Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 4.632
1.
Neuron ; 112(11): 1727-1729, 2024 Jun 05.
Article En | MEDLINE | ID: mdl-38843778

While effective analgesics, TRPV1 antagonists can dangerously alter thermoregulation. In this issue of Neuron, Huang et al.1 demonstrate that interaction with the S4-S5 linker of TRPV1 determines whether an antagonist affects core body temperature, with promising implications for analgesic development.


Body Temperature Regulation , Hyperthermia , TRPV Cation Channels , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism , Hyperthermia/chemically induced , Animals , Body Temperature Regulation/drug effects , Body Temperature Regulation/physiology , Humans , Body Temperature/drug effects , Analgesics/pharmacology
2.
FASEB J ; 38(11): e23731, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38855909

Ca2+ permeation through TRPV4 in fibroblasts is associated with pathological matrix degradation. In human gingival fibroblasts, IL-1ß binding to its signaling receptor (IL-1R1) induces activation of extracellular regulated kinase (ERK) and MMP1 expression, processes that require Ca2+ flux across the plasma membrane. It is not known how IL-1R1, which does not conduct Ca2+, generates Ca2+ signals in response to IL-1. We examined whether TRPV4 mediates the Ca2+ fluxes required for ERK signaling in IL-1 stimulated gingival fibroblasts. TRPV4 was immunostained in fibroblasts of human gingival connective tissue and in focal adhesions of cultured mouse gingival fibroblasts. Human gingival fibroblasts treated with IL-1ß showed no change of TRPV4 expression but there was increased MMP1 expression. In mouse, gingival fibroblasts expressing TRPV4, IL-1 strongly increased [Ca2+]i. Pre-incubation of cells with IL-1 Receptor Antagonist blocked Ca2+ entry induced by IL-1 or the TRPV4 agonist GSK101. Knockout of TRPV4 or expression of a non-Ca2+-conducting TRPV4 pore-mutant or pre-incubation with the TRPV4 inhibitor RN1734, blocked IL-1-induced Ca2+ transients and expression of the mouse interstitial collagenase, MMP13. Treatment of mouse gingival fibroblasts with GSK101 phenocopied Ca2+ and ERK responses induced by IL-1; these responses were absent in TRPV4-null cells or cells expressing a non-conducting TRPV4 pore-mutant. Immunostained IL-1R1 localized with TRPV4 in adhesions within cell extensions. While TRPV4 immunoprecipitates analyzed by mass spectrometry showed no association with IL-1R1, TRPV4 associated with Src-related proteins and Src co-immunoprecipitated with TRPV4. Src inhibition reduced IL-1-induced Ca2+ responses. The functional linkage of TRPV4 with IL-1R1 expands its repertoire of innate immune signaling processes by mediating IL-1-driven Ca2+ responses that drive matrix remodeling in fibroblasts. Thus, inhibiting TRPV4 activity may provide a new pharmacological approach for blunting matrix degradation in inflammatory diseases.


Calcium Signaling , Fibroblasts , Gingiva , TRPV Cation Channels , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Animals , Humans , Mice , Fibroblasts/metabolism , Gingiva/metabolism , Gingiva/cytology , Calcium/metabolism , MAP Kinase Signaling System , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-1/metabolism , Interleukin-1/pharmacology , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 1/genetics , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology
3.
Braz J Med Biol Res ; 57: e13238, 2024.
Article En | MEDLINE | ID: mdl-38808885

Osteoarthritis (OA) is a highly prevalent joint disorder characterized by progressive degeneration of articular cartilage, subchondral bone remodeling, osteophyte formation, synovial inflammation, and meniscal damage. Although the etiology of OA is multifactorial, pro-inflammatory processes appear to play a key role in disease pathogenesis. Previous studies indicate that electroacupuncture (EA) exerts chondroprotective, anti-inflammatory, and analgesic effects in preclinical models of OA, but the mechanisms underlying these potential therapeutic benefits remain incompletely defined. This study aimed to investigate the effects of EA on OA development in a rat model, as well as to explore associated molecular mechanisms modulated by EA treatment. Forty rats were divided into OA, EA, antagomiR-214, and control groups. Following intra-articular injection of monosodium iodoacetate to induce OA, EA and antagomiR-214 groups received daily EA stimulation at acupoints around the knee joint for 21 days. Functional pain behaviors and chondrocyte apoptosis were assessed as outcome measures. The expression of microRNA-214 (miR-214) and its downstream targets involved in apoptosis and nociception, BAX and TRPV4, were examined. Results demonstrated that EA treatment upregulated miR-214 expression in OA knee cartilage. By suppressing pro-apoptotic BAX and pro-nociceptive TRPV4, this EA-induced miR-214 upregulation ameliorated articular pain and prevented chondrocyte apoptosis. These findings suggested that miR-214 plays a key role mediating EA's therapeutic effects in OA pathophysiology, and represents a promising OA treatment target for modulation by acupuncture.


Apoptosis , Chondrocytes , Disease Models, Animal , Electroacupuncture , MicroRNAs , Osteoarthritis , Rats, Sprague-Dawley , TRPV Cation Channels , bcl-2-Associated X Protein , Animals , TRPV Cation Channels/metabolism , MicroRNAs/metabolism , Electroacupuncture/methods , Male , Osteoarthritis/therapy , Chondrocytes/metabolism , bcl-2-Associated X Protein/metabolism , Rats
4.
Front Biosci (Landmark Ed) ; 29(5): 175, 2024 May 07.
Article En | MEDLINE | ID: mdl-38812310

Dry eye disease (DED) is a prevalent ophthalmic ailment with intricate pathogenesis and that occurs primarily due to various factors which affect the ocular surface. DED is characterized by the disruption of tear film homeostasis, inflammatory reaction, and neuroparesthesia. Transient receptor potential vanilloid 1 (TRPV1) is a versatile receptor that can be stimulated by heat, acid, capsaicin (CAP), hyperosmolarity, and numerous inflammatory agents. There is accumulating evidence that implicates TRPV1 in the initiation and progression of DED through its detection of hypertonic conditions and modulation of inflammatory pathways. In this article, we present a comprehensive review of the expression and function of the TRPV1 channel in tissues and cells associated with DED. In addition, we outline the potential mechanisms that implicate TRPV1 in the pathophysiology of DED. The aim of this review is to establish a theoretical basis for TRPV1 as a possible therapeutic target in DED, thereby encouraging further investigations into its role in DED.


Dry Eye Syndromes , TRPV Cation Channels , TRPV Cation Channels/metabolism , Humans , Dry Eye Syndromes/metabolism , Dry Eye Syndromes/physiopathology , Animals
5.
FASEB J ; 38(10): e23661, 2024 May 31.
Article En | MEDLINE | ID: mdl-38733310

Itching is an aversive somatosensation that triggers the desire to scratch. Transient receptor potential (TRP) channel proteins are key players in acute and chronic itch. However, whether the modulatory effect of fibroblast growth factor 13 (FGF13) on acute and chronic itch is associated with TRP channel proteins is unclear. Here, we demonstrated that conditional knockout of Fgf13 in dorsal root ganglion neurons induced significant impairment in scratching behaviors in response to acute histamine-dependent and chronic dry skin itch models. Furthermore, FGF13 selectively regulated the function of the TRPV1, but not the TRPA1 channel on Ca2+ imaging and electrophysiological recordings, as demonstrated by a significant reduction in neuronal excitability and current density induced by TRPV1 channel activation, whereas TRPA1 channel activation had no effect. Changes in channel currents were also verified in HEK cell lines. Subsequently, we observed that selective modulation of TRPV1 by FGF13 required its microtubule-stabilizing effect. Furthermore, in FGF13 knockout mice, only the overexpression of FGF13 with a tubulin-binding domain could rescue TRP channel function and the impaired itch behavior. Our findings reveal a novel mechanism by which FGF13 is involved in TRPV1-dependent itch transduction and provide valuable clues for alleviating pathological itch syndrome.


Fibroblast Growth Factors , Mice, Knockout , Microtubules , Pruritus , TRPV Cation Channels , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Pruritus/metabolism , Pruritus/genetics , Animals , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , Mice , Humans , HEK293 Cells , Microtubules/metabolism , Ganglia, Spinal/metabolism , Male , Mice, Inbred C57BL , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/genetics
6.
Int J Mol Sci ; 25(10)2024 May 11.
Article En | MEDLINE | ID: mdl-38791297

Globally, breast cancer is a significant cause of mortality. Recent research focused on identifying compounds regulating the transient receptor potential vanilloid 1 (TRPV1) ion channel activity for the possibility of developing cancer therapeutics. In this study, the antiproliferative properties and mechanisms of action through TRPV1 of Maclura pomifera, a dioecious tree native to the south-central USA, have been investigated. Male and female extracts of spring branch tissues and leaves (500 µg/mL) significantly reduced the viability of MCF-7 and T47D cells by 75-80%. M. pomifera extracts induced apoptosis by triggering intracellular calcium overload via TRPV1. Blocking TRPV1 with the capsazepine antagonist and pretreating cells with the BAPTA-AM chelator boosted cell viability, revealing that M. pomifera phytochemicals activate TRPV1. Both male and female M. pomifera extracts initiated apoptosis through multiple pathways, the mitochondrial, ERK-induced, and endoplasmic reticulum-stress-mediated apoptotic pathways, demonstrated by the expression of activated caspase 3, caspase 9, caspase 8, FADD, FAS, ATF4, and CHOP, the overexpression of phosphorylated PERK and ERK proteins, and the reduction of BCL-2 levels. In addition, AKT and pAKT protein expressions were reduced in female M. pomifera-treated cells, revealing that female plant extract also inhibits PI3K/Akt signaling pathways. These results suggest that phytochemicals in M. pomifera extracts could be promising for developing breast cancer therapeutics.


Apoptosis , Breast Neoplasms , Cell Proliferation , Plant Extracts , Receptors, Estrogen , TRPV Cation Channels , Humans , Apoptosis/drug effects , TRPV Cation Channels/metabolism , Plant Extracts/pharmacology , Plant Extracts/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Cell Proliferation/drug effects , Receptors, Estrogen/metabolism , Cell Line, Tumor , MCF-7 Cells , Signal Transduction/drug effects , Cell Survival/drug effects
7.
J Histochem Cytochem ; 72(5): 275-287, 2024 05.
Article En | MEDLINE | ID: mdl-38725415

The TRPA1 ion channel is a sensitive detector of reactive chemicals, found primarily on sensory neurons. The phenotype exhibited by mice lacking TRPA1 suggests its potential as a target for pharmacological intervention. Antibody-based detection for distribution analysis is a standard technique. In the case of TRPA1, however, there is no antibody with a plausible validation in knockout animals or functional studies, but many that have failed in this regard. To this end we employed the single molecule in situ hybridization technique RNAscope on sensory neurons immediately after detection of calcium responses to the TRPA1 agonist allyl isothiocyanate. There is a clearly positive correlation between TRPA1 calcium imaging and RNAscope detection (R = 0.43), although less than what might have been expected. Thus, the technique of choice should be carefully considered to suit the research question. The marginal correlation between TRPV1 RNAscope and the specific agonist capsaicin indicates that such validation is advisable for every RNAscope target. Given the recent description of a long-awaited TRPA1 reporter mouse, TRPA1 RNAscope detection might still have its use cases, for detection of RNA at particular sites, for example, defined structurally or by other molecular markers.


Calcium , Isothiocyanates , TRPA1 Cation Channel , Animals , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/genetics , Isothiocyanates/pharmacology , Mice , Calcium/metabolism , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/agonists , Capsaicin/pharmacology , In Situ Hybridization , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/agonists , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Mice, Inbred C57BL , Calcium Channels/metabolism , Calcium Channels/genetics , Male
8.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article En | MEDLINE | ID: mdl-38732143

This study explores low-intensity extracorporeal shock wave therapy (LiESWT)'s efficacy in alleviating detrusor hyperactivity with impaired contractility (DHIC) induced by ovarian hormone deficiency (OHD) in ovariectomized rats. The rats were categorized into the following four groups: sham group; OVX group, subjected to bilateral ovariectomy (OVX) for 12 months to induce OHD; OVX + SW4 group, underwent OHD for 12 months followed by 4 weeks of weekly LiESWT; and OVX + SW8 group, underwent OHD for 12 months followed by 8 weeks of weekly LiESWT. Cystometrogram studies and voiding behavior tracing were used to identify the symptoms of DHIC. Muscle strip contractility was evaluated through electrical-field, carbachol, ATP, and KCl stimulations. Western blot and immunofluorescence analyses were performed to assess the expressions of various markers related to bladder dysfunction. The OVX rats exhibited significant bladder deterioration and overactivity, alleviated by LiESWT. LiESWT modified transient receptor potential vanilloid (TRPV) channel expression, regulating calcium concentration and enhancing bladder capacity. It also elevated endoplasmic reticulum (ER) stress proteins, influencing ER-related Ca2+ channels and receptors to modulate detrusor muscle contractility. OHD after 12 months led to neuronal degeneration and reduced TRPV1 and TRPV4 channel activation. LiESWT demonstrated potential in enhancing angiogenic remodeling, neurogenesis, and receptor response, ameliorating DHIC via TRPV channels and cellular signaling in the OHD-induced DHIC rat model.


Disease Models, Animal , Extracorporeal Shockwave Therapy , Muscle Contraction , TRPV Cation Channels , Urinary Bladder , Animals , Female , Rats , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Extracorporeal Shockwave Therapy/methods , Urinary Bladder/physiopathology , Urinary Bladder/metabolism , Urinary Bladder, Overactive/therapy , Urinary Bladder, Overactive/metabolism , Urinary Bladder, Overactive/physiopathology , Urinary Bladder, Overactive/etiology , Ovariectomy , Rats, Sprague-Dawley , Ovary/metabolism
9.
Brain Res ; 1839: 149008, 2024 Sep 15.
Article En | MEDLINE | ID: mdl-38761846

A synthetic inhibitor of capsaicin-induced TRPV1 channel activation is called capsazepine (CPZ). In this study, we aimed to explore the effects of CPZ on hyperpolarization-activated cationic current (Ih) and voltage-gated Na + current (INa) in pituitary tumor (GH3) cells. Through patch-clamp recordings, we found that CPZ concentration-dependently inhibited Ih amplitude and slowed its activation time course. The IC50 and KD values were 3.1 and 3.16 µM, respectively. CPZ also shifted the steady-state activation curve of Ih towards a more hyperpolarized potential. However, there was no change in the gating charge of the curve. A modified Markovian model predicted the CPZ-induced decrease in the voltage-dependent hysteresis of Ih. CPZ suppressed INa in GH3 cells, without altering its activation or inactivation time course. Additionally, exposure to CPZ reduced spontaneous firing. These findings suggest that CPZ's inhibitory effects on Ih and INa are direct and not dependent on vanilloid receptor binding. This could provide light on an unidentified ionic mechanism influencing the membrane excitability of neurons and endocrine or neuroendocrine cells in vivo.


Capsaicin , TRPV Cation Channels , TRPV Cation Channels/metabolism , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/drug effects , Capsaicin/pharmacology , Capsaicin/analogs & derivatives , Animals , Rats , Cell Line, Tumor , Patch-Clamp Techniques , Membrane Potentials/drug effects , Neurons/drug effects , Neurons/metabolism , Action Potentials/drug effects
10.
Bioorg Med Chem ; 107: 117750, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38776567

Analgesia and blood sugar control are considered as two main unmet clinical needs for diabetes related neuropathic pain patients. Transient receptor potential vanilloid type-1 (TRPV1) channel is a highly validated target for pain perception, while no TRPV1 antagonists have been approved due to hyperthermia side effects. Herein, two series of new TRPV1 antagonists with flavonoid skeleton were designed by the structure-based drug design (SBDD) strategy. After comprehensive evaluation, compound CX-3 was identified as a promising TRPV1 antagonist. CX-3 exhibited equivalent TRPV1 antagonistic activity with classical TRPV1 antagonist BCTC in vitro, and exerted better analgesic activity in vivo than that of BCTC in the formalin induced inflammatory pain model without hyperthermia risk. Moreover, CX-3 exhibited robust glucose-lowering effects and showed high selectivity over other ion channels. Overall, these findings identified a first-in-class highly selective TRPV1 antagonist CX-3, which is a promising candidate to target the pathogenesis of diabetes related neuropathic pain.


Analgesics , Hypoglycemic Agents , TRPV Cation Channels , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism , Analgesics/pharmacology , Analgesics/chemistry , Analgesics/chemical synthesis , Animals , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/chemical synthesis , Humans , Structure-Activity Relationship , Mice , Molecular Structure , Male , Drug Discovery , Dose-Response Relationship, Drug , Rats , Rats, Sprague-Dawley
11.
Nat Commun ; 15(1): 4609, 2024 May 30.
Article En | MEDLINE | ID: mdl-38816425

The protection of the replication fork structure under stress conditions is essential for genome maintenance and cancer prevention. A key signaling pathway for fork protection involves TRPV2-mediated Ca2+ release from the ER, which is triggered after the generation of cytosolic DNA and the activation of cGAS/STING. This results in CaMKK2/AMPK activation and subsequent Exo1 phosphorylation, which prevent aberrant fork processing, thereby ensuring genome stability. However, it remains poorly understood how the TRPV2 channel is activated by the presence of cytosolic DNA. Here, through a genome-wide CRISPR-based screen, we identify TRPM8 channel-associated factor 1 (TCAF1) as a key factor promoting TRPV2-mediated Ca2+ release under replication stress or other conditions that activate cGAS/STING. Mechanistically, TCAF1 assists Ca2+ release by facilitating the dissociation of STING from TRPV2, thereby relieving TRPV2 repression. Consistent with this function, TCAF1 is required for fork protection, chromosomal stability, and cell survival after replication stress.


Calcium , Cytosol , DNA Replication , Membrane Proteins , TRPV Cation Channels , Humans , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Calcium/metabolism , Cytosol/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , HEK293 Cells , DNA/metabolism , HeLa Cells , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Phosphorylation , Genomic Instability , DNA Damage , Animals
12.
Drug Res (Stuttg) ; 74(5): 195-207, 2024 Jun.
Article En | MEDLINE | ID: mdl-38729182

This study examines the fundamental chemical mechanisms responsible for capsaicin's advantageous impact on cancer, specifically investigating its influence on several biological processes such as inflammation in cancer metastasis, apoptosis, angiogenesis, and cellular proliferation. This entity's connections with other signaling pathways, including PI3K/AKT, NF-B, and TRPV channels, which have been linked to tumor growth, are thoroughly examined in this work. This study presents a thorough analysis of preclinical studies and clinical trials investigating the efficacy of capsaicin in treating many forms of cancer, such as breast, prostate, colorectal, pancreatic, and others. Through tests conducted in both live organisms and laboratory settings, it has been determined that capsaicin has the ability to inhibit tumor growth and induce apoptosis in cancer cells. (in vitro and in vivo). Researchers have also looked at the results of combining capsaicin with chemotherapy medications in traditional treatment. The efficacy and bioavailability of capsaicin as a viable medicinal drug are being studied, along with ways to improve its clinical value. The present investigation carefully assesses the challenges and potential options for maximizing the therapeutic benefits of capsaicin, including customized drug delivery and personalized therapeutic strategies. In finalization, this comprehensive investigation brings together the evidence currently obtainable on the anticancer properties of capsaicin, underscoring its potential as an autonomous treatment option in the struggle against cancer. Capsaicin is a compound of significant relevance for continuing research and clinical exploration in the field of cancer treatment due to its diverse mechanisms of action and ability for boosting prevailing therapy approaches.


Capsaicin , Neoplasms , Capsaicin/pharmacology , Capsaicin/therapeutic use , Humans , Neoplasms/drug therapy , Neoplasms/prevention & control , Animals , Apoptosis/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Signal Transduction/drug effects , TRPV Cation Channels/metabolism
13.
ACS Appl Bio Mater ; 7(5): 2836-2850, 2024 05 20.
Article En | MEDLINE | ID: mdl-38717017

High-altitude regions, cold deserts, permafrost regions, and the polar region have some of the severest cold conditions on earth and pose immense perils of cold injuries to exposed individuals. Accidental and unintended exposures to severe cold, either unintentionally or due to occupational risks, can greatly increase the risk of serious conditions including hypothermia, trench foot, and cold injuries like frostbite. Cold-induced vasoconstriction and intracellular/intravascular ice crystal formation lead to hypoxic conditions at the cellular level. The condition is exacerbated in individuals having inadequate and proper covering and layering, particularly when large area of the body are exposed to extremely cold environments. There is a paucity of preventive and therapeutic pharmacological modalities that have been explored for managing and treating cold injuries. Given this, an efficient modality that can potentiate the healing of frostbite was investigated by studying various complex pathophysiological changes that occur during severe cold injuries. In the current research, we report the effectiveness and healing properties of a standardized formulation, i.e., a herbosomal-loaded PEG-poloxamer topical formulation (n-HPTF), on frostbite. The intricate mechanistic pathways modulated by the novel formulation have been elucidated by studying the pathophysiological sequelae that occur following severe cold exposures leading to frostbite. The results indicate that n-HPTF ameliorates the outcome of frostbite, as it activates positive sensory nerves widely distributed in the epidermis transient receptor potential vanilloid 1 (TRPV1), significantly (p < 0.05) upregulates cytokeratin-14, promotes angiogenesis (VEGF-A), prominently represses the expression of thromboxane formation (TXA2), and significantly (p < 0.05) restores levels of enzymatic (glutathione reductase, superoxide dismutase, and catalase) and nonenzymatic antioxidants (glutathione). Additionally, n-HPTF attenuates oxidative stress and the expression of inflammatory proteins PGF-2α, NFκB-p65, TNF-α, IL-6, IL-1ß, malondialdehyde (MDA), advanced oxidative protein products (AOPP), and protein carbonylation (PCO). Masson's Trichrome staining showed that n-HPTF stimulates cellular proliferation, and increases collagen fiber deposition, which significantly (p < 0.05) promotes the healing of frostbitten tissue, as compared to control. We conclude that protection against severe cold injuries by n-HPTF is mediated via modulation of pathways involving TRPV1, VEGF-A, TXA2, redox homeostasis, and inflammatory cascades. The study is likely to have widespread implications for the prophylaxis and management of moderate-to-severe frostbite conditions.


Homeostasis , Poloxamer , Polyethylene Glycols , TRPV Cation Channels , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor A/metabolism , TRPV Cation Channels/metabolism , Animals , Poloxamer/chemistry , Poloxamer/pharmacology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Homeostasis/drug effects , Oxidation-Reduction , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Rats , Materials Testing , Cold Injury/metabolism , Cold Injury/drug therapy , Particle Size , Inflammation/drug therapy , Inflammation/metabolism , Male , Liposomes/chemistry , Humans , Administration, Topical , Frostbite/metabolism , Frostbite/drug therapy
14.
Exp Cell Res ; 439(1): 114087, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38735619

Diabetic retinopathy (DR) is a common microvascular complication that causes visual impairment or loss. Aquaporin 4 (AQP4) is a regulatory protein involved in water transport and metabolism. In previous studies, we found that AQP4 is related to hypoxia injury in Muller cells. Transient receptor potential cation channel subfamily V member 4 (TRPV4) is a non-selective cation channel protein involved in the regulation of a variety of ophthalmic diseases. However, the effects of AQP4 and TRPV4 on ferroptosis and oxidative stress in high glucose (HG)-treated Muller cells are unclear. In this study, we investigated the functions of AQP4 and TRPV4 in DR. HG was used to treat mouse Muller cells. Reverse transcription quantitative polymerase chain reaction was used to measure AQP4 mRNA expression. Western blotting was used to detect the protein levels of AQP4, PTGS2, GPX4, and TRPV4. Cell count kit-8, flow cytometry, 5,5',6,6'-tetrachloro-1,1,3,3'-tetraethylbenzimidazolyl carbocyanine iodide staining, and glutathione (GSH), superoxide dismutase (SOD), and malondialdehyde (MDA) kits were used to evaluate the function of the Muller cells. Streptozotocin was used to induce DR in rats. Haematoxylin and eosin staining was performed to stain the retina of rats. GSH, SOD, and MDA detection kits, immunofluorescence, and flow cytometry assays were performed to study the function of AQP4 and TRPV4 in DR rats. Results found that AQP4 and TRPV4 were overexpressed in HG-induced Muller cells and streptozotocin-induced DR rats. AQP4 inhibition promoted proliferation and cell cycle progression, repressed cell apoptosis, ferroptosis, and oxidative stress, and alleviated retinal injury in DR rats. Mechanistically, AQP4 positively regulated TRPV4 expression. Overexpression of TRPV4 enhanced ferroptosis and oxidative stress in HG-treated Muller cells, and inhibition of TRPV4 had a protective effect on DR-induced retinal injury in rats. In conclusion, inhibition of AQP4 inhibits the ferroptosis and oxidative stress in Muller cells by downregulating TRPV4, which may be a potential target for DR therapy.


Aquaporin 4 , Diabetic Retinopathy , Ependymoglial Cells , Ferroptosis , Oxidative Stress , TRPV Cation Channels , Animals , Male , Mice , Rats , Aquaporin 4/metabolism , Aquaporin 4/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Diabetic Retinopathy/genetics , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Glucose/metabolism , Glucose/pharmacology , Mice, Inbred C57BL , Rats, Sprague-Dawley , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics
15.
Eur J Pharmacol ; 975: 176648, 2024 Jul 15.
Article En | MEDLINE | ID: mdl-38759706

Opioids are used for pain relief in patients suffering from acute myocardial ischemia or infarction. Clinical and laboratory studies demonstrate that morphine treated patients or the experimental animal model suffering acute myocardial ischemia and reperfusion, may worsen myocardial viability. As transient receptor potential vanilloid 1 (TRPV1) plays important roles in pain sensation and cardio-protection, we query whether opioids may exacerbate myocardial viability via interaction with TRPV1 activity in the pain relief. We found the co-expressions of TRPV1 and opioid µ, δ and κ receptors in adult rat cardiomyocytes. Intravenous injection of morphine (0.3 mg/kg) at 20 min after induction of myocardial ischemia, in the rat model of acute myocardial ischemia and reperfusion, induced significant reduction of phosphorylated TRPV1 (p-TRPV1) in the ventricular myocardium and increase in serum cardiac troponin I (cTnI), compared with the ischemia/reperfusion controls (all P < 0.05). The effects of morphine were completely reversed by selective opioid µ, δ and κ receptor antagonists. While significant upregulation of p-TRPV1 (P < 0.05) and improvement of ±dP/dt max (all P < 0.05) were detected in the animals giving the same dose of morphine before induction of myocardial ischemia. The changes in p-TRPV1 correlate with the alterations of cTnI (r = -0.5840, P = 0.0283) and ±dP/dt max (r = 0.8084, P = 0.0005 and r = -0.8133, P = 0.0004, respectively). The findings of this study may indicate that potentiation and attenuation of TRPV1 sensitivity correlate with the improvement of the cardiac performance and the aggravation of myocardial viability, respectively, by giving morphine before and during myocardial ischemia and reperfusion.


Morphine , Myocardial Reperfusion Injury , Rats, Sprague-Dawley , TRPV Cation Channels , Animals , TRPV Cation Channels/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/drug therapy , Morphine/pharmacology , Phosphorylation/drug effects , Male , Rats , Time Factors , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Analgesics, Opioid/pharmacology , Receptors, Opioid/metabolism , Troponin I/metabolism , Troponin I/blood , Myocardium/metabolism , Myocardium/pathology
16.
Sci Transl Med ; 16(748): eadk1358, 2024 May 22.
Article En | MEDLINE | ID: mdl-38776392

Blood-CNS barrier disruption is a hallmark of numerous neurological disorders, yet whether barrier breakdown is sufficient to trigger neurodegenerative disease remains unresolved. Therapeutic strategies to mitigate barrier hyperpermeability are also limited. Dominant missense mutations of the cation channel transient receptor potential vanilloid 4 (TRPV4) cause forms of hereditary motor neuron disease. To gain insights into the cellular basis of these disorders, we generated knock-in mouse models of TRPV4 channelopathy by introducing two disease-causing mutations (R269C and R232C) into the endogenous mouse Trpv4 gene. TRPV4 mutant mice exhibited weakness, early lethality, and regional motor neuron loss. Genetic deletion of the mutant Trpv4 allele from endothelial cells (but not neurons, glia, or muscle) rescued these phenotypes. Symptomatic mutant mice exhibited focal disruptions of blood-spinal cord barrier (BSCB) integrity, associated with a gain of function of mutant TRPV4 channel activity in neural vascular endothelial cells (NVECs) and alterations of NVEC tight junction structure. Systemic administration of a TRPV4-specific antagonist abrogated channel-mediated BSCB impairments and provided a marked phenotypic rescue of symptomatic mutant mice. Together, our findings show that mutant TRPV4 channels can drive motor neuron degeneration in a non-cell autonomous manner by precipitating focal breakdown of the BSCB. Further, these data highlight the reversibility of TRPV4-mediated BSCB impairments and identify a potential therapeutic strategy for patients with TRPV4 mutations.


Blood-Brain Barrier , Endothelial Cells , Gain of Function Mutation , Motor Neurons , TRPV Cation Channels , Animals , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Motor Neurons/pathology , Motor Neurons/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Mice , Nerve Degeneration/pathology , Nerve Degeneration/genetics , Phenotype , Spinal Cord/pathology , Spinal Cord/metabolism
17.
Cell Calcium ; 121: 102894, 2024 Jul.
Article En | MEDLINE | ID: mdl-38728789

TRPV2 voltage-insensitive, calcium-permeable ion channels play important roles in cancer progression, immune response, and neuronal development. Despite TRPV2's physiological impact, underlying endogenous proteins mediating TRPV2 responses and affected signaling pathways remain elusive. Using quantitative peroxidase-catalyzed (APEX2) proximity proteomics we uncover dynamic changes in the TRPV2-proximal proteome and identify calcium signaling and cell adhesion factors recruited to the molecular channel neighborhood in response to activation. Quantitative TRPV2 proximity proteomics further revealed activation-induced enrichment of protein clusters with biological functions in neural and cellular projection. We demonstrate a functional connection between TRPV2 and the neural immunoglobulin cell adhesion molecules NCAM and L1CAM. NCAM and L1CAM stimulation robustly induces TRPV2 [Ca2+]I flux in neuronal PC12 cells and this TRPV2-specific [Ca2+]I flux requires activation of the protein kinase PKCα. TRPV2 expression directly impacts neurite lengths that are modulated by NCAM or L1CAM stimulation. Hence, TRPV2's calcium signaling plays a previously undescribed, yet vital role in cell adhesion, and TRPV2 calcium flux and neurite development are intricately linked via NCAM and L1CAM cell adhesion proteins.


Calcium , Neural Cell Adhesion Molecule L1 , Neural Cell Adhesion Molecules , Neuronal Outgrowth , Proteome , TRPV Cation Channels , TRPV Cation Channels/metabolism , Animals , Neural Cell Adhesion Molecules/metabolism , Rats , PC12 Cells , Neural Cell Adhesion Molecule L1/metabolism , Proteome/metabolism , Calcium/metabolism , Calcium Signaling , Cell Adhesion , Humans , Neurites/metabolism , Protein Kinase C-alpha/metabolism
18.
Environ Pollut ; 353: 124127, 2024 Jul 15.
Article En | MEDLINE | ID: mdl-38759746

Allergic asthma is a chronic inflammatory airway disease with a high mortality rate and a rapidly increasing prevalence in recent decades that is closely linked to environmental change. Previous research found that high humidity (HH) and the traffic-related air pollutant NO2 both aggregated allergic asthma. Their combined effect and mechanisms on asthma exacerbation, however, are unknown. Our study aims to toxicologically clarify the role of HH (90%) and NO2 (5 ppm) on allergic asthma. Ninety male Balb/c mice were randomly assigned to one of six groups (n = 15 in each): saline control, ovalbumin (OVA)-sensitized, OVA + HH, OVA + NO2, OVA + HH + NO2, and OVA + HH + NO2+Capsazepine (CZP). After 38 days of treatment, the airway function, pathological changes in lung tissue, blood inflammatory cells, and oxidative stress and inflammatory biomarkers were comprehensively assessed. Co-exposure to HH and NO2 exacerbated histopathological changes and airway hyperresponsiveness, increased IgE, oxidative stress markers malonaldehyde (MDA) and allergic asthma-related inflammation markers (IL-1ß, TNF-α and IL-17), and upregulated the expressions of the transient receptor potential (TRP) ion channels (TRPA1, TRPV1 and TRPV4). Our findings show that co-exposure to HH and NO2 disrupted the Th1/Th2 immune balance, promoting allergic airway inflammation and asthma susceptibility, and increasing TRPV1 expression, whereas CZP reduced TRPV1 expression and alleviated allergic asthma symptoms. Thus, therapeutic treatments that target the TRPV1 ion channel have the potential to effectively manage allergic asthma.


Air Pollutants , Asthma , Humidity , Lung , Mice, Inbred BALB C , Nitrogen Dioxide , Oxidative Stress , TRPV Cation Channels , Animals , Asthma/metabolism , Oxidative Stress/drug effects , Male , Mice , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Lung/drug effects , Lung/immunology , Lung/metabolism , Air Pollutants/toxicity , Nitrogen Dioxide/toxicity , Inflammation/metabolism , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/genetics , Immunoglobulin E , Capsaicin/analogs & derivatives
19.
Biochem Biophys Res Commun ; 721: 150128, 2024 Aug 20.
Article En | MEDLINE | ID: mdl-38776831

PURPOSE: Chronic stress is a significant risk factor for mood disorders such as depression, where synaptic plasticity plays a central role in pathogenesis. Transient Receptor Potential Vanilloid Type-2 (TRPV2) Ion Channels are implicated in hypothalamic-pituitary-adrenal axis disorders. Previous proteomic analysis indicated a reduction in TRPV2 levels in the chronic unpredictable mild stress (CUMS) rat model, yet its role in synaptic plasticity during depression remains to be elucidated. This study aims to investigate TRPV2's role in depression and its underlying mechanisms. METHODS: In vivo and in vitro experiments were conducted using the TRPV2-specific agonist probenecid and ERK1/2 inhibitors SCH772984. In vivo, rats underwent six weeks of CUMS before probenecid administration. Depressive-like behaviors were assessed through behavioral tests. ELISA kits measured 5-HT, DA, NE levels in rat hippocampal tissues. Hippocampal morphology was examined via Nissl staining. In vitro, rat hippocampal neuron cell lines were treated with ERK1/2 inhibitors SCH772984 and probenecid. Western blot, immunofluorescence, immunohistochemical staining, and RT-qPCR assessed TRPV2 expression, neurogenesis-related proteins, synaptic markers, and ERK1/2-CREB-BDNF signaling proteins. RESULTS: Decreased hippocampal TRPV2 levels were observed in CUMS rats. Probenecid treatment mitigated depressive-like behavior and enhanced hippocampal 5-HT, NE, and DA levels in CUMS rats. TRPV2 activation countered CUMS-induced synaptic plasticity inhibition. Probenecid activated the ERK1/2-CREB-BDNF pathway, suggesting TRPV2's involvement in this pathway via ERK1/2. CONCLUSION: These findings indicate that TRPV2 activation offers protective effects against depressive-like behaviors and enhances hippocampal synaptic plasticity in CUMS rats via the ERK1/2-CREB-BDNF pathway. TRPV2 emerges as a potential therapeutic target for depression.


Brain-Derived Neurotrophic Factor , Cyclic AMP Response Element-Binding Protein , Hippocampus , MAP Kinase Signaling System , Neuronal Plasticity , Rats, Sprague-Dawley , Stress, Psychological , TRPV Cation Channels , Animals , TRPV Cation Channels/metabolism , TRPV Cation Channels/antagonists & inhibitors , Neuronal Plasticity/drug effects , Male , Brain-Derived Neurotrophic Factor/metabolism , Stress, Psychological/complications , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Hippocampus/metabolism , Hippocampus/drug effects , Rats , MAP Kinase Signaling System/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Depression/metabolism , Depression/drug therapy , Chronic Disease , Probenecid/pharmacology
20.
Front Immunol ; 15: 1386719, 2024.
Article En | MEDLINE | ID: mdl-38694510

Introduction: B-cell activation triggers the release of endoplasmic reticulum calcium stores through the store-operated calcium entry (SOCE) pathway resulting in calcium influx by calcium release-activated calcium (CRAC) channels on the plasma membrane. B-cell-specific murine knockouts of SOCE do not impact humoral immunity suggesting that alternative channels may be important. Methods: We identified a member of the calcium-permeable transient receptor potential (TRP) ion channel family, TRPV5, as a candidate channel expressed in B cells by a quantitative polymerase chain reaction (qPCR) screen. To further investigate the role of TRPV5 in B-cell responses, we generated a murine TRPV5 knockout (KO) by CRISPR-Cas9. Results: We found TRPV5 polarized to B-cell receptor (BCR) clusters upon stimulation in a PI3K-RhoA-dependent manner. TRPV5 KO mice have normal B-cell development and mature B-cell numbers. Surprisingly, calcium influx upon BCR stimulation in primary TRPV5 KO B cells was not impaired; however, differential expression of other calcium-regulating proteins, such as ORAI1, may contribute to a compensatory mechanism for calcium signaling in these cells. We demonstrate that TRPV5 KO B cells have impaired spreading and contraction in response to membrane-bound antigen. Consistent with this, TRPV5 KO B cells have reduced BCR signaling measured through phospho-tyrosine residues. Lastly, we also found that TRPV5 is important for early T-dependent antigen specific responses post-immunization. Discussion: Thus, our findings identify a role for TRPV5 in BCR signaling and B-cell activation.


B-Lymphocytes , Calcium Signaling , Lymphocyte Activation , Mice, Knockout , Receptors, Antigen, B-Cell , TRPV Cation Channels , Animals , Mice , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Calcium/metabolism , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, B-Cell/immunology , Signal Transduction , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
...