal Transduction/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Humans , Female , Extracellular Signal-Regulated MAP Kinases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Rats, Sprague-Dawley , MAP Kinase Signaling System/genetics
19.
Front Immunol ; 15: 1396000, 2024.
Article in English | MEDLINE | ID: mdl-39192982

ABSTRACT

Endometriosis is a chronic inflammatory disease that causes debilitating pelvic pain in women. Macrophages are considered to be key players in promoting disease progression, as abundant macrophages are present in ectopic lesions and elevated in the peritoneum. In the present study, we examined the role of GATA6+ peritoneal macrophages on endometriosis-associated hyperalgesia using mice with a specific myeloid deficiency of GATA6. Lesion induction induced the disappearance of TIM4hi MHCIIlo residential macrophages and the influx of increased Ly6C+ monocytes and TIM4lo MHCIIhi macrophages. The recruitment of MHCIIhi inflammatory macrophages was extensive in Mac Gata6 KO mice due to the severe disappearance of TIM4hi MHCIIlo residential macrophages. Ki67 expression confirmed GATA6-dependent proliferative ability, showing different proliferative phenotypes of TIM4+ residential macrophages in Gata6f/f and Mac Gata6 KO mice. Peritoneal proinflammatory cytokines were elevated after lesion induction. When cytokine levels were compared between Gata6f/f and Mac Gata6 KO mice, TNFα at day 21 in Gata6f/f mice was higher than in Mac Gata6 KO mice. Lesion induction increased both abdominal and hind paw sensitivities. Gata6f/f mice tended to show higher sensitivity in the abdomen after day 21. Elevated expression of TRPV1 and CGRP was observed in the dorsal root ganglia after ELL induction in Gata6f/f mice until days 21 and 42, respectively. These results support that peritoneal GATA6+ macrophages are involved in the recruitment and reprogramming of monocyte-derived macrophages. The extensive recruitment of monocyte-derived macrophages in Mac Gata6 KO mice might protect against inflammatory stimuli during the resolution phase, whereas GATA6 deficiency did not affect lesion initiation and establishment at the acute phase of inflammation. GATA6+ residential macrophages act to sustain local inflammation in the peritoneum and sensitivities in the neurons, reflecting endometriosis-associated hyperalgesia.


Subject(s)
Endometriosis , GATA6 Transcription Factor , Macrophages, Peritoneal , Animals , Female , Mice , Cytokines/metabolism , Disease Models, Animal , Endometriosis/immunology , Endometriosis/pathology , Endometriosis/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/immunology , GATA6 Transcription Factor/metabolism , GATA6 Transcription Factor/genetics , Hyperalgesia/etiology , Hyperalgesia/metabolism , Hyperalgesia/immunology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice, Inbred C57BL , Mice, Knockout , Peritoneum/pathology , Peritoneum/immunology , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics
20.
Proc Natl Acad Sci U S A ; 121(36): e2411846121, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39190348

ABSTRACT

Optogenetic techniques provide genetically targeted, spatially and temporally precise approaches to correlate cellular activities and physiological outcomes. In the nervous system, G protein-coupled receptors (GPCRs) have essential neuromodulatory functions through binding extracellular ligands to induce intracellular signaling cascades. In this work, we develop and validate an optogenetic tool that disrupts Gαq signaling through membrane recruitment of a minimal regulator of G protein signaling (RGS) domain. This approach, Photo-induced Gα Modulator-Inhibition of Gαq (PiGM-Iq), exhibited potent and selective inhibition of Gαq signaling. Using PiGM-Iq we alter the behavior of Caenorhabditis elegans and Drosophila with outcomes consistent with GPCR-Gαq disruption. PiGM-Iq changes axon guidance in cultured dorsal root ganglia neurons in response to serotonin. PiGM-Iq activation leads to developmental deficits in zebrafish embryos and larvae resulting in altered neuronal wiring and behavior. Furthermore, by altering the minimal RGS domain, we show that this approach is amenable to Gαi signaling. Our unique and robust optogenetic Gα inhibiting approaches complement existing neurobiological tools and can be used to investigate the functional effects neuromodulators that signal through GPCR and trimeric G proteins.


Subject(s)
Caenorhabditis elegans , GTP-Binding Protein alpha Subunits, Gq-G11 , Optogenetics , RGS Proteins , Signal Transduction , Zebrafish , Animals , Optogenetics/methods , Caenorhabditis elegans/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , RGS Proteins/metabolism , RGS Proteins/genetics , Zebrafish/embryology , Neurons/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Protein Domains , Ganglia, Spinal/metabolism , Ganglia, Spinal/cytology , Drosophila/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL