Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.473
Filter
1.
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
2.
J Orthop Surg Res ; 19(1): 466, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39118176

ABSTRACT

OBJECTIVE: Delayed fracture healing increases the suffering of patients. An in-depth investigation of the pathogenesis of delayed fracture healing may offer new direction for the prevention and treatment. METHODS: The study included 63 normal healing tibial fractures and 58 delayed healing tibial fractures patients. Long non-coding RNA (lncRNA)TRPM2-AS, microRNA-545-3p (miR-545-3p), bone morphogenetic protein 2 (Bmp2) mRNA and osteogenic differentiation markers, including runt-related transcription factor 2 (Runx2), osteocalcin (Ocn), and alkaline phosphatase (Alp) mRNA expression were determined by Real-time quantitative reverse transcription-polymerase chain reaction in serum and MC3T3-E1 cells. The prediction potential of TRPM2-AS in delayed healing fracture patients was verified by receiver operating characteristic curves. The binding relationship of TRPM2-AS/miR-545-3p/Bmp2 was evaluated by dual luciferase reporter gene assay. Cell proliferation and apoptosis were detected by CCK-8 and flow cytometry. RESULTS: TRPM2-AS was remarkably down-regulated in patients with delayed fracture healing and could better predict the fracture healing status. TRPM2-AS downregulation inhibited osteogenic markers mRNA expression, restrained proliferation, and promoted apoptosis of MC3T3-E1 cells (p < 0.05). In delayed fracture healing, miR-545-3p was dramatically up-regulated and was negatively regulated by TRPM2-AS. Reducing miR-545-3p eliminate the negative effect of TRPM2-AS down-regulation on osteoblast proliferation and differentiation (p < 0.05). miR-545-3p targets Bmp2, which plays a positive role in osteoblast differentiation (p < 0.05). CONCLUSION: This study found that TRPM2-AS has the potential to be a diagnostic marker for delayed fracture healing and revealed that the TRPM2-AS/miR-545-3p/Bmp2 axis affects fracture healing by regulating osteoblast.


Subject(s)
Bone Morphogenetic Protein 2 , Fracture Healing , MicroRNAs , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Humans , MicroRNAs/genetics , Fracture Healing/genetics , Fracture Healing/physiology , Mice , Animals , RNA, Long Noncoding/genetics , Female , Male , Tibial Fractures/genetics , Osteogenesis/genetics , Osteogenesis/physiology , TRPM Cation Channels/genetics , Cell Proliferation/genetics , Cell Differentiation/genetics , Adult , Apoptosis/genetics , Middle Aged , Osteoblasts/metabolism
3.
Biotechnol J ; 19(8): e2400310, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39212193

ABSTRACT

The differentiation of bone marrow mesenchymal stem cells (BMSCs) toward osteogenesis can be induced by low-intensity pulsed ultrasound (LIPUS). However, the molecular mechanisms responsible for LIPUS stimulation are unclear. The possible molecular mechanisms by which LIPUS promotes osteogenic differentiation of BMSCs were investigated in this study. The quantification of alkaline phosphatase (ALP) activity, Alizarin Red S staining, ALP staining, and the establishment of a calvarial defect model were used to evaluate osteogenic effects. Immunofluorescence was performed to observe the expression of microfilaments and transient receptor potential melastatin 7 (TRPM7). The levels of F-actin/G-actin and osteogenesis-related proteins under LIPUS alone or LIPUS combined with cytoskeleton interfering drugs (Cytochalasin D [CytoD] or Jasplakinolide [JA]) were assayed by western blot. Quantitative real-time reverse transcription polymerase chain reaction was utilized to measure the expression of Trpm7 mRNA. Moreover, adenoviral Trpm7 knockdown was verified using western blot. The results demonstrated that LIPUS promoted bone formation in vivo. Under osteogenic induction in vitro, the osteogenesis of BMSCs induced by LIPUS was accompanied by the depolymerization and rearrangement of microfilaments and increased levels of TRPM7. By perturbing intracellular actin dynamics, CytoD enhanced the pro-osteogenicity of LIPUS and increased TRPM7 level, while JA inhibited the pro-osteogenicity of LIPUS and reduced TRPM7 level. Additionally, the knockdown of Trpm7 suppressed the osteogenic promotion of BMSCs induced by LIPUS. The transient depolymerization and rearrangement of the cytoskeleton microfilaments mediated by LIPUS can affect TRPM7 expression and subsequently promote the osteogenesis of BMSCs. This study provides further direction for exploring the molecular mechanism of LIPUS, as a mechanical stress, in facilitating the osteogenic differentiation of BMSCs.


Subject(s)
Actin Cytoskeleton , Actins , Cell Differentiation , Mesenchymal Stem Cells , Osteogenesis , TRPM Cation Channels , Animals , TRPM Cation Channels/metabolism , TRPM Cation Channels/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Actin Cytoskeleton/metabolism , Actins/metabolism , Actins/genetics , Rats , Rats, Sprague-Dawley , Ultrasonic Waves , Cells, Cultured , Male , Depsipeptides
4.
Channels (Austin) ; 18(1): 2396339, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39212541

ABSTRACT

The transient receptor potential melastatin 7 channel (TRPM7) is a nonselective cation channel highly expressed in some human cancer tissues. TRPM7 is involved in the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) of cancer cells. Modulation of TRPM7 could be a promising therapeutic strategy for treating cancer; however, efficient and selective pharmacological TRPM7 modulators are lacking. In this study we investigated N- [4- (4, 6-dimethyl- 2-pyrimidinyloxy) - 3- methylphenyl] -N' - [2 -(dimethylamino)] benzoylurea (SUD), a newly synthesized benzoylurea derivative, for its effects on cancer cell migration and EMT and on functional expression of TRPM7. Our previous studies showed that SUD induces cell cycle arrest and apoptosis of MCF-7 and BGC-823 cells (human breast cancer and gastric cancer cell lines, respectively). Here, we show that SUD significantly decreased the migration of both types of cancer cells. Moreover, SUD decreased vimentin expression and increased E-cadherin expression in both cell types, indicating that EMT is also decreased by SUD. Importantly, SUD potentially reduced the TRPM7-like current in a concentration-dependent manner and decreased TRPM7 expression through the PI3K/Akt signaling pathway. Finally, molecular docking simulations were used to investigate potential SUD binding sites on TRPM7. In summary, our research demonstrated that SUD is an effective TRPM7 inhibitor and a potential agent to suppress the metastasis of breast and gastric cancer by inhibiting TRPM7 expression and function.


Subject(s)
Cell Movement , Epithelial-Mesenchymal Transition , Protein Serine-Threonine Kinases , TRPM Cation Channels , Urea , Humans , TRPM Cation Channels/metabolism , TRPM Cation Channels/genetics , TRPM Cation Channels/antagonists & inhibitors , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Urea/analogs & derivatives , Urea/pharmacology , Urea/chemistry , Cell Line, Tumor , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Molecular Docking Simulation , MCF-7 Cells
5.
Sci Rep ; 14(1): 18447, 2024 08 08.
Article in English | MEDLINE | ID: mdl-39117690

ABSTRACT

Disorders of gallbladder motility can lead to serious pathology. Bitter tastants acting upon bitter taste receptors (TAS2R family) have been proposed as a novel class of smooth muscle relaxants to combat excessive contraction in the airways and other organs. To explore whether this might also emerge as an option for gallbladder diseases, we here tested bitter tastants for relaxant properties and profiled Tas2r expression in the mouse gallbladder. In organ bath experiments, the bitter tastants denatonium, quinine, dextromethorphan, and noscapine, dose-dependently relaxed the pre-contracted gallbladder. Utilizing gene-deficient mouse strains, neither transient receptor potential family member 5 (TRPM5), nor the Tas2r143/Tas2r135/Tas2r126 gene cluster, nor tuft cells proved to be required for this relaxation, indicating direct action upon smooth muscle cells (SMC). Accordingly, denatonium, quinine and dextromethorphan increased intracellular calcium concentration preferentially in isolated gallbladder SMC and, again, this effect was independent of TRPM5. RT-PCR revealed transcripts of Tas2r108, Tas2r126, Tas2r135, Tas2r137, and Tas2r143, and analysis of gallbladders from mice lacking tuft cells revealed preferential expression of Tas2r108 and Tas2r137 in tuft cells. A TAS2R143-mCherry reporter mouse labeled tuft cells in the gallbladder epithelium. An in silico analysis of a scRNA sequencing data set revealed Tas2r expression in only few cells of different identity, and from in situ hybridization histochemistry, which did not label distinct cells. Our findings demonstrate profound tuft cell- and TRPM5-independent relaxing effects of bitter tastants on gallbladder smooth muscle, but do not support the concept that these effects are mediated by bitter receptors.


Subject(s)
Gallbladder , Muscle, Smooth , Receptors, G-Protein-Coupled , TRPM Cation Channels , Animals , Mice , Calcium/metabolism , Dextromethorphan/pharmacology , Gallbladder/metabolism , Mice, Inbred C57BL , Mice, Knockout , Muscle Relaxation/drug effects , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Noscapine/pharmacology , Quaternary Ammonium Compounds/pharmacology , Quinine/pharmacology , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Taste/physiology , TRPM Cation Channels/metabolism , TRPM Cation Channels/genetics , Tuft Cells/metabolism
6.
Cell Mol Biol Lett ; 29(1): 93, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956502

ABSTRACT

BACKGROUND: Anti-angiogenic therapy has become one of the effective treatment methods for tumors. Long noncoding RNAs (lncRNAs) are emerging as important regulators of tumorigenesis and angiogenesis in EC. However, the underlying mechanisms of lncRNA TRPM2-AS in EC are still not clear. METHODS: We screened the differently expressed lncRNAs that were highly associated with poor prognosis and angiogenesis of EC by bioinformatics analysis, and constructed a ceRNA network based on the prognostic lncRNAs. The subcellular localization of TRPM2-AS was determined by fluorescence in situ hybridization (FISH) and nuclear cytoplasmic fractionation assay. CCK-8, EdU, transwell, western blot, qRT-PCR and endothelial tube formation assay were used to evaluate the effects of TRPM2-AS on the proliferation, invasion, migration of EC cells and angiogenesis. The targeted microRNA (miRNA) of TRPM2-AS was predicted by bioinformatic methods. The interaction between TRPM2-AS and miR497-5p, miR497-5p and SPP1 were analyzed by RNA immunoprecipitation and dual-luciferase reporter assay. A subcutaneous tumor model was used to explore TRPM2-AS's function in vivo. CIBERSORT was used to analyze the correlation between TRPM2-AS and immune cell immersion in EC. RESULTS: We found that the expression of TRPM2-AS and SPP1 was aberrantly upregulated, while miR-497-5p expression was significantly downregulated in EC tissues and cells. TRPM2-AS was closely correlated with the angiogenesis and poor prognosis in EC patients. Mechanistically, TRPM2-AS could sponge miR-497-5p to release SPP1, thus promoting the proliferation, invasion and migration of EC cells and angiogenesis of HUVECs. Knockdown of TRPM2-AS in xenograft mouse model inhibited tumor proliferation and angiogenesis in vivo. In addition, TRPM2-AS plays a vital role in regulating the tumor immune microenvironment of EC, overexpression of TRPM2-AS in EC cells stimulated the polarization of M2 macrophages and angiogenesis through secreting SPP1 enriched exosomes. CONCLUSION: The depletion of TRPM2-AS inhibits the oncogenicity of EC by targeting the miR-497-5p/SPP1 axis. This study offers a better understanding of TRPM2-AS's role in regulating angiogenesis and provides a novel target for EC treatment.


Subject(s)
Cell Movement , Cell Proliferation , Endometrial Neoplasms , Gene Expression Regulation, Neoplastic , MicroRNAs , Neovascularization, Pathologic , RNA, Long Noncoding , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Neovascularization, Pathologic/genetics , Female , Animals , Cell Proliferation/genetics , Cell Line, Tumor , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrial Neoplasms/metabolism , Cell Movement/genetics , Mice , Disease Progression , Mice, Nude , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Mice, Inbred BALB C , Prognosis , Angiogenesis
7.
Nat Commun ; 15(1): 5887, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003305

ABSTRACT

Memory engrams are a subset of learning activated neurons critical for memory recall, consolidation, extinction and separation. While the transcriptional profile of engrams after learning suggests profound neural changes underlying plasticity and memory formation, little is known about how memory engrams are selected and allocated. As epigenetic factors suppress memory formation, we developed a CRISPR screening in the hippocampus to search for factors controlling engram formation. We identified histone lysine-specific demethylase 4a (Kdm4a) as a negative regulator for engram formation. Kdm4a is downregulated after neural activation and controls the volume of mossy fiber boutons. Mechanistically, Kdm4a anchors to the exonic region of Trpm7 gene loci, causing the stalling of nascent RNAs and allowing burst transcription of Trpm7 upon the dismissal of Kdm4a. Furthermore, the YTH domain containing protein 2 (Ythdc2) recruits Kdm4a to the Trpm7 gene and stabilizes nascent RNAs. Reducing the expression of Kdm4a in the hippocampus via genetic manipulation or artificial neural activation facilitated the ability of pattern separation in rodents. Our work indicates that Kdm4a is a negative regulator of engram formation and suggests a priming state to generate a separate memory.


Subject(s)
Hippocampus , Memory , TRPM Cation Channels , Animals , Hippocampus/metabolism , Mice , Memory/physiology , TRPM Cation Channels/metabolism , TRPM Cation Channels/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Humans , Down-Regulation/genetics , Neurons/metabolism , Male , Mice, Inbred C57BL , Rats , CRISPR-Cas Systems , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Neuronal Plasticity/genetics , HEK293 Cells , Histone Demethylases
8.
Dev Growth Differ ; 66(6): 349-356, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38970357

ABSTRACT

Parkinson's disease is a neurological disorder characterized by reduced motility, depression and dementia. Guamanian parkinsonism dementia with amyotrophic sclerosis is a local case of Parkinson's disease reported in the Western Pacific Islands of Guam and Rota as well as in the Kii Peninsula of Japan. A previous genetic study has suggested that Guamanian parkinsonism is attributable to a variant of the TRPM7 gene, which encodes for melastatin-related transient receptor potential (TRP) ion channels. But the link between parkinsonism and the TRPM7 gene remains elusive. Previous studies have addressed that trpm7-deficient zebrafish embryos showed defects in pigmentation and touch-evoked motor response. In this study, we identified a new viable allele of trpm7 mutant causing an I756N amino acid substitution in the first transmembrane domain. Behavioral analyses revealed that trpm7 mutants showed compromised motility with their movement distance shorter than wild-type larvae. The velocity of the movement was significantly reduced in trpm7 mutants than in wild-type larvae. Along with a previous finding of reduced dopaminergic neurons in zebrafish trpm7 mutants, reduced motility of trpm7 mutants can suggest another similarity between trpm7 phenotypes and Parkinson's disease symptoms.


Subject(s)
TRPM Cation Channels , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Mutation , Protein Serine-Threonine Kinases
9.
Physiol Genomics ; 56(9): 634-647, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39037434

ABSTRACT

Although age-dependent alterations in urinary magnesium (Mg2+) excretion have been described, the underlying mechanism remains elusive. As heritability significantly contributes to variations in urinary Mg2+ excretion, we measured urinary Mg2+ excretion at different ages in a cohort of genetically variable Diversity Outbred (DO) mice. Compared with animals aged 6 mo, an increase in Mg2+ excretion was observed at 12 and 18 mo. Quantitative trait locus (QTL) analysis revealed an association of a locus on chromosome 10 with Mg2+ excretion at 6 mo of age, with Oit3 (encoding oncoprotein-induced transcript 3; OIT3) as our primary candidate gene. To study the possible role of OIT3 in renal Mg2+ handling, we generated and characterized Oit3 knockout (Oit3-/-) mice. Although a slightly lower serum Mg2+ concentration was present in male Oit3-/- mice, this effect was not observed in female Oit3-/- mice. In addition, urinary Mg2+ excretion and the expression of renal magnesiotropic genes were unaltered in Oit3-/- mice. For animals aged 12 and 18 mo, QTL analysis revealed an association with a locus on chromosome 19, which contains the gene encoding TRPM6, a known Mg2+ channel involved in renal Mg2+ reabsorption. Comparison with RNA sequencing (RNA-Seq) data revealed that Trpm6 mRNA expression is inversely correlated with the QTL effect, implying that TRPM6 may be involved in age-dependent changes in urinary Mg2+ excretion in mice. In conclusion, we show here that variants in Oit3 and Trpm6 are associated with urinary Mg2+ excretion at distinct periods of life, although OIT3 is unlikely to affect renal Mg2+ handling.NEW & NOTEWORTHY Aging increased urinary magnesium (Mg2+) excretion in mice. We show here that variation in Oit3, a candidate gene for the locus associated with Mg2+ excretion in young mice, is unlikely to be involved as knockout of Oit3 did not affect Mg2+ excretion. Differences in the expression of the renal Mg2+ channel TRPM6 may contribute to the variation in urinary Mg2+ excretion in older mice.


Subject(s)
Aging , Magnesium , Mice, Knockout , Quantitative Trait Loci , TRPM Cation Channels , Animals , Magnesium/urine , Magnesium/metabolism , Magnesium/blood , Quantitative Trait Loci/genetics , Male , Female , Mice , Aging/genetics , TRPM Cation Channels/genetics , Kidney/metabolism
10.
Physiol Res ; 73(3): 351-367, 2024 07 17.
Article in English | MEDLINE | ID: mdl-39027953

ABSTRACT

Diabetic cardiomyopathy may result from the overproduction of ROS, TRPM2 and TRPV2. Moreover, the therapeutic role of ginger, omega-3 fatty acids, and their combinations on the expression of TRPM2 and TRPV2 and their relationship with apoptosis, inflammation, and oxidative damage in heart tissue of rats with type 2 diabetes have not yet been determined. Therefore, this study aimed to investigate the therapeutic effects of ginger and omega-3 fatty acids on diabetic cardiomyopathy by evaluating the cardiac gene expression of TRPM2 and TRPV2, oxidative damage, inflammation, and apoptosis in male rats. Ninety adult male Wistar rats were equally divided into nine control, diabetes, and treated diabetes groups. Ginger extract (100 mg/kg) and omega-3 fatty acids (50, 100, and 150 mg/kg) were orally administrated in diabetic rats for 6 weeks. Type 2 diabetes was induced by feeding a high-fat diet and a single dose of STZ (40 mg/kg). Glucose, cardiac troponin I (cTnI), lipid profile, insulin in serum, and TNF-alpha IL-6, SOD, MDA, and CAT in the left ventricle of the heart were measured. The cardiac expression of TRPM2, TRPV2, NF-kappaB, Bcl2, Bax, Cas-3, and Nrf-2 genes was also measured in the left ventricle of the heart. An electrocardiogram (ECG) was continuously recorded to monitor arrhythmia at the end of the course. The serum levels of cTnI, glucose, insulin, and lipid profile, and the cardiac levels of MDA, IL-6, and TNF-alpha increased in the diabetic group compared to the control group (p<0.05). Moreover, the cardiac levels of SOD and CAT decreased in the diabetic group compared to the control group (p<0.05). The cardiac expression of TRPM2, TRPV2, NF-kappaB, Bax, and Cas-3 increased and Bcl2 and Nrf-2 expression decreased in the diabetic group compared to the control group (p<0.05). However, simultaneous and separate treatment with ginger extract and omega-3 fatty acids (50, 100, and 150 mg/kg) could significantly moderate these changes (p<0.05). The results also showed that the simultaneous treatment of ginger extract and different doses of omega-3 fatty acids have improved therapeutic effects than their individual treatments (p<0.05). It can be concluded that ginger and omega-3 fatty acids showed protective effects against diabetic cardiomyopathy by inhibiting inflammation, apoptosis and oxidative damage of the heart and reducing blood glucose and cardiac expression of TRPM2 and TRPV2. Combining ginger and omega-3 in the diet may provide a natural approach to reducing the risk or progression of diabetic cardiomyopathy while preserving heart structure and function.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Fatty Acids, Omega-3 , Plant Extracts , Rats, Wistar , Zingiber officinale , Animals , Male , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Plant Extracts/administration & dosage , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/therapeutic use , Zingiber officinale/chemistry , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/prevention & control , Rats , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Dietary Supplements , Oxidative Stress/drug effects , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/complications , TRPM Cation Channels/metabolism , TRPM Cation Channels/genetics
11.
Int J Biol Macromol ; 275(Pt 1): 133659, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38969045

ABSTRACT

The age-related alterations in pituitary function, including changes in prolactin (PRL) production contributes to the systemic susceptibility to age-related diseases. Our previous research has shown the involvement of Nrg1 in regulating the expression and secretion of PRL. However, the precise role of Nrg1 in mitigating the senescence of pituitary lactotrophs and the underlying mechanisms are yet to be comprehended. Here, data from the GEPIA database was used to evaluate the association between transient receptor potential cation channel subfamily M member 8 (TRPM8) and PRL in normal human pituitary tissues, followed by immunofluorescence verification using a human pituitary tissue microarray. TRPM8 levels showed a significant positive association with PRL expression in normal human pituitary tissues, and both TRPM8 and PRL levels declined during aging, suggesting that TRPM8 may regulate pituitary aging by affecting PRL production. It was also found that treatment with exogenous neuregulin 1 (Nrg1) markedly delayed the senescence of GH3 cells (rat lactotroph cell line) generated by D-galactose (D-gal). In addition, melatonin reduced the levels of senescence-related markers in senescent pituitary cells by promoting Nrg1 / ErbB4 signaling, stimulating PRL expression and secretion. Further investigation showed that Nrg1 attenuated senescence in pituitary cells by increasing TRPM8 expression. Downregulation of TRPM8 activation eliminated Nrg1-mediated amelioration of pituitary cell senescence. These findings demonstrate the critical function of Nrg1 / ErbB signaling in delaying pituitary lactotroph cell senescence and enhancing PRL production via promoting TRPM8 expression under the modulation of melatonin.


Subject(s)
Cellular Senescence , Lactotrophs , Melatonin , Neuregulin-1 , Prolactin , Signal Transduction , TRPM Cation Channels , Prolactin/metabolism , TRPM Cation Channels/metabolism , TRPM Cation Channels/genetics , Humans , Cellular Senescence/drug effects , Signal Transduction/drug effects , Lactotrophs/metabolism , Lactotrophs/drug effects , Melatonin/pharmacology , Animals , Rats , Neuregulin-1/metabolism , Neuregulin-1/genetics , Pituitary Gland/metabolism , Pituitary Gland/drug effects , Aging/metabolism , Receptor, ErbB-4/metabolism , Receptor, ErbB-4/genetics , Cell Line , Male , Female
12.
Poult Sci ; 103(9): 103984, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38986357

ABSTRACT

Improving immune function is an important indicator for establishing cold adaptation in broilers. In the study, to explore the effects and molecular mechanisms of intermittent and mild cold stimulation (IMCS) on the immune function of broilers, CIRP and TRPM8, induced by cold stimulation, as well as the NF-κB and MAPK pathways which play an important role in immune response, were selected to investigate. A total of 192 one-day-old broilers (Ross 308) were selected and randomly divided into the control group (CC) and the cold stimulation group (CS). The broilers in CC were raised at normal feeding temperature from d 1 to 43, while the broilers in CS were subjected to cold stimulation from day 15 to 35, with a temperature 3 °C below that of the CC group for 5 h, at 1 d intervals. The results showed that IMCS had little effect on the broiler hearts, and the myocardial structure was not damaged. On d 22, IMCS significantly increased the mRNA levels of CIRP, TRPM8, P65, P38, COX-2, TNF-α, IFN- γ, IL-6, IL-10, and the protein levels of CIRP, P65, P38, IL-1ß and iNOS in the hearts, and the levels of CIRP and all cytokines in the serum (P ≤ 0.05). The mRNA and protein levels of IκB-α were significantly reduced (P ≤ 0.05). On d 36, the mRNA levels of TRPM8, P65, ERK, and IL-10 in the hearts and the content of COX-2 in the serum in CS were increased significantly (P ≤ 0.05), while the mRNA levels of IκB-α, P38, and IL-1ß were decreased significantly (P ≤ 0.05). On d 43, IMCS significantly upregulated the mRNA levels of TRPM8, IFN- γ, IL-4, IL-6, IL-10, and the protein levels of IκB-α, P38, and the levels of iNOS, TNF-α, IL6 and IL10 in the serum (P ≤ 0.05); whereas it significantly downregulated CIRP, JNK, P38, iNOS, TNF-α mRNA levels, and CIRP, P65, ERK, JNK, IL1ß and iNOS protein levels (P ≤ 0.05). Therefore, IMCS can enhance broiler immune function through co-regulation of CIRP and TRPM8 on the NF-κB and MAPK pathways, which facilitate the cold adaptation in broilers.


Subject(s)
Avian Proteins , Chickens , Cold Temperature , MAP Kinase Signaling System , NF-kappa B , TRPM Cation Channels , Animals , Chickens/immunology , Chickens/physiology , Avian Proteins/genetics , Avian Proteins/metabolism , NF-kappa B/metabolism , NF-kappa B/genetics , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Random Allocation , Male , Immunity, Innate , Signal Transduction
13.
Int J Biochem Cell Biol ; 174: 106618, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39053766

ABSTRACT

Ferroptosis is an emerging target in rheumatoid arthritis (RA). We previously reported that transient receptor potential melastatin 7 (TRPM7) expression is correlated with RA cartilage destruction and demonstrated that TRPM7 mediates ferroptosis in chondrocytes. Here, we further determined the role and mechanism of (R)-N-(Benzimidazol-2-yl)-1,2,3,4-tetrahydro-1-naphthylamine (NS8593), a TRPM7 inhibitor, in chondrocyte ferroptosis of RA. We established in vitro models of ferroptosis in human chondrocytes (C28/I2 cells) by using ferroptosis inducer Erastin. The results showed that NS8593 could protect C28/I2 cells from ferroptosis by inhibiting TRPM7 channel, which was manifested by restoring cell viability, reducing cytotoxicity, affecting the expression of ferroptosis marker protein, and restoring redox balance to alleviate Erastin-induced oxidative stress injury. Mechanistically, the Heme oxygenase-1 (HO-1) axis responded to Erastin stimulation, which resulted in TRPM7-mediated chondrocyte ferroptosis, NS8593 could reduce the expression of HO-1 by inhibiting TRPM7 channel. Moreover, NS8593 alleviated articular cartilage destruction and inhibited chondrocyte ferroptosis in AA rats. In conclusion, NS8593 mitigated articular cartilage damage and chondrocyte ferroptosis through the TRPM7/HO-1 pathway, suggesting that NS8593 may be a potential novel drug for the treatment of RA.


Subject(s)
1-Naphthylamine , Arthritis, Experimental , Chondrocytes , Ferroptosis , TRPM Cation Channels , Animals , Humans , Male , Rats , 1-Naphthylamine/analogs & derivatives , 1-Naphthylamine/pharmacology , Arthritis, Experimental/pathology , Arthritis, Experimental/metabolism , Arthritis, Experimental/drug therapy , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrocytes/pathology , Ferroptosis/drug effects , Heme Oxygenase (Decyclizing) , Heme Oxygenase-1/metabolism , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Signal Transduction/drug effects , TRPM Cation Channels/metabolism , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/genetics
14.
Elife ; 122024 Jul 02.
Article in English | MEDLINE | ID: mdl-38953285

ABSTRACT

We studied lysosomal Ca2+ in inflammasome. Lipopolysaccharide (LPS) + palmitic acid (PA) decreased lysosomal Ca2+ ([Ca2+]Lys) and increased [Ca2+]i through mitochondrial ROS, which was suppressed in Trpm2-KO macrophages. Inflammasome activation and metabolic inflammation in adipose tissue of high-fat diet (HFD)-fed mice were ameliorated by Trpm2 KO. ER→lysosome Ca2+ refilling occurred after lysosomal Ca2+ release whose blockade attenuated LPS + PA-induced inflammasome. Subsequently, store-operated Ca2+entry (SOCE) was activated whose inhibition suppressed inflammasome. SOCE was coupled with K+ efflux whose inhibition reduced ER Ca2+ content ([Ca2+]ER) and impaired [Ca2+]Lys recovery. LPS + PA activated KCa3.1 channel, a Ca2+-activated K+ channel. Inhibitors of KCa3.1 channel or Kcnn4 KO reduced [Ca2+]ER, attenuated increase of [Ca2+]i or inflammasome activation by LPS + PA, and ameliorated HFD-induced inflammasome or metabolic inflammation. Lysosomal Ca2+ release induced delayed JNK and ASC phosphorylation through CAMKII-ASK1. These results suggest a novel role of lysosomal Ca2+ release sustained by ER→lysosome Ca2+ refilling and K+ efflux through KCa3.1 channel in inflammasome activation and metabolic inflammation.


Subject(s)
Calcium , Endoplasmic Reticulum , Inflammasomes , Inflammation , Lysosomes , Mice, Knockout , Potassium , Animals , Inflammasomes/metabolism , Mice , Lysosomes/metabolism , Calcium/metabolism , Potassium/metabolism , Inflammation/metabolism , Endoplasmic Reticulum/metabolism , Lipopolysaccharides , TRPM Cation Channels/metabolism , TRPM Cation Channels/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Mice, Inbred C57BL , Macrophages/metabolism , Male , Diet, High-Fat
15.
Cancer Res Commun ; 4(8): 2101-2111, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39041239

ABSTRACT

Triple-negative breast cancer (TNBC) is clinically aggressive and relatively unresponsive to current therapies. Therefore, the development of new anticancer agents is needed to satisfy clinical needs. Oxyphenisatin acetate (Acetalax), which had been used as a laxative, has recently been reported to have anticancer activity in murine models. In this study, we demonstrate that Acetalax and its diphenolic laxative structural analogue bisacodyl (Dulcolax) exhibit potent antiproliferative activity in TNBC cell lines and cause oncosis, a nonapoptotic cell death characterized by cellular and nuclear swelling and cell membrane blebbing, leading to mitochondrial dysfunction, ATP depletion, and enhanced immune and inflammatory responses. Mechanistically, we provide evidence that transient receptor potential melastatin member 4 (TRPM4) is poisoned by Acetalax and bisacodyl in MDA-MB468, BT549, and HS578T TNBC cells. MDA-MB231 and MDA-MB436 TNBC cells without endogenous TRPM4 expression as well as TRPM4-knockout TNBC cells were found to be Acetalax- and bisacodyl-resistant. Conversely, ectopic expression of TRPM4 sensitized MDA-MB231 and MDA-MB436 cells to Acetalax. TRPM4 was also lost in cells with acquired Acetalax resistance. Moreover, TRPM4 is rapidly degraded by the ubiquitin-proteasome system upon acute exposure to Acetalax and bisacodyl. Together, these results demonstrate that TRPM4 is a previously unknown target of Acetalax and bisacodyl and that TRPM4 expression in cancer cells is a predictor of Acetalax and bisacodyl efficacy and could be used for the clinical development of these drugs as anticancer agents. SIGNIFICANCE: Acetalax and bisacodyl kill cancer cells by causing oncosis following poisoning of the plasma membrane sodium transporter TRPM4 and represent a new therapeutic approach for TNBC.


Subject(s)
TRPM Cation Channels , Triple Negative Breast Neoplasms , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/genetics , Humans , Cell Line, Tumor , Female , Cell Proliferation/drug effects , Animals , Mice , Antineoplastic Agents/pharmacology
16.
PeerJ ; 12: e17559, 2024.
Article in English | MEDLINE | ID: mdl-38854798

ABSTRACT

Background: To investigate the effects of arsenic trioxide (ATO) on human colorectal cancer cells (HCT116) growth and the role of transient receptor potential melastatin 4 (TRPM4) channel in this process. Methods: The viability of HCT116 cells was assessed using the CCK-8 assay. Western blot analysis was employed to examine the protein expression of TRPM4. The apoptosis of HCT116 cells was determined using TUNEL and Flow cytometry. Cell migration was assessed through the cell scratch recovery assay and Transwell cell migration assay. Additionally, Transwell cell invasion assay was performed to determine the invasion ability of HCT116 cells. Results: ATO suppressed the viability of HCT116 cells in a dose-dependent manner, accompanied by a decline in cell migration and invasion, and an increase in apoptosis. 9-phenanthroline (9-Ph), a specific inhibitor of TRPM4, abrogated the ATO-induced upregulation of TRPM4 expression. Additionally, blocking TRPM4 reversed the effects of ATO on HCT116 cells proliferation, including restoration of cell viability, migration and invasion, as well as the inhibition of apoptosis. Conclusion: ATO inhibits CRC cell growth by inducing TRPM4 expression, our findings indicate that ATO is a promising therapeutic strategy and TRPM4 may be a novel target for the treatment of CRC.


Subject(s)
Apoptosis , Arsenic Trioxide , Cell Movement , Cell Proliferation , Cell Survival , Colorectal Neoplasms , TRPM Cation Channels , Humans , TRPM Cation Channels/metabolism , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/genetics , Arsenic Trioxide/pharmacology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , HCT116 Cells , Cell Movement/drug effects , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Oxides/pharmacology , Antineoplastic Agents/pharmacology , Neoplasm Invasiveness , Arsenicals/pharmacology
17.
J Therm Biol ; 122: 103868, 2024 May.
Article in English | MEDLINE | ID: mdl-38852485

ABSTRACT

Transient Receptor Potential (TRP) ion channels are important for sensing environmental temperature. In rodents, TRPV4 senses warmth (25-34 °C), TRPV1 senses heat (>42 °C), TRPA1 putatively senses cold (<17 °C), and TRPM8 senses cool-cold (18-26 °C). We investigated if knockout (KO) mice lacking these TRP channels exhibited changes in thermal preference. Thermal preference was tested using a dual hot-cold plate with one thermoelectric surface set at 30 °C and the adjacent surface at a temperature of 15-45 °C in 5 °C increments. Blinded observers counted the number of times mice crossed through an opening between plates and the percentage of time spent on the 30 °C plate. In a separate experiment, observers blinded as to genotype also assessed the temperature at the location on a thermal gradient (1.83 m, 4-50 °C) occupied by the mouse at 5- or 10-min intervals over 2 h. Male and female wildtype mice preferred 30 °C and significantly avoided colder (15-20 °C) and hotter (40-45 °C) temperatures. Male TRPV1KOs and TRPA1KOs, and TRPV4KOs of both sexes, were similar, while female WTs, TRPV1KOs, TRPA1KOs and TRPM8KOs did not show significant thermal preferences across the temperature range. Male and female TRPM8KOs did not significantly avoid the coldest temperatures. Male mice (except for TRPM8KOs) exhibited significantly fewer plate crossings at hot and cold temperatures and more crossings at thermoneutral temperatures, while females exhibited a similar but non-significant trend. Occupancy temperatures along the thermal gradient exhibited a broad distribution that shrank somewhat over time. Mean occupancy temperatures (recorded at 90-120 min) were significantly higher for females (30-34 °C) compared to males (26-27 °C) of all genotypes, except for TRPA1KOs which exhibited no sex difference. The results indicate (1) sex differences with females (except TRPA1KOs) preferring warmer temperatures, (2) reduced thermosensitivity in female TRPV1KOs, and (3) reduced sensitivity to cold and innocuous warmth in male and female TRPM8KOs consistent with previous studies.


Subject(s)
Mice, Knockout , TRPA1 Cation Channel , TRPV Cation Channels , Thermosensing , Animals , Female , Male , Mice , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , TRPA1 Cation Channel/genetics , TRPA1 Cation Channel/metabolism , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/physiology , Mice, Inbred C57BL , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Hot Temperature , Cold Temperature
18.
Molecules ; 29(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38893478

ABSTRACT

Transient receptor potential melastatin-8 (TRPM8) is a cation channel that is activated by cold and "cooling agents" such as menthol and icilin, which induce a cold sensation. The stimulation of TRPM8 activates an intracellular signaling cascade that ultimately leads to a change in the gene expression pattern of the cells. Here, we investigate the TRPM8-induced signaling pathway that links TRPM8 channel activation to gene transcription. Using a pharmacological approach, we show that the inhibition of phosphatidylinositol 4-phosphate 5 kinase α (PIP5K), an enzyme essential for the biosynthesis of phosphatidylinositol 4,5-bisphosphate, attenuates TRPM8-induced gene transcription. Analyzing the link between TRPM8 and Gq proteins, we show that the pharmacological inhibition of the ßγ subunits impairs TRPM8 signaling. In addition, genetic studies show that TRPM8 requires an activated Gα subunit for signaling. In the nucleus, the TRPM8-induced signaling cascade triggers the activation of the transcription factor AP-1, a complex consisting of a dimer of basic region leucine zipper (bZIP) transcription factors. Here, we identify the bZIP protein c-Jun as an essential component of AP-1 within the TRPM8-induced signaling cascade. In summary, with PIP5K, Gq subunits, and c-Jun, we identified key molecules in TRPM8-induced signaling from the plasma membrane to the nucleus.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11 , Phosphotransferases (Alcohol Group Acceptor) , Signal Transduction , TRPM Cation Channels , TRPM Cation Channels/metabolism , TRPM Cation Channels/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Transcription Factor AP-1/metabolism , HEK293 Cells , Proto-Oncogene Proteins c-jun/metabolism , Animals
19.
Sci Adv ; 10(25): eadm9228, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905339

ABSTRACT

Transient receptor potential melastatin 8 (TRPM8) is a temperature- and menthol-sensitive ion channel that contributes to diverse physiological roles, including cold sensing and pain perception. Clinical trials targeting TRPM8 have faced repeated setbacks predominantly due to the knowledge gap in unraveling the molecular underpinnings governing polymodal activation. A better understanding of the molecular foundations between the TRPM8 activation modes may aid the development of mode-specific, thermal-neutral therapies. Ancestral sequence reconstruction was used to explore the origins of TRPM8 activation modes. By resurrecting key TRPM8 nodes along the human evolutionary trajectory, we gained valuable insights into the trafficking, stability, and function of these ancestral forms. Notably, this approach unveiled the differential emergence of cold and menthol sensitivity over evolutionary time, providing a fresh perspective on complex polymodal behavior. These studies provide a paradigm for understanding polymodal behavior in TRPM8 and other proteins with the potential to enhance our understanding of sensory receptor biology and pave the way for innovative therapeutic interventions.


Subject(s)
Cold Temperature , Menthol , TRPM Cation Channels , TRPM Cation Channels/metabolism , TRPM Cation Channels/genetics , Humans , Menthol/pharmacology , Evolution, Molecular , Phylogeny , Thermosensing
20.
J Ethnopharmacol ; 334: 118464, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-38908492

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Paeonol (PAE) and glycyrrhizic acid (GLY) are predominate components of 14 blood-entering ones of Piantongtang No. 1, which is a traditional Chinese medicine prescription for chronic migraine with minimal side effects. Both paeonol and glycyrrhizic acid exhibit analgesic, neuroprotective and anti-inflammatory properties individually. Our previous research has highlighted their combined effect (PAE + GLY) in ameliorating migraine symptoms. However, there are not yet any studies exploring the mechanism of action of PAE + GLY in the treatment of migraine. AIM OF THE STUDY: This research aimed to determine the mechanism of PAE + GLY in ameliorating the recurrent nitroglycerin-induced migraine-like phenotype in rats. MATERIALS AND METHODS: Using a nitroglycerin-induced migraine model via subcutaneous injection in the neck, we evaluated the effect of PAE + GLY on migraine-like symptoms. Behavioural tests and biomarkers analysis were employed, alongside transcriptome sequencing (RNA-seq). Mechanistic insights were further verified utilising reverse transcription quantitative PCR (RT-qPCR), Western blot (WB), ELISA and immunofluorescence (IF) techniques. RESULTS: Following treatment with PAE + GLY, hyperalgesia threshold and 5-hydroxytryptamine (5-HT) levels increased, and migraine-like head scratching, histamine and calcitonin gene-related peptide (CGRP) levels were reduced. RNA-Seq experiments revealed that PAE + GLY upregulated the expression of Glutamate decarboxylase 2 (GAD2) and γ-aminobutyric acid type B receptor subunit 2 (GABBR2) genes. This upregulation activated the GABAergic synapse pathway, effectively inhibiting migraine attacks. Further validation demonstrated an increase in γ-aminobutyric acid (GABA) content in cerebrospinal fluid post PAE + GLY treatment, coupled with increased expression of dural GAD2, GABBR2 and transient receptor potential channel M8 (TRPM8). Consequently, this inhibited the expression of dural cAMP-dependent protein kinase catalytic subunit alpha (PRKACA) and transient receptor potential channel type 1 (TRPV1), subsequently downregulating p-ERK1/2, p-AKT1, IL-1ß and TNF-α. CONCLUSIONS: Our findings underscore that PAE + GLY ameliorates inflammatory hyperalgesia migraine by upregulating inhibitory neurotransmitters and modulating the GABBR2/TRPM8/PRKACA/TRPV1 pathway.


Subject(s)
Acetophenones , Glycyrrhizic Acid , Migraine Disorders , Nitroglycerin , TRPM Cation Channels , TRPV Cation Channels , Animals , Male , Rats , Acetophenones/pharmacology , Disease Models, Animal , Drug Therapy, Combination , Glycyrrhizic Acid/pharmacology , Glycyrrhizic Acid/therapeutic use , Migraine Disorders/drug therapy , Migraine Disorders/chemically induced , Migraine Disorders/metabolism , Nitroglycerin/toxicity , Nitroglycerin/pharmacology , Phenotype , Protein Kinase C-alpha/metabolism , Protein Kinase C-alpha/genetics , Rats, Sprague-Dawley , Receptors, GABA/metabolism , Receptors, GABA/genetics , Signal Transduction/drug effects , TRPM Cation Channels/metabolism , TRPM Cation Channels/genetics , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL