Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 201
Filter
1.
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
2.
Bull Exp Biol Med ; 177(2): 231-234, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39093477

ABSTRACT

Acute nociceptive pain in mice caused by subcutaneous (intraplantar) injection of TRPV1 ion channel agonist capsaicin (1.6 µg/mouse) and the effects of protein kinase A inhibitor H-89 (0.05 mg/mouse, intraplantar injection) and NMDA receptor channel antagonists MK-801 (7.5 and 15 µg/mouse, topical application) and hemantane (0.5 mg/mouse, topical application) on the pain were assessed. MK-801 and hemantane were found to reduce the duration of the pain response. H-89 did not significantly affect the pain in animals, but preliminary administration of this drug abolished the antinociceptive effect of MK-801 (7.5 µg/mouse) and weakens the effect of hemantane (0.5 mg/mouse).


Subject(s)
Analgesics , Capsaicin , Dizocilpine Maleate , Receptors, N-Methyl-D-Aspartate , Animals , Capsaicin/pharmacology , Mice , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Male , Dizocilpine Maleate/pharmacology , Analgesics/pharmacology , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Nociceptive Pain/drug therapy , Nociceptive Pain/chemically induced , Pain Measurement/drug effects , Pain Measurement/methods
3.
J Gen Physiol ; 156(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39162763

ABSTRACT

Regulation of ion channel expression on the plasma membrane is a major determinant of neuronal excitability, and identifying the underlying mechanisms of this expression is critical to our understanding of neurons. Here, we present two orthogonal strategies to label extracellular sites of the ion channel TRPV1 that minimally perturb its function. We use the amber codon suppression technique to introduce a non-canonical amino acid (ncAA) with tetrazine click chemistry, compatible with a trans-cyclooctene coupled fluorescent dye. Additionally, by inserting the circularly permutated HaloTag (cpHaloTag) in an extracellular loop of TRPV1, we can incorporate a fluorescent dye of our choosing. Optimization of ncAA insertion sites was accomplished by screening residue positions between the S1 and S2 transmembrane domains with elevated missense variants in the human population. We identified T468 as a rapid labeling site (∼5 min) based on functional and biochemical assays in HEK293T/17 cells. Through adapting linker lengths and backbone placement of cpHaloTag on the extracellular side of TRPV1, we generated a fully functional channel construct, TRPV1exCellHalo, with intact wild-type gating properties. We used TRPV1exCellHalo in a single molecule experiment to track TRPV1 on the cell surface and validate studies that show decreased mobility of the channel upon activation. The application of these extracellular label TRPV1 (exCellTRPV1) constructs to track surface localization of the channel will shed significant light on the mechanisms regulating its expression and provide a general scheme to introduce similar modifications to other cell surface receptors.


Subject(s)
Cell Membrane , Fluorescent Dyes , TRPV Cation Channels , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Humans , HEK293 Cells , Cell Membrane/metabolism , Animals
4.
J Mol Neurosci ; 74(3): 79, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39162890

ABSTRACT

Diabetic neuropathic pain (DNP) is a diabetic complication that causes severe pain and deeply impacts the quality of the sufferer's daily life. Currently, contemporary clinical treatments for DNP generally exhibit a deficiency in effectiveness. Electroacupuncture (EA) is recognized as a highly effective and safe treatment for DNP with few side effects. Regrettably, the processes via which EA alleviates DNP are still poorly characterized. Transient receptor potential vanilloid 1 (TRPV1) and phosphorylated calcium/calmodulin-dependent protein kinase II (p-CaMKII) are overexpressed on spinal cord dorsal horn (SCDH) in DNP rats, and co-localization is observed between them. Capsazepine, a TRPV1 antagonist, effectively reduced nociceptive hypersensitivity and downregulated the overexpression of phosphorylated CaMKIIα in rats with DNP. Conversely, the CaMKII inhibitor KN-93 did not have any impact on TRPV1. EA alleviated heightened sensitivity to pain caused by nociceptive stimuli and downregulated the level of TRPV1, p-CaMKIIα, and phosphorylated cyclic adenosine monophosphate response element-binding protein (p-CREB) in DNP rats. Intrathecal injection of capsaicin, on the other hand, reversed the above effects of EA. These findings indicated that the CaMKII/CREB pathway on SCDH is located downstream of TRPV1 and is affected by TRPV1. EA alleviates DNP through the TRPV1-mediated CaMKII/CREB pathway.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cyclic AMP Response Element-Binding Protein , Diabetic Neuropathies , Electroacupuncture , Rats, Sprague-Dawley , TRPV Cation Channels , Animals , TRPV Cation Channels/metabolism , TRPV Cation Channels/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Electroacupuncture/methods , Rats , Male , Cyclic AMP Response Element-Binding Protein/metabolism , Diabetic Neuropathies/therapy , Diabetic Neuropathies/metabolism , Capsaicin/pharmacology , Capsaicin/analogs & derivatives , Signal Transduction , Spinal Cord Dorsal Horn/metabolism , Benzenesulfonamides , Benzylamines
5.
Elife ; 122024 Aug 20.
Article in English | MEDLINE | ID: mdl-39162616

ABSTRACT

Ligands such as insulin, epidermal growth factor, platelet-derived growth factor, and nerve growth factor (NGF) initiate signals at the cell membrane by binding to receptor tyrosine kinases (RTKs). Along with G-protein-coupled receptors, RTKs are the main platforms for transducing extracellular signals into intracellular signals. Studying RTK signaling has been a challenge, however, due to the multiple signaling pathways to which RTKs typically are coupled, including MAP/ERK, PLCγ, and Class 1A phosphoinositide 3-kinases (PI3K). The multi-pronged RTK signaling has been a barrier to isolating the effects of any one downstream pathway. Here, we used optogenetic activation of PI3K to decouple its activation from other RTK signaling pathways. In this context, we used genetic code expansion to introduce a click chemistry noncanonical amino acid into the extracellular side of membrane proteins. Applying a cell-impermeant click chemistry fluorophore allowed us to visualize delivery of membrane proteins to the plasma membrane in real time. Using these approaches, we demonstrate that activation of PI3K, without activating other pathways downstream of RTK signaling, is sufficient to traffic the TRPV1 ion channels and insulin receptors to the plasma membrane.


Subject(s)
Click Chemistry , Phosphatidylinositol 3-Kinases , Protein Transport , Receptor Protein-Tyrosine Kinases , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Signal Transduction , Cell Membrane/metabolism , Optogenetics , Genetic Code , Light , Animals , HEK293 Cells
6.
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
7.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39125864

ABSTRACT

The potential role of the transient receptor potential Vanilloid 1 (TRPV1) non-selective cation channel in gastric carcinogenesis remains unclear. The main objective of this study was to evaluate TRPV1 expression in gastric cancer (GC) and precursor lesions compared with controls. Patient inclusion was based on a retrospective review of pathology records. Patients were subdivided into five groups: Helicobacter pylori (H. pylori)-associated gastritis with gastric intestinal metaplasia (GIM) (n = 12), chronic atrophic gastritis (CAG) with GIM (n = 13), H. pylori-associated gastritis without GIM (n = 19), GC (n = 6) and controls (n = 5). TRPV1 expression was determined with immunohistochemistry and was significantly higher in patients with H. pylori-associated gastritis compared with controls (p = 0.002). TRPV1 expression was even higher in the presence of GIM compared with patients without GIM and controls (p < 0.001). There was a complete loss of TRPV1 expression in patients with GC. TRPV1 expression seems to contribute to gastric-mucosal inflammation and precursors of GC, which significantly increases in cancer precursor lesions but is completely lost in GC. These findings suggest TRPV1 expression to be a potential marker for precancerous conditions and a target for individualized treatment. Longitudinal studies are necessary to further address the role of TRPV1 in gastric carcinogenesis.


Subject(s)
Helicobacter Infections , Stomach Neoplasms , TRPV Cation Channels , Humans , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Male , Female , Middle Aged , Aged , Helicobacter Infections/metabolism , Helicobacter Infections/complications , Helicobacter Infections/pathology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Retrospective Studies , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Helicobacter pylori/pathogenicity , Metaplasia/metabolism , Metaplasia/pathology , Gastritis/metabolism , Gastritis/pathology , Gastritis/microbiology , Adult , Immunohistochemistry , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gastritis, Atrophic/metabolism , Gastritis, Atrophic/pathology
8.
Article in English | MEDLINE | ID: mdl-39136188

ABSTRACT

Recently, physical tools for remotely stimulating mechanical force-sensitive and temperature-sensitive proteins to regulate intracellular pathways have opened up novel and exciting avenues for basic research and clinical applications. Among the numerous modes of physical stimulation, magnetic stimulation is significantly attractive for biological applications due to the advantages of depth penetration and spatial-temporally controlled transduction. Herein, the physicochemical parameters (e.g., shape, size, composition) that influence the magnetic properties of magnetic nanosystems as well as the characteristics of transient receptor potential vanilloid-1 (TRPV1) and transient receptor potential vanilloid-4 (TRPV4) channels are systematically summarized, which offer opportunities for magnetic manipulation of cell fate in a precise and effective manner. In addition, representative regulatory applications involving magnetic nanosystem-based TRPV1 and TRPV4 channel activation are highlighted, both at the cellular level and in animal models. Furthermore, perspectives on the further development of this magnetic stimulation mode are commented on, with emphasis on scientific limitations and possible directions for exploitation. This article is categorized under: Diagnostic Tools > Biosensing Diagnostic Tools > In Vivo Nanodiagnostics and Imaging.


Subject(s)
TRPV Cation Channels , TRPV Cation Channels/metabolism , Animals , Humans , Mice
9.
PLoS One ; 19(8): e0304874, 2024.
Article in English | MEDLINE | ID: mdl-39106272

ABSTRACT

INTRODUCTION: The olfactory and trigeminal system are closely interlinked. Existing literature has primarily focused on characterizing trigeminal stimulation through mechanical and chemical stimulation, neglecting thermal stimulation thus far. The present study aimed to characterize the intranasal sensitivity to heat and the expression of trigeminal receptors (transient receptor potential channels, TRP). METHODS: A total of 20 healthy participants (aged 21-27 years, 11 women) were screened for olfactory function and trigeminal sensitivity using several tests. Under endoscopic control, a thermal stimulator was placed in 7 intranasal locations: anterior septum, lateral vestibulum, interior nose tip, lower turbinate, middle septum, middle turbinate, and olfactory cleft to determine the thermal threshold. Nasal swabs were obtained in 3 different locations (anterior septum, middle turbinate, olfactory cleft) to analyze the expression of trigeminal receptors TRP: TRPV1, TRPV3, TRPA1, TRPM8. RESULTS: The thermal threshold differed between locations (p = 0.018), with a trend for a higher threshold at the anterior septum (p = 0.092). There were no differences in quantitative receptor expression (p = 0.46) at the different sites. The highest overall receptor RNA expression was detected for TRPV1 over all sites (p<0.001). The expression of TRPV3 was highest at the anterior septum compared to the middle turbinate or the olfactory cleft. The thermal sensitivity correlated with olfactory sensitivity and results from tests were related to trigeminal function like intensity ratings of ammonium, a questionnaire regarding trigeminal function, nasal patency, and CO2 thresholds. However, no correlation was found between receptor expression and psychophysical measures of trigeminal function. DISCUSSION: This study provided the first insights about intranasal thermal sensitivity and suggested the presence of topographical differences in thermal thresholds. There was no correlation between thermal sensitivity and trigeminal mRNA receptor expression. However, thermal sensitivity was found to be associated with psychophysical measures of trigeminal and olfactory function.


Subject(s)
Nasal Mucosa , TRPV Cation Channels , Humans , Female , Adult , Male , Nasal Mucosa/metabolism , Young Adult , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Hot Temperature , Trigeminal Nerve/physiology , Trigeminal Nerve/metabolism , Sensory Thresholds/physiology , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/genetics , TRPM Cation Channels/metabolism , TRPM Cation Channels/genetics , Thermosensing/physiology , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/genetics
10.
Sci Transl Med ; 16(759): eadn2140, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39110778

ABSTRACT

Hearing loss is a major health concern in our society, affecting more than 400 million people worldwide. Among the causes, aminoglycoside therapy can result in permanent hearing loss in 40% to 60% of patients receiving treatment, and despite these high numbers, no drug for preventing or treating this type of hearing loss has yet been approved by the US Food and Drug Administration. We have previously conducted high-throughput screenings of bioactive compounds, using zebrafish as our discovery platform, and identified piplartine as a potential therapeutic molecule. In the present study, we expanded this work and characterized piplartine's physicochemical and therapeutic properties. We showed that piplartine had a wide therapeutic window and neither induced nephrotoxicity in vivo in zebrafish nor interfered with aminoglycoside antibacterial activity. In addition, a fluorescence-based assay demonstrated that piplartine did not inhibit cytochrome C activity in microsomes. Coadministration of piplartine protected from kanamycin-induced hair cell loss in zebrafish and protected hearing function, outer hair cells, and presynaptic ribbons in a mouse model of kanamycin ototoxicity. Last, we investigated piplartine's mechanism of action by phospho-omics, immunoblotting, immunohistochemistry, and molecular dynamics experiments. We found an up-regulation of AKT1 signaling in the cochleas of mice cotreated with piplartine. Piplartine treatment normalized kanamycin-induced up-regulation of TRPV1 expression and modulated the gating properties of this receptor. Because aminoglycoside entrance to the inner ear is, in part, mediated by TRPV1, these results suggested that by regulating TRPV1 expression, piplartine blocked aminoglycoside's entrance, thereby preventing the long-term deleterious effects of aminoglycoside accumulation in the inner ear compartment.


Subject(s)
Aminoglycosides , Hearing Loss , TRPV Cation Channels , Zebrafish , Animals , TRPV Cation Channels/metabolism , Aminoglycosides/pharmacology , Hearing Loss/chemically induced , Hearing Loss/metabolism , Hearing Loss/prevention & control , Hearing Loss/pathology , Mice , Ototoxicity/metabolism , Kanamycin , Dioxolanes/pharmacology , Piperidones
11.
Int J Mol Sci ; 25(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39126095

ABSTRACT

Olfactory perception is an important physiological function for human well-being and health. Loss of olfaction, or anosmia, caused by viral infections such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has received considerable attention, especially in persistent cases that take a long time to recover. This review discusses the integration of different components of the olfactory epithelium to serve as a structural and functional unit and explores how they are affected during viral infections, leading to the development of olfactory dysfunction. The review mainly focused on the role of receptors mediating the disruption of olfactory signal transduction pathways such as angiotensin converting enzyme 2 (ACE2), transmembrane protease serine type 2 (TMPRSS2), neuropilin 1 (NRP1), basigin (CD147), olfactory, transient receptor potential vanilloid 1 (TRPV1), purinergic, and interferon gamma receptors. Furthermore, the compromised function of the epithelial sodium channel (ENaC) induced by SARS-CoV-2 infection and its contribution to olfactory dysfunction are also discussed. Collectively, this review provides fundamental information about the many types of receptors that may modulate olfaction and participate in olfactory dysfunction. It will help to understand the underlying pathophysiology of virus-induced anosmia, which may help in finding and designing effective therapies targeting molecules involved in viral invasion and olfaction. To the best of our knowledge, this is the only review that covered all the receptors potentially involved in, or mediating, the disruption of olfactory signal transduction pathways during COVID-19 infection. This wide and complex spectrum of receptors that mediates the pathophysiology of olfactory dysfunction reflects the many ways in which anosmia can be therapeutically managed.


Subject(s)
Anosmia , COVID-19 , SARS-CoV-2 , Humans , COVID-19/metabolism , COVID-19/complications , COVID-19/physiopathology , COVID-19/virology , Anosmia/physiopathology , Anosmia/etiology , Anosmia/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Olfactory Mucosa/metabolism , Olfactory Mucosa/virology , Signal Transduction , Serine Endopeptidases/metabolism , Neuropilin-1/metabolism , Basigin/metabolism , TRPV Cation Channels/metabolism
12.
Nutrients ; 16(15)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39125403

ABSTRACT

Osteoporosis in menopausal women requires alternatives to current medications, considering their adverse effects. In this context, probiotics and isoflavone products are promising dietary interventions. The objective of our study was to examine the impacts of Lactobacillus acidophilus and its combination with daidzein and tempeh on calcium status, calcium transporters, and bone metabolism biomarkers in a post-menopausal osteoporotic rat model. A total of 48 female Wistar rats were exposed to a two-stage experiment involving calcium deficit induction and subsequent dietary interventions across six groups. Calcium levels, the gene expression of TRPV5 and TRPV6 calcium transporters, bone histopathology, serum bone metabolism markers, and blood biochemistry were evaluated. The results revealed that, while decreasing serum calcium levels, the groups that received the probiotic L. acidophilus and isoflavone combination exhibited increased bone metabolism biomarkers and decreased calcium transporter expressions, akin to the effects of bisphosphonate. Additionally, significant improvements in bone histopathology were observed in these groups. However, the group receiving probiotic L. acidophilus alone did not exhibit significant changes in bone resorption biomarkers, calcium transporter expression, or various blood parameters. Meanwhile, the combination of probiotic L. acidophilus with tempeh positively influenced hematological parameters and reduced cholesterol and triglyceride levels, but it led to elevated blood glucose levels. Correlation analyses highlighted associations between serum calcium levels, calcium transporter expression, and bone metabolism biomarkers. In conclusion, our findings suggest that the daily consumption of probiotic L. acidophilus in combination with isoflavone products may improve bone health in ovariectomized rats, warranting further research to elucidate potential interactions with other nutrients.


Subject(s)
Biomarkers , Bone and Bones , Calcium , Disease Models, Animal , Isoflavones , Lactobacillus acidophilus , Probiotics , Rats, Wistar , Animals , Female , Isoflavones/pharmacology , Probiotics/pharmacology , Calcium/blood , Biomarkers/blood , Rats , Bone and Bones/metabolism , Bone and Bones/drug effects , TRPV Cation Channels/metabolism , Osteoporosis, Postmenopausal , Postmenopause
13.
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
14.
Nat Commun ; 15(1): 6689, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107321

ABSTRACT

Transient Receptor Potential Vanilloid 1 (TRPV1) plays a central role in pain sensation and is thus an attractive pharmacological drug target. SAF312 is a potent, selective, and non-competitive antagonist of TRPV1 and shows promising potential in treating ocular surface pain. However, the precise mechanism by which SAF312 inhibits TRPV1 remains poorly understood. Here, we present the cryo-EM structure of human TRPV1 in complex with SAF312, elucidating the structural foundation of its antagonistic effects on TRPV1. SAF312 binds to the vanilloid binding pocket, preventing conformational changes in S4 and S5 helices, which are essential for channel gating. Unexpectedly, a putative cholesterol was found to contribute to SAF312's inhibition. Complemented by mutagenesis experiments and molecular dynamics simulations, our research offers substantial mechanistic insights into the regulation of TRPV1 by SAF312, highlighting the interplay between the antagonist and cholesterol in modulating TRPV1 function. This work not only expands our understanding of TRPV1 inhibition by SAF312 but also lays the groundwork for further developments in the design and optimization of TRPV1-related therapies.


Subject(s)
Cholesterol , Cryoelectron Microscopy , Molecular Dynamics Simulation , TRPV Cation Channels , TRPV Cation Channels/metabolism , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/chemistry , TRPV Cation Channels/genetics , Cholesterol/metabolism , Humans , Binding Sites , HEK293 Cells , Protein Binding
15.
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
17.
Cell Physiol Biochem ; 58(4): 292-310, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38973197

ABSTRACT

BACKGROUND/AIMS: Tactile perception relies on mechanoreceptors and nerve fibers, including c-fibers, Aß-fibers and Aδ-fibers. Schwann cells (SCs) play a crucial role in supporting nerve fibers, with non-myelinating SCs enwrapping c-fibers and myelinating SCs ensheathing Aß and Aδ fibers. Recent research has unveiled new functions for cutaneous sensory SCs, highlighting the involvement of nociceptive SCs in pain perception and Meissner corpuscle SCs in tactile sensation. Furthermore, Piezo2, previously associated with Merkel cell tactile sensitivity, has been identified in SCs. The goal of this study was to investigate the channels implicated in SC mechanosensitivity and the release process of neurotrophic factor secretion. METHODS: Immortalized IFRS1 SCs and human primary SCs generated two distinct subtypes of SCs: undifferentiated and differentiated SCs. Quantitative PCR was employed to evaluate the expression of differentiation markers and mechanosensitive channels, including TRP channels (TRPV4, TRPM7 and TRPA1) and Piezo channels (Piezo1 and Piezo2). To validate the functionality of specific mechanosensitive channels, Ca2+ imaging and electronic cell sizing experiments were conducted under hypotonic conditions, and inhibitors and siRNAs were used. Protein expression was assessed by Western blotting and immunostaining. Additionally, secretome analysis was performed to evaluate the release of neurotrophic factors in response to hypotonic stimulation, with BDNF, a representative trophic factor, quantified using ELISA. RESULTS: Induction of differentiation increased Piezo2 mRNA expression levels both in IFRS1 and in human primary SCs. Both cell types were responsive to hypotonic solutions, with differentiated SCs displaying a more pronounced response. Gd3+ and FM1-43 effectively inhibited hypotonicity-induced Ca2+ transients in differentiated SCs, implicating Piezo2 channels. Conversely, inhibitors of Piezo1 and TRPM7 (Dooku1 and NS8593, respectively) had no discernible impact. Moreover, Piezo2 in differentiated SCs appeared to participate in regulatory volume decreases (RVD) after cell swelling induced by hypotonic stimulation. A Piezo2 deficiency correlated with reduced RVD and prolonged cell swelling, leading to heightened release of the neurotrophic factor BDNF by upregulating the function of endogenously expressed Ca2+-permeable TRPV4. CONCLUSION: Our study unveils the mechanosensitivity of SCs and implicates Piezo2 channels in the release of neurotrophic factors from SCs. These results suggest that Piezo2 may contribute to RVD, thereby maintaining cellular homeostasis, and may also serve as a negative regulator of neurotrophic factor release. These findings underscore the need for further investigation into the role of Piezo2 in SC function and neurotrophic regulation.


Subject(s)
Brain-Derived Neurotrophic Factor , Cell Size , Ion Channels , Schwann Cells , Schwann Cells/metabolism , Schwann Cells/cytology , Humans , Ion Channels/metabolism , Cell Size/drug effects , Brain-Derived Neurotrophic Factor/metabolism , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , RNA, Small Interfering/metabolism , Cell Differentiation , Cells, Cultured , RNA Interference , Calcium/metabolism , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/genetics , Mechanotransduction, Cellular
18.
CNS Neurosci Ther ; 30(7): e14829, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38961264

ABSTRACT

AIMS: Paclitaxel (PTX) is extensively utilized in the management of diverse solid tumors, frequently resulting in paclitaxel-induced peripheral neuropathy (PIPN). The present study aimed to investigate sex differences in the behavioral manifestations and underlying pathogenesis of PIPN and search for clinically efficacious interventions. METHODS: Male and female C57BL/6 mice (5-6 weeks and 12 months, weighing 18-30 g) were intraperitoneally (i.p.) administered paclitaxel diluted in saline (NaCl 0.9%) at a dose of 2 mg/kg every other day for a total of 4 injections. Von Frey and hot plate tests were performed before and after administration to confirm the successful establishment of the PIPN model and also to evaluate the pain of PIPN and the analgesic effect of PD-L1. On day 14 after PTX administration, PD-L1 protein (10 ng/pc) was injected into the PIPN via the intrathecal (i.t.) route. To knock down TRPV1 in the spinal cord, adeno-associated virus 9 (AAV9)-Trpv1-RNAi (5 µL, 1 × 1013 vg/mL) was slowly injected via the i.t. route. Four weeks after AAV9 delivery, the downregulation of TRPV1 expression was verified by immunofluorescence staining and Western blotting. The levels of PD-L1, TRPV1 and CGRP were measured via Western blotting, RT-PCR, and immunofluorescence staining. The levels of TNF-α and IL-1ß were measured via RT-PCR. RESULTS: TRPV1 and CGRP protein and mRNA levels were higher in the spinal cords of control female mice than in those of control male mice. PTX-induced nociceptive behaviors in female PIPN mice were greater than those in male PIPN mice, as indicated by increased expression of TRPV1 and CGRP. The analgesic effects of PD-L1 on mechanical hyperalgesia and thermal sensitivity were significantly greater in female mice than in male mice, with calculated relative therapeutic levels increasing by approximately 2.717-fold and 2.303-fold, respectively. PD-L1 and CGRP were partly co-localized with TRPV1 in the dorsal horn of the mouse spinal cord. The analgesic effect of PD-L1 in PIPN mice was observed to be mediated through the downregulation of TRPV1 and CGRP expression following AAV9-mediated spinal cord specific decreased TRPV1 expression. CONCLUSIONS: PTX-induced nociceptive behaviors and the analgesic effect of PD-L1 in PIPN mice were sexually dimorphic, highlighting the significance of incorporating sex as a crucial biological factor in forthcoming mechanistic studies of PIPN and providing insights for potential sex-specific therapeutic approaches.


Subject(s)
B7-H1 Antigen , Calcitonin Gene-Related Peptide , Mice, Inbred C57BL , Paclitaxel , Peripheral Nervous System Diseases , Sex Characteristics , TRPV Cation Channels , Animals , Paclitaxel/toxicity , Male , Female , Mice , Calcitonin Gene-Related Peptide/metabolism , TRPV Cation Channels/metabolism , TRPV Cation Channels/antagonists & inhibitors , B7-H1 Antigen/metabolism , Peripheral Nervous System Diseases/chemically induced , Antineoplastic Agents, Phytogenic/toxicity , Spinal Cord/drug effects , Spinal Cord/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Hyperalgesia/metabolism
19.
J Cell Mol Med ; 28(13): e18509, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38957035

ABSTRACT

Pruritus is often accompanied with bacterial infections, but the underlying mechanism is not fully understood. Although previous studies revealed that lipopolysaccharides (LPS) could directly activate TRPV4 channel and TRPV4 is involved in the generation of both acute itch and chronic itch, whether and how LPS affects TRPV4-mediated itch sensation remains unclear. Here, we showed that LPS-mediated TRPV4 sensitization exacerbated GSK101-induced scratching behaviour in mice. Moreover, this effect was compromised in TLR4-knockout mice, suggesting LPS acted through a TLR4-dependent mechanism. Mechanistically, LPS enhanced GSK101-evoked calcium influx in mouse ear skin cells and HEK293T cells transfected with TRPV4. Further, LPS sensitized TRPV4 channel through the intracellular TLR4-PI3K-AKT signalling. In summary, our study found a modulatory role of LPS in TRPV4 function and highlighted the TLR4-TRPV4 interaction in itch signal amplification.


Subject(s)
Lipopolysaccharides , Phosphatidylinositol 3-Kinases , Pruritus , Signal Transduction , TRPV Cation Channels , Toll-Like Receptor 4 , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Animals , Toll-Like Receptor 4/metabolism , Pruritus/metabolism , Pruritus/chemically induced , Pruritus/pathology , Lipopolysaccharides/pharmacology , Humans , Mice , HEK293 Cells , Phosphatidylinositol 3-Kinases/metabolism , Mice, Knockout , Mice, Inbred C57BL , Male , Calcium/metabolism , Proto-Oncogene Proteins c-akt/metabolism
20.
Cell Mol Life Sci ; 81(1): 285, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38969941

ABSTRACT

Volume regulation is essential for cell homeostasis and physiological function. Amongst the sensory molecules that have been associated with volume regulation is the transient receptor potential vanilloid 4 (TRPV4), which is a non-selective cation channel that in conjunction with aquaporins, typically controls regulatory volume decrease (RVD). Here we show that the interaction between orthologous AQP4 (Aqp4a) and TRPV4 (Trpv4) is important for regulatory volume increase (RVI) in post-activated marine fish spermatozoa under high osmotic stress. Based upon electrophysiological, volumetric, and in vivo and ex vivo functional experiments using the pharmacological and immunological inhibition of Aqp4a and Trpv4 our model suggests that upon ejaculation and exposure to the hypertonic seawater, spermatozoon shrinkage is initially mediated by water efflux through Aqp1aa in the flagellar tail. The shrinkage results in an increase in intracellular Ca2+ concentration, and the activation of sperm motility and a Na+/K+/2Cl- (NKCC1) cotransporter. The activity of NKCC1 is required for the initiation of cell swelling, which secondarily activates the Aqp4a-Trpv4 complex to facilitate the influx of water via Aqp4a-M43 and Ca2+ via Trpv4 and L-type channels for the mediation of RVI. The inhibitory experiments show that blocking of each of these events prevents either shrinkage or RVI. Our data thus reveal that post-activated marine fish spermatozoa are capable of initiating RVI under a high hypertonic stress, which is essential for the maintenance of sperm motility.


Subject(s)
Cell Size , Osmotic Pressure , Sperm Motility , Spermatozoa , TRPV Cation Channels , Animals , Male , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Spermatozoa/metabolism , Sperm Motility/physiology , Aquaporin 4/metabolism , Aquaporin 4/genetics , Calcium/metabolism , Fishes/metabolism , Fishes/physiology , Swimming , Solute Carrier Family 12, Member 2/metabolism , Solute Carrier Family 12, Member 2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL