1
|
Asuthkar S, Bayen S, Saldes EB, Tom B, Velpula J, Siddharth S, Koeltzow TE, Vander Griend DJ. The Impact of TRPM8 on Prostate Cancer Transcriptomic Dynamics. Cells 2025; 14:501. [PMID: 40214455 PMCID: PMC11988096 DOI: 10.3390/cells14070501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/21/2025] [Accepted: 03/22/2025] [Indexed: 04/14/2025] Open
Abstract
Prostate cancer (PC) remains a significant health challenge, with androgen receptor (AR) signaling playing a pivotal role in its progression. This study investigates the expression and functional implications of the transient receptor potential melastatin 8 (TRPM8) channel in PC, focusing on its interaction with AR and its impact on oncogenic pathways. We analyzed mRNA expression levels of TRPM8 and AR in PC tissues, revealing that TRPM8 is upregulated in benign and early-stage tumors but significantly downregulated in metastatic samples. This decline correlates with increased AR expression, suggesting a compensatory mechanism that enhances AR-driven tumorigenesis. RNA sequencing and pathway enrichment analyses demonstrated that TRPM8 knockout (KO) prostates exhibited significant alterations in gene expression, particularly in pathways related to extracellular matrix (ECM) remodeling, cell proliferation, and survival signaling. Notably, genes associated with metastasis, such as MMP2 and FAP, were upregulated in TRPM8 KO samples, indicating a potential role for TRPM8 in inhibiting tumor invasion. Furthermore, Gene Set Enrichment Analysis (GSEA) revealed positive enrichment of androgen response, angiogenesis, and epithelial-mesenchymal transition (EMT) pathways in TRPM8 KO prostates, reinforcing the notion that TRPM8 loss creates a pro-tumorigenic environment. Our findings suggest that TRPM8 functions as a molecular brake on PC progression, and its loss may contribute to the development of aggressive disease phenotypes. This study underscores the importance of TRPM8 as a potential therapeutic target and biomarker in PC, warranting further investigation into its role in cancer biology and treatment response.
Collapse
Affiliation(s)
- Swapna Asuthkar
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA (E.B.S.); (B.T.); (S.S.); (T.E.K.)
- Department of Pediatrics, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| | - Susovon Bayen
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA (E.B.S.); (B.T.); (S.S.); (T.E.K.)
| | - Erick B. Saldes
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA (E.B.S.); (B.T.); (S.S.); (T.E.K.)
| | - Benny Tom
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA (E.B.S.); (B.T.); (S.S.); (T.E.K.)
| | | | - Sarangi Siddharth
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA (E.B.S.); (B.T.); (S.S.); (T.E.K.)
- OSF Saint Francis Medical Center, Peoria, IL 61637, USA
| | - Timothy E. Koeltzow
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA (E.B.S.); (B.T.); (S.S.); (T.E.K.)
- Department of Psychology, Bradley University, Peoria, IL 61625, USA
| | | |
Collapse
|
2
|
Liu TT, Chen PY, Tseng CY, Chen YN, Chen JB, Ni TH, Wang SJ, Chen SP, Yen JC. Activation of central and peripheral transient receptor potential melastatin 8 increases susceptibility to spreading depolarization and facilitates trigeminal neuroinflammation. J Headache Pain 2025; 26:55. [PMID: 40087597 PMCID: PMC11907788 DOI: 10.1186/s10194-025-01997-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 03/09/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Transient receptor potential melastatin 8 (TRPM8), a gene encoding a nonselective cation channel responsive to cold stimuli, has been implicated in migraine susceptibility. Despite this association, the role of TRPM8 to migraine pathogenesis remains elusive. This study aims to elucidate the potential role of TRPM8 in migraine pathophysiology. METHODS TRPM8 expression in the cortex and primary trigeminal ganglion (TG) cells was analyzed via immunostaining. The central role of TRPM8 was assessed using a spreading depolarization (SD) model, where intracerebroventricular injections or topical applications of TRPM8 agonists and antagonists were administered to rats to investigate their effects on KCl-evoked SD and SD-induced cortical inflammation. The peripheral role of TRPM8 in migraine was evaluated using primary cultures of rat TG cells by analyzing the effects of TRPM8 activation on calcitonin gene-related peptide (CGRP) expression, release, and trigeminal neuroinflammation. RESULTS TRPM8 was homogeneously distributed in the cerebral cortex, predominantly co-localizing with cortical neurons. Activation of cortical TRPM8 increased the frequency of KCl-evoked SD and exacerbated SD-induced cortical inflammation. Interestingly. Interestingly, inhibition of cerebral TRPM8 had negligible effects. In TG primary cultures, TRPM8 activation upregulated CGRP expression and release and induced cyclooxygenase-2 (Cox2) upregulation via a calmodulin kinase II (CaMKII)-dependent mechanism. CONCLUSIONS TRPM8 activation increased susceptibility to SD and facilitated the effects of CGRP and trigeminal neuroinflammation, implicating that TRPM8 may contribute to migraine pathophysiology through central and peripheral mechanisms.
Collapse
Affiliation(s)
- Tzu-Ting Liu
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, 5th floor, Shouren Building, No. 155, Sec. 2, Linong St., Beitou District, Taipei, 112, Taiwan
| | - Pin-Yu Chen
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Faculty of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Chyun-Yea Tseng
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, 5th floor, Shouren Building, No. 155, Sec. 2, Linong St., Beitou District, Taipei, 112, Taiwan
| | - Yun-Ning Chen
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, 5th floor, Shouren Building, No. 155, Sec. 2, Linong St., Beitou District, Taipei, 112, Taiwan
| | - Jian-Bang Chen
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, 5th floor, Shouren Building, No. 155, Sec. 2, Linong St., Beitou District, Taipei, 112, Taiwan
| | - Tz-Han Ni
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, 5th floor, Shouren Building, No. 155, Sec. 2, Linong St., Beitou District, Taipei, 112, Taiwan
| | - Shuu-Jiun Wang
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Faculty of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, 112201, Taiwan
| | - Shih-Pin Chen
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, 112201, Taiwan
- Division of Translational Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, 11217, Taiwan
| | - Jiin-Cherng Yen
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, 5th floor, Shouren Building, No. 155, Sec. 2, Linong St., Beitou District, Taipei, 112, Taiwan.
| |
Collapse
|
3
|
Vigano M, Wang L, As’sadiq A, Samarani S, Ahmad A, Costiniuk CT. Impact of cannabinoids on cancer outcomes in patients receiving immune checkpoint inhibitor immunotherapy. Front Immunol 2025; 16:1497829. [PMID: 40109334 PMCID: PMC11919899 DOI: 10.3389/fimmu.2025.1497829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025] Open
Abstract
Cannabinoids relieve pain, nausea, anorexia and anxiety, and improve quality of life in several cancer patients. The immunotherapy with checkpoint inhibitors (ICIs), although very successful in a subset of patients, is accompanied by moderate to severe immune-related adverse events (ir-AE) that often necessitate its discontinuation. Because of their role in symptomatic relief, cannabinoids have been used in combination with immune checkpoint inhibitor (ICI) immunotherapy. A few studies strongly suggest that the use of medicinal cannabis in cancer patients attenuates many of the ir-AE associated with the use of ICI immunotherapy and increase its tolerability. However, no significant beneficial effects on overall survival, progression free survival or cancer relapses were observed; rather, some of the studies noted adverse effects of concurrent administration of cannabinoids with ICI immunotherapy on the clinical benefits of the latter. Because of cannabinoids' well documented immunosuppressive effects mediated through the cannabinoid recptor-2 (CB2), we propose considering this receptor as an inhibitory immune checkpoint per se. A simultaneous neutralization of CB2, concurrent with cannabinoid treatment, may lead to better clinical outcomes in cancer patients receiving ICI immunotherapy. In this regard, cannabinoids such as cannabidiol (CBD) and cannabigerol (CBG), with little agonism for CB2, may be better therapeutic choices. Additional strategies e.g., the use of monoacylglycerol lipase (MAGL) inhibitors that degrade some endocannabinoids as well as lipogenesis and formation of lipid bilayers in cancer cells may also be explored. Future studies should take into consideration gut microbiota, CYP450 polymorphism and haplotypes, cannabinoid-drug interactions as well as genetic and somatic variations occurring in the cannabinoid receptors and their signaling pathways in cancer cells for personalized cannabis-based therapies in cancer patients receiving ICIs. This may lead to rational knowledge-based regimens tailored to individual cancer patients.
Collapse
Affiliation(s)
- MariaLuisa Vigano
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Lixing Wang
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Alia As’sadiq
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Suzanne Samarani
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Ali Ahmad
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Cecilia T. Costiniuk
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Infectious Diseases and Chronic Viral Illnesses Service, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
4
|
Becker J, Ellerkmann CS, Schmelzer H, Hermann C, Lützel K, Gudermann T, Konrad DB, Trauner D, Storch U, Mederos Y Schnitzler M. Optical Control of TRPM8 Channels with Photoswitchable Menthol. Angew Chem Int Ed Engl 2025; 64:e202416549. [PMID: 39660776 DOI: 10.1002/anie.202416549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/21/2024] [Accepted: 12/10/2024] [Indexed: 12/12/2024]
Abstract
Transient receptor potential melastatin 8 (TRPM8) channels are well known as sensors for cold temperatures and cooling agents such as menthol and icilin and these channels are tightly regulated by the membrane lipid phosphoinositol-4,5-bisphosphate (PIP2). Since TRPM8 channels emerged as promising drug targets for treating pain, itching, obesity, cancer, dry eye disease, and inflammation, we aimed at developing a high-precision TRPM8 channel activator, to achieve spatiotemporal control of TRPM8 activity with light. In this study, we designed, synthesized and characterized the first photoswitchable TRPM8 activator azo-menthol (AzoM). AzoM enables optical control of endogenously and heterologously expressed TRPM8 channels with UV and blue light which is demonstrated by performing patch-clamp experiments. Moreover, AzoM facilitates the reliable determination of activation, inactivation, and deactivation kinetics thereby providing further insights into the channel gating. Using AzoM, the specific roles of individual amino acids for AzoM or PIP2 binding and for sensitization by PIP2 can be elucidated. Altogether, AzoM represents as a high-precision pharmaceutical tool for reversible control of TRPM8 channel function that enhances our biophysical understanding of TRPM8 channels and holds the potential to support the development of novel pharmaceuticals.
Collapse
Affiliation(s)
- Jasmin Becker
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, Goethestr. 33, 80336, Munich, Germany
| | - Clara S Ellerkmann
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, Goethestr. 33, 80336, Munich, Germany
| | - Hannah Schmelzer
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, Goethestr. 33, 80336, Munich, Germany
| | - Christian Hermann
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, Goethestr. 33, 80336, Munich, Germany
| | - Kyra Lützel
- Department of Pharmacy, Ludwig Maximilian University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, Goethestr. 33, 80336, Munich, Germany
- Comprehensive Pneumology Center Munich (CPC-M), German Center for Lung Research, 81377, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Munich Heart Alliance, 80336, Munich, Germany
| | - David B Konrad
- Department of Pharmacy, Ludwig Maximilian University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Dirk Trauner
- Department of Chemistry College of Arts and Sciences, University of Pennsylvania, 231 South 34th Street, Philadelphia, Pennsylvania, 19104-6323, United States
| | - Ursula Storch
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, Goethestr. 33, 80336, Munich, Germany
- Institute of Pharmacy, University of Regensburg, Universitätsstr. 31, 93040, Regensburg, Germany
| | - Michael Mederos Y Schnitzler
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, Goethestr. 33, 80336, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Munich Heart Alliance, 80336, Munich, Germany
| |
Collapse
|
5
|
Kashio M. Thermo-TRP regulation by endogenous factors and its physiological function at core body temperature. Physiol Rep 2025; 13:e70164. [PMID: 39793986 PMCID: PMC11723785 DOI: 10.14814/phy2.70164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 11/01/2024] [Accepted: 12/10/2024] [Indexed: 01/13/2025] Open
Abstract
Transient receptor potential (TRP) channels with temperature sensitivities (thermo-TRPs) are involved in various physiological processes. Thermo-TRPs that detect temperature changes in peripheral sensory neurons possess indispensable functions in thermosensation, eliciting defensive behavior against noxious temperatures and driving autonomic/behavioral thermoregulatory responses to maintain body temperature in mammals. Moreover, most thermo-TRPs are functionally expressed in cells and tissues where the temperature is maintained at a constant core body temperature. To perform physiological functions, the activity of each thermo-TRP channel must be regulated by endogenous mechanisms at body temperature. Dysregulation of this process can lead to various diseases. This review highlights the endogenous factors regulating thermo-TRP activity and physiological functions at constant core body temperature.
Collapse
Affiliation(s)
- Makiko Kashio
- Department of Cell PhysiologyKumamoto UniversityKumamotoJapan
| |
Collapse
|
6
|
Saldes EB, Erdmier A, Velpula J, Koeltzow TE, Zhu MX, Asuthkar S. Transcriptomic Profile Analysis of Brain Tissue in the Absence of Functional TRPM8 Calcium Channel. Biomedicines 2024; 13:75. [PMID: 39857659 PMCID: PMC11760472 DOI: 10.3390/biomedicines13010075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/20/2024] [Accepted: 12/25/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Transient Receptor Potential Melastatin 8 (TRPM8) is a non-selective, Ca2+-permeable cation channel involved in thermoregulation and other physiological processes, such as basal tear secretion, cell differentiation, and insulin homeostasis. The activation and deactivation of TRPM8 occur through genetic modifications, channel interactions, and signaling cascades. Recent evidence suggests a significant role of TRPM8 in the hypothalamus and amygdala related to pain sensation and sexual behavior. Notably, TRPM8 has been implicated in neuropathic pain, migraines, and neurodegenerative diseases such as Parkinson's disease. Our laboratory has identified testosterone as a high-affinity ligand of TRPM8. TRPM8 deficiency appears to influence behavioral traits in mice, like increased aggression and deficits in sexual satiety. Here, we aim to explore the pathways altered in brain tissues of TRPM8-deficient mice using the expression and methylation profiles of messenger RNA (mRNA) and long non-coding RNA (lncRNA). Specifically, we focused on brain regions integral to behavioral and hormonal control, including the olfactory bulb, hypothalamus, amygdala, and insula. Methods: RNA was isolated and purified for microarray analysis collected from male wild-type and TRPM8 knockout mice. Results: We identified various differentially expressed genes tied to multiple signaling pathways. Among them, the androgen-estrogen receptor (AR-ER) pathway, steroidogenesis pathway, sexual reward pathway, and cocaine reward pathway are particularly worth noting. Conclusions: These results should bridge the existing gaps in the knowledge regarding TRPM8 and inform potential targets for future studies to elucidate its role in the behavior changes and pathology of the diseases associated with TRPM8 activity.
Collapse
Affiliation(s)
- Erick B. Saldes
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (E.B.S.); (A.E.); (T.E.K.)
| | - Alexandra Erdmier
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (E.B.S.); (A.E.); (T.E.K.)
| | | | - Timothy E. Koeltzow
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (E.B.S.); (A.E.); (T.E.K.)
- Department of Psychology, Bradley University, Peoria, IL 61625, USA
| | - Michael X. Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| | - Swapna Asuthkar
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (E.B.S.); (A.E.); (T.E.K.)
- Department of Pediatrics, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| |
Collapse
|
7
|
Zadmajid V, Shahriar S, Gorelick DA. Testosterone acts through the membrane protein GPRC6A to cause cardiac edema in zebrafish embryos. Development 2024; 151:dev204390. [PMID: 39479956 PMCID: PMC11634029 DOI: 10.1242/dev.204390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 10/06/2024] [Indexed: 11/02/2024]
Abstract
Androgens are classically thought to act through intracellular androgen receptors (AR/NR3C4), but they can also trigger non-genomic effects via membrane proteins. Although several membrane androgen receptors have been characterized in vitro, their functions in vivo remain unclear. Using a chemical-genetic screen in zebrafish, we found that GPRC6A, a G-protein-coupled receptor, mediates non-genomic androgen actions during embryonic development. Exposure to androgens (androstanedione, DHT and testosterone) caused cardiac edema or tail curvature in wild-type embryos, as well as in ar mutants, suggesting AR-independent pathways. We then mutated putative membrane androgen receptors [gprc6a, hcar1-4 and zip9 (slc39a9)] and found that only gprc6a mutants exhibited a significant reduction in cardiac edema after testosterone exposure. Additionally, co-treatment of wild-type embryos with testosterone and GPRC6A antagonists significantly suppressed the cardiac edema phenotype. Using RNA-seq and RNA rescue approaches, we found that testosterone and GPRC6A cause cardiac phenotypes by reducing Pak1 signaling. Our results indicate that testosterone induces cardiac edema in zebrafish embryos through GPRC6A, independent of nuclear androgen receptors, highlighting a previously unappreciated non-genomic androgen signaling pathway in embryonic development.
Collapse
MESH Headings
- Animals
- Zebrafish/embryology
- Zebrafish/metabolism
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Testosterone/metabolism
- Testosterone/pharmacology
- Zebrafish Proteins/metabolism
- Zebrafish Proteins/genetics
- Edema, Cardiac/metabolism
- Edema, Cardiac/pathology
- Edema, Cardiac/genetics
- Embryo, Nonmammalian/metabolism
- Embryo, Nonmammalian/drug effects
- Receptors, Androgen/metabolism
- Receptors, Androgen/genetics
- Signal Transduction/drug effects
- Gene Expression Regulation, Developmental/drug effects
- Heart/embryology
- Heart/drug effects
- Androgens/pharmacology
- Androgens/metabolism
- Mutation/genetics
Collapse
Affiliation(s)
- Vahid Zadmajid
- Center for Precision Environmental Health, Department of Molecular & Cellular Biology, Baylor College of Medicine, One Baylor Plaza, BCM229, Houston, TX 77030, USA
| | - Shayan Shahriar
- Center for Precision Environmental Health, Department of Molecular & Cellular Biology, Baylor College of Medicine, One Baylor Plaza, BCM229, Houston, TX 77030, USA
| | - Daniel A. Gorelick
- Center for Precision Environmental Health, Department of Molecular & Cellular Biology, Baylor College of Medicine, One Baylor Plaza, BCM229, Houston, TX 77030, USA
| |
Collapse
|
8
|
Shikha D, Dalai R, Kumar S, Goswami C. Residues of TRPM8 at the Lipid-Water-Interface have Coevolved with Cholesterol Interaction and are Relevant for Diverse Health Disorders. J Membr Biol 2024; 257:345-364. [PMID: 39150496 PMCID: PMC11584472 DOI: 10.1007/s00232-024-00319-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024]
Abstract
TRPM8 is a non-selective cation channel that is expressed in several tissues and cells and also has a unique property to be activated by low-temperature. In this work, we have analyzed the conservation of amino acids that are present in the lipid-water-interface (LWI) region of TRPM8, the region which experiences a microenvironment near the membrane surface. We demonstrate that the amino acids present in the LWI region are more conserved than the transmembrane or even full-length TRPM8, suggesting strong selection pressure in these residues. TRPM8 also has several conserved cholesterol-binding motifs where cholesterol can bind in different modes and energies. We suggest that mutations and/or physiological conditions can potentially alter these TRPM8-cholesterol complexes and can lead to physiological disorders or even apparently irreversible diseases such as cancer and neurodegeneration.
Collapse
Affiliation(s)
- Deep Shikha
- School of Biological Sciences, National Institute of Science Education and Research, An OCC of Homi Bhabha National Institute, Khordha, Jatni, Odisha, 752050, India
| | - Ritesh Dalai
- School of Biological Sciences, National Institute of Science Education and Research, An OCC of Homi Bhabha National Institute, Khordha, Jatni, Odisha, 752050, India
| | - Shamit Kumar
- School of Biological Sciences, National Institute of Science Education and Research, An OCC of Homi Bhabha National Institute, Khordha, Jatni, Odisha, 752050, India
| | - Chandan Goswami
- School of Biological Sciences, National Institute of Science Education and Research, An OCC of Homi Bhabha National Institute, Khordha, Jatni, Odisha, 752050, India.
| |
Collapse
|
9
|
Ghovanloo MR, Effraim PR, Tyagi S, Aldrich AM, Cheng X, Yuan JH, Schulman BR, Jacobs DS, Dib-Hajj SD, Waxman SG. TRPM8 Mutations Associated With Persistent Pain After Surgical Injury of Corneal Trigeminal Axons. Neurol Genet 2024; 10:e200206. [PMID: 39555137 PMCID: PMC11567650 DOI: 10.1212/nxg.0000000000200206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/09/2024] [Indexed: 11/19/2024]
Abstract
Background and Objectives Despite extensive efforts, the mechanisms underlying pain after axonal injury remain incompletely understood. Pain following corneal refractive surgery offers a valuable human model for investigating trigeminal axonal injury because laser-assisted in situ keratomileusis (LASIK) severs axons of trigeminal ganglion neurons innervating the cornea. While the majority of patients are pain-free shortly after surgery, a minority endure persistent postoperative ocular pain. Through genomic analysis of patients experiencing persistent postoperative ocular pain, we identified rare variants in genes encoding ion channels and receptors, including TRPM8, which codes for the menthol-sensitive and cold-sensing transient receptor potential cation channel. Methods We conducted a profiling of 2 TRPM8 mutant variants, D665N and V915M, which were identified in patients suffering from persistent pain after LASIK surgery. We used patch-clamp and multielectrode array (MEA) recordings to investigate the biophysical and pharmacologic properties of mutant vs wild-type (WT) channels. Results Patch-clamp analysis shows that these mutations shift the activation curves of TRPM8 in a hyperpolarized direction, with this effect being significantly different between WT and D665N channels. In addition, both mutations significantly increase channel sensitivity to the canonical ligand, menthol. MEA recordings from transfected rat trigeminal ganglion neurons indicate that expression of D665N and V915M mutant channels increases spontaneous activity compared with WT channels. Consistent with patch-clamp results, neuronal activity in MEA recordings was increased on exposure to menthol. Discussion Collectively, our findings suggest that proexcitatory mutations of TRPM8, in the context of axonal injury within the cornea, can produce trigeminal ganglion neuron hyperexcitability that contributes to persistent postoperative ocular pain. In addition to providing additional evidence for a role of TRPM8 in human pain, our results suggest that inhibitors of this channel merit future study.
Collapse
Affiliation(s)
- Mohammad-Reza Ghovanloo
- From the Department of Neurology (M.-R.G., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University School of Medicine, New Haven; Center for Neuroscience and Regeneration Research (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University, New Haven; Neuro-Rehabilitation Research Center (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Veterans Affairs Connecticut Healthcare System, West Haven; Department of Anesthesiology (P.R.E.), Yale University School of Medicine, New Haven, CT; and Department of Ophthalmology (D.S.J.), Massachusetts Eye and Ear, Harvard Medical School, Boston
| | - Philip R Effraim
- From the Department of Neurology (M.-R.G., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University School of Medicine, New Haven; Center for Neuroscience and Regeneration Research (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University, New Haven; Neuro-Rehabilitation Research Center (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Veterans Affairs Connecticut Healthcare System, West Haven; Department of Anesthesiology (P.R.E.), Yale University School of Medicine, New Haven, CT; and Department of Ophthalmology (D.S.J.), Massachusetts Eye and Ear, Harvard Medical School, Boston
| | - Sidharth Tyagi
- From the Department of Neurology (M.-R.G., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University School of Medicine, New Haven; Center for Neuroscience and Regeneration Research (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University, New Haven; Neuro-Rehabilitation Research Center (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Veterans Affairs Connecticut Healthcare System, West Haven; Department of Anesthesiology (P.R.E.), Yale University School of Medicine, New Haven, CT; and Department of Ophthalmology (D.S.J.), Massachusetts Eye and Ear, Harvard Medical School, Boston
| | - Alecia M Aldrich
- From the Department of Neurology (M.-R.G., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University School of Medicine, New Haven; Center for Neuroscience and Regeneration Research (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University, New Haven; Neuro-Rehabilitation Research Center (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Veterans Affairs Connecticut Healthcare System, West Haven; Department of Anesthesiology (P.R.E.), Yale University School of Medicine, New Haven, CT; and Department of Ophthalmology (D.S.J.), Massachusetts Eye and Ear, Harvard Medical School, Boston
| | - Xiaoyang Cheng
- From the Department of Neurology (M.-R.G., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University School of Medicine, New Haven; Center for Neuroscience and Regeneration Research (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University, New Haven; Neuro-Rehabilitation Research Center (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Veterans Affairs Connecticut Healthcare System, West Haven; Department of Anesthesiology (P.R.E.), Yale University School of Medicine, New Haven, CT; and Department of Ophthalmology (D.S.J.), Massachusetts Eye and Ear, Harvard Medical School, Boston
| | - Jun-Hui Yuan
- From the Department of Neurology (M.-R.G., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University School of Medicine, New Haven; Center for Neuroscience and Regeneration Research (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University, New Haven; Neuro-Rehabilitation Research Center (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Veterans Affairs Connecticut Healthcare System, West Haven; Department of Anesthesiology (P.R.E.), Yale University School of Medicine, New Haven, CT; and Department of Ophthalmology (D.S.J.), Massachusetts Eye and Ear, Harvard Medical School, Boston
| | - Betsy R Schulman
- From the Department of Neurology (M.-R.G., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University School of Medicine, New Haven; Center for Neuroscience and Regeneration Research (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University, New Haven; Neuro-Rehabilitation Research Center (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Veterans Affairs Connecticut Healthcare System, West Haven; Department of Anesthesiology (P.R.E.), Yale University School of Medicine, New Haven, CT; and Department of Ophthalmology (D.S.J.), Massachusetts Eye and Ear, Harvard Medical School, Boston
| | - Deborah S Jacobs
- From the Department of Neurology (M.-R.G., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University School of Medicine, New Haven; Center for Neuroscience and Regeneration Research (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University, New Haven; Neuro-Rehabilitation Research Center (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Veterans Affairs Connecticut Healthcare System, West Haven; Department of Anesthesiology (P.R.E.), Yale University School of Medicine, New Haven, CT; and Department of Ophthalmology (D.S.J.), Massachusetts Eye and Ear, Harvard Medical School, Boston
| | - Sulayman D Dib-Hajj
- From the Department of Neurology (M.-R.G., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University School of Medicine, New Haven; Center for Neuroscience and Regeneration Research (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University, New Haven; Neuro-Rehabilitation Research Center (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Veterans Affairs Connecticut Healthcare System, West Haven; Department of Anesthesiology (P.R.E.), Yale University School of Medicine, New Haven, CT; and Department of Ophthalmology (D.S.J.), Massachusetts Eye and Ear, Harvard Medical School, Boston
| | - Stephen G Waxman
- From the Department of Neurology (M.-R.G., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University School of Medicine, New Haven; Center for Neuroscience and Regeneration Research (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Yale University, New Haven; Neuro-Rehabilitation Research Center (M.-R.G., P.R.E., S.T., A.M.A., X.C., J.-H.Y., B.R.S., S.D.D.-H., S.G.W.), Veterans Affairs Connecticut Healthcare System, West Haven; Department of Anesthesiology (P.R.E.), Yale University School of Medicine, New Haven, CT; and Department of Ophthalmology (D.S.J.), Massachusetts Eye and Ear, Harvard Medical School, Boston
| |
Collapse
|
10
|
Behrendt M. Implications of TRPM3 and TRPM8 for sensory neuron sensitisation. Biol Chem 2024; 405:583-599. [PMID: 39417661 DOI: 10.1515/hsz-2024-0045] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Sensory neurons serve to receive and transmit a wide range of information about the conditions of the world around us as well as the external and internal state of our body. Sensitisation of these nerve cells, i.e. becoming more sensitive to stimuli or the emergence or intensification of spontaneous activity, for example in the context of inflammation or nerve injury, can lead to chronic diseases such as neuropathic pain. For many of these disorders there are only very limited treatment options and in order to find and establish new therapeutic approaches, research into the exact causes of sensitisation with the elucidation of the underlying mechanisms and the identification of the molecular components is therefore essential. These components include plasma membrane receptors and ion channels that are involved in signal reception and transmission. Members of the transient receptor potential (TRP) channel family are also expressed in sensory neurons and some of them play a crucial role in temperature perception. This review article focuses on the heat-sensitive TRPM3 and the cold-sensitive TRPM8 (and TRPA1) channels and their importance in sensitisation of dorsal root ganglion sensory neurons is discussed based on studies related to inflammation and injury- as well as chemotherapy-induced neuropathy.
Collapse
Affiliation(s)
- Marc Behrendt
- Experimental Pain Research, Medical Faculty Mannheim, Heidelberg University, MCTN, Tridomus, Building C, Ludolf-Krehl-Straße 13-17, D-68167 Mannheim, Germany
| |
Collapse
|
11
|
Arcas JM, Oudaha K, González A, Fernández-Trillo J, Peralta FA, Castro-Marsal J, Poyraz S, Taberner F, Sala S, de la Peña E, Gomis A, Viana F. The ion channel TRPM8 is a direct target of the immunosuppressant rapamycin in primary sensory neurons. Br J Pharmacol 2024; 181:3192-3214. [PMID: 38741464 DOI: 10.1111/bph.16402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND AND PURPOSE The mechanistic target of rapamycin (mTOR) signalling pathway is a key regulator of cell growth and metabolism. Its deregulation is implicated in several diseases. The macrolide rapamycin, a specific inhibitor of mTOR, has immunosuppressive, anti-inflammatory and antiproliferative properties. Recently, we identified tacrolimus, another macrolide immunosuppressant, as a novel activator of TRPM8 ion channels, involved in cold temperature sensing, thermoregulation, tearing and cold pain. We hypothesized that rapamycin may also have agonist activity on TRPM8 channels. EXPERIMENTAL APPROACH Using calcium imaging and electrophysiology in transfected HEK293 cells and wildtype or Trpm8 KO mouse DRG neurons, we characterized rapamycin's effects on TRPM8 channels. We also examined the effects of rapamycin on tearing in mice. KEY RESULTS Micromolar concentrations of rapamycin activated rat and mouse TRPM8 channels directly and potentiated cold-evoked responses, effects also observed in human TRPM8 channels. In cultured mouse DRG neurons, rapamycin increased intracellular calcium levels almost exclusively in cold-sensitive neurons. Responses were markedly decreased in Trpm8 KO mice or by TRPM8 channel antagonists. Cutaneous cold thermoreceptor endings were also activated by rapamycin. Topical application of rapamycin to the eye surface evokes tearing in mice by a TRPM8-dependent mechanism. CONCLUSION AND IMPLICATIONS These results identify TRPM8 cationic channels in sensory neurons as novel molecular targets of the immunosuppressant rapamycin. These findings may help explain some of its therapeutic effects after topical application to the skin and the eye surface. Moreover, rapamycin could be used as an experimental tool in the clinic to explore cold thermoreceptors.
Collapse
Affiliation(s)
- José Miguel Arcas
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Khalid Oudaha
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Alejandro González
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Jorge Fernández-Trillo
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | | | - Júlia Castro-Marsal
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Seyma Poyraz
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Francisco Taberner
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Salvador Sala
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Elvira de la Peña
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Ana Gomis
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Félix Viana
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| |
Collapse
|
12
|
Qiao S, Wu F, Wang H. Genetic and immune identification and functional analysis of TRPM8 as a potential biomarker for pancreatic adenocarcinoma proliferation. Cancer Rep (Hoboken) 2024; 7:e2108. [PMID: 38837874 PMCID: PMC11150080 DOI: 10.1002/cnr2.2108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 03/26/2024] [Accepted: 04/30/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma (PAAD), a member of highly lethal malignant tumors, has a poor outcome and extremely poor prognosis. The transient receptor potential (TRP) superfamily, a group of nonselective cation channels, is capable of influencing cellular functions by regulating calcium homeostasis. In addition, it has been shown that TRP channels can also affect various cellular phenotypes by regulating gene transcription levels and are involved in the development of a variety of malignant tumors. AIMS In order to find new therapeutic targets and biomarkers to improve the clinical prognosis of pancreatic cancer, we performed genetic and immunological characterization of TRP channels in PAAD, as well as related functional and prognostic analyses. METHODS AND RESULTS We investigated the expression, genetic alterations, methylation levels, and immune infiltration levels of TRP channels in PAAD, and further also analyzed the function of TRP channels in PAAD and their prognostic value for PAAD patients. Our results suggest that TRPM8 may contribute to tumor proliferation by controlling the PI3K-AKT-mTOR signaling pathway in PAAD. CONCLUSION After careful evaluation of the accumulated data, we concluded that TRPM8 has potential as a prognostic indicator and prospective therapeutic target in PAAD.
Collapse
Affiliation(s)
- Sen Qiao
- Assisted Reproduction CenterNorthwest Women's and Children's HospitalXi'anChina
| | - Fengming Wu
- School of MedicineSoutheast UniversityNanjingJiangsuChina
| | - Hongmei Wang
- School of MedicineSoutheast UniversityNanjingJiangsuChina
- Shaanxi University of Chinese MedicineXianyangChina
| |
Collapse
|
13
|
Carstens MI, Mahroke A, Selescu T, Carstens E. Role of thermosensitive transient receptor potential (TRP) channels in thermal preference of male and female mice. J Therm Biol 2024; 122:103868. [PMID: 38852485 PMCID: PMC11185440 DOI: 10.1016/j.jtherbio.2024.103868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 02/14/2024] [Accepted: 04/26/2024] [Indexed: 06/11/2024]
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.
Collapse
Affiliation(s)
- Mirela Iodi Carstens
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA, 95616, USA
| | - Avina Mahroke
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA, 95616, USA
| | - Tudor Selescu
- Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - E Carstens
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
14
|
Akosah Y, Yang J, Pavlov E. Inorganic polyphosphate and ion transport across biological membranes. Biochem Soc Trans 2024; 52:671-679. [PMID: 38630434 DOI: 10.1042/bst20230522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/22/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024]
Abstract
Inorganic polyphosphate (polyP) is widely recognized for playing important roles and processes involved in energy and phosphate storage, regulation of gene expression, and calcium signaling. The less well-known role of polyP is as a direct mediator of ion transport across biological membranes. Here, we will briefly summarize current knowledge of the molecular mechanisms of how polyP can be involved in membrane ion transport. We discuss three types of mechanisms that might involve polyP: (1) formation of non-protein channel complex that includes calcium, polyP, and polyhydroxybutyrate (PHB); (2) modulation of the channel activity of PHBlated protein channels; and (3) direct effects of polyP on the function of the voltage-gated ion channels in the process that do not involve PHB.
Collapse
Affiliation(s)
- Yaw Akosah
- Department of Molecular Pathobiology, New York University, New York, NY, U.S.A
| | - Jingyi Yang
- Department of Molecular Pathobiology, New York University, New York, NY, U.S.A
| | - Evgeny Pavlov
- Department of Molecular Pathobiology, New York University, New York, NY, U.S.A
| |
Collapse
|
15
|
Choi ME, Lee JH, Jung CJ, Lee WJ, Won CH, Lee MW, Chang SE. A randomized, double-blinded, vehicle-controlled clinical trial of topical cryosim-1, a synthetic TRPM8 agonist, in prurigo nodularis. J Cosmet Dermatol 2024; 23:931-937. [PMID: 38169089 DOI: 10.1111/jocd.16079] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/09/2023] [Accepted: 11/01/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Prurigo nodularis (PN) is an intensively pruritic skin disease that negatively influences quality of life. Cryosim-1 (Intrinsic IB Spot) is a synthetic, selective transient receptor potential melastatin 8 agonist. AIMS To investigate the efficacy and safety of cryosim-1 in PN patients. PATIENTS/METHODS A randomized, double-blinded, placebo-controlled clinical trial including 30 patients was conducted. The numerical rating scale (NRS) of pruritus was evaluated before and 2 h after cryosim-1 application at every visit. RESULTS At week 8, the mean pruritus NRS before serum application (4.7 ± 0.4 treatment, 6.1 ± 0.5 placebo; p = 0.045) and 2 h after serum application (2.8 ± 0.4 treatment, 4.3 ± 0.5 placebo; p = 0.031) were significantly lower in the treatment group, and the mean NRS for sleep disorder was significantly lower in the treatment group (2.2 ± 0.5 treatment, 4.2 ± 0.8 placebo; p = 0.031). The mean satisfaction scales for pruritus improvement were significantly higher in the treatment group (7.2 ± 0.6) than in the placebo group (4.0 ± 0.9; p = 0.005). There was no difference in TEWL between the two groups, and no adverse reactions were reported. CONCLUSIONS Cryosim-1 is a safe and effective topical treatment for PN patients.
Collapse
Affiliation(s)
- Myoung Eun Choi
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong Hyeon Lee
- Bio-Medical Institute of Technology (BMIT), University of Ulsan, College of Medicine, Ulsan, Korea
| | - Chang Jin Jung
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Woo Jin Lee
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chong Hyun Won
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mi Woo Lee
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung Eun Chang
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
16
|
Alaimo A, Genovesi S, Annesi N, De Felice D, Subedi S, Macchia A, La Manna F, Ciani Y, Vannuccini F, Mugoni V, Notarangelo M, Libergoli M, Broso F, Taulli R, Ala U, Savino A, Cortese M, Mirzaaghaei S, Poli V, Bonapace IM, Papotti MG, Molinaro L, Doglioni C, Caffo O, Anesi A, Nagler M, Bertalot G, Carbone FG, Barbareschi M, Basso U, Dassi E, Pizzato M, Romanel A, Demichelis F, Kruithof-de Julio M, Lunardi A. Sterile inflammation via TRPM8 RNA-dependent TLR3-NF-kB/IRF3 activation promotes antitumor immunity in prostate cancer. EMBO J 2024; 43:780-805. [PMID: 38316991 PMCID: PMC10907604 DOI: 10.1038/s44318-024-00040-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 01/06/2024] [Accepted: 01/10/2024] [Indexed: 02/07/2024] Open
Abstract
Inflammation is a common condition of prostate tissue, whose impact on carcinogenesis is highly debated. Microbial colonization is a well-documented cause of a small percentage of prostatitis cases, but it remains unclear what underlies the majority of sterile inflammation reported. Here, androgen- independent fluctuations of PSA expression in prostate cells have lead us to identify a prominent function of the Transient Receptor Potential Cation Channel Subfamily M Member 8 (TRPM8) gene in sterile inflammation. Prostate cells secret TRPM8 RNA into extracellular vesicles (EVs), which primes TLR3/NF-kB-mediated inflammatory signaling after EV endocytosis by epithelial cancer cells. Furthermore, prostate cancer xenografts expressing a translation-defective form of TRPM8 RNA contain less collagen type I in the extracellular matrix, significantly more infiltrating NK cells, and larger necrotic areas as compared to control xenografts. These findings imply sustained, androgen-independent expression of TRPM8 constitutes as a promoter of anticancer innate immunity, which may constitute a clinically relevant condition affecting prostate cancer prognosis.
Collapse
Affiliation(s)
- Alessandro Alaimo
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy.
| | - Sacha Genovesi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Nicole Annesi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Dario De Felice
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Saurav Subedi
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland
| | - Alice Macchia
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Federico La Manna
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland
| | - Yari Ciani
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Federico Vannuccini
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Vera Mugoni
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Michela Notarangelo
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Michela Libergoli
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Francesca Broso
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Riccardo Taulli
- Department of Oncology, University of Torino, Torino, Italy
- Center for Experimental Research and Medical Studies (CeRMS), AOU Città della Salute e della Scienza di Torino, Torino, Italy
| | - Ugo Ala
- Department of Veterinary Sciences, University of Torino, Torino, Italy
| | - Aurora Savino
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Martina Cortese
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Somayeh Mirzaaghaei
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Molecular Biotechnology Center (MBC) "Guido Tarone", University of Torino, Torino, Italy
| | - Valeria Poli
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Molecular Biotechnology Center (MBC) "Guido Tarone", University of Torino, Torino, Italy
| | - Ian Marc Bonapace
- Department of Biotechnology and Life Sciences, University of Insubria, Busto Arsizio, VA, Italy
| | - Mauro Giulio Papotti
- Department of Pathology, University of Torino and AOU Città della Salute e della Scienza di Torino, Torino, Italy
| | - Luca Molinaro
- Department of Pathology, University of Torino and AOU Città della Salute e della Scienza di Torino, Torino, Italy
| | - Claudio Doglioni
- Division of Pathology, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS Vita Salute, San Raffaele University, Milano, Italy
| | - Orazio Caffo
- Medical Oncology Department, Santa Chiara Hospital-APSS, Trento, Italy
| | - Adriano Anesi
- Operative Unit of Clinical Pathology, Santa Chiara Hospital-APSS, Trento, Italy
| | - Michael Nagler
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Giovanni Bertalot
- Operative Unit of Anatomy Pathology, Santa Chiara Hospital-APSS, Trento, Italy
- Centre for Medical Sciences-CISMed, University of Trento, Trento, Italy
| | | | - Mattia Barbareschi
- Operative Unit of Anatomy Pathology, Santa Chiara Hospital-APSS, Trento, Italy
- Centre for Medical Sciences-CISMed, University of Trento, Trento, Italy
| | - Umberto Basso
- Oncology 1 Unit, Department of Oncology, Istituto Oncologico Veneto IOV IRCCS, Padova, Italy
| | - Erik Dassi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Massimo Pizzato
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Alessandro Romanel
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Francesca Demichelis
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Marianna Kruithof-de Julio
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Andrea Lunardi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy.
| |
Collapse
|
17
|
Sim J, O'Guin E, Monahan K, Sugimoto C, McLean SA, Albertorio-Sáez L, Zhao Y, Laumet S, Dagenais A, Bernard MP, Folger JK, Robison AJ, Linnstaedt SD, Laumet G. Interleukin-10-producing monocytes contribute to sex differences in pain resolution in mice and humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.03.565129. [PMID: 37961295 PMCID: PMC10635095 DOI: 10.1101/2023.11.03.565129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Pain is closely associated with the immune system, which exhibits sexual dimorphism. For these reasons, neuro-immune interactions are suggested to drive sex differences in pain pathophysiology. However, our understanding of peripheral neuro-immune interactions on sex differences in pain resolution remains limited. Here, we have shown, in both a mouse model of inflammatory pain and in humans following traumatic pain, that males had higher levels of interleukin (IL)-10 than females, which were correlated with faster pain resolution. Following injury, we identified monocytes (CD11b+ Ly6C+ Ly6G-F4/80 mid ) as the primary source of IL-10, with IL-10-producing monocytes being more abundant in males than females. In a mouse model, neutralizing IL-10 signaling through antibodies, genetically ablating IL-10R1 in sensory neurons, or depleting monocytes with clodronate all impaired the resolution of pain hypersensitivity in both sexes. Furthermore, manipulating androgen levels in mice reversed the sexual dimorphism of pain resolution and the levels of IL-10-producing monocytes. These results highlight a novel role for androgen-driven peripheral IL-10-producing monocytes in the sexual dimorphism of pain resolution. These findings add to the growing concept that immune cells play a critical role in resolving pain and preventing the transition into chronic pain. Graphical abstract
Collapse
|
18
|
Jesus RLC, Araujo FA, Alves QL, Dourado KC, Silva DF. Targeting temperature-sensitive transient receptor potential channels in hypertension: far beyond the perception of hot and cold. J Hypertens 2023; 41:1351-1370. [PMID: 37334542 DOI: 10.1097/hjh.0000000000003487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Transient receptor potential (TRP) channels are nonselective cation channels and participate in various physiological roles. Thus, changes in TRP channel function or expression have been linked to several disorders. Among the many TRP channel subtypes, the TRP ankyrin type 1 (TRPA1), TRP melastatin type 8 (TRPM8), and TRP vanilloid type 1 (TRPV1) channels are temperature-sensitive and recognized as thermo-TRPs, which are expressed in the primary afferent nerve. Thermal stimuli are converted into neuronal activity. Several studies have described the expression of TRPA1, TRPM8, and TRPV1 in the cardiovascular system, where these channels can modulate physiological and pathological conditions, including hypertension. This review provides a complete understanding of the functional role of the opposing thermo-receptors TRPA1/TRPM8/TRPV1 in hypertension and a more comprehensive appreciation of TRPA1/TRPM8/TRPV1-dependent mechanisms involved in hypertension. These channels varied activation and inactivation have revealed a signaling pathway that may lead to innovative future treatment options for hypertension and correlated vascular diseases.
Collapse
Affiliation(s)
- Rafael Leonne C Jesus
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Salvador
| | - Fênix A Araujo
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation - FIOCRUZ, Bahia, Brazil
| | - Quiara L Alves
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Salvador
| | - Keina C Dourado
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Salvador
| | - Darizy F Silva
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Salvador
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation - FIOCRUZ, Bahia, Brazil
| |
Collapse
|
19
|
Silvestri R, Nicolì V, Gangadharannambiar P, Crea F, Bootman MD. Calcium signalling pathways in prostate cancer initiation and progression. Nat Rev Urol 2023; 20:524-543. [PMID: 36964408 DOI: 10.1038/s41585-023-00738-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2023] [Indexed: 03/26/2023]
Abstract
Cancer cells proliferate, differentiate and migrate by repurposing physiological signalling mechanisms. In particular, altered calcium signalling is emerging as one of the most widespread adaptations in cancer cells. Remodelling of calcium signalling promotes the development of several malignancies, including prostate cancer. Gene expression data from in vitro, in vivo and bioinformatics studies using patient samples and xenografts have shown considerable changes in the expression of various components of the calcium signalling toolkit during the development of prostate cancer. Moreover, preclinical and clinical evidence suggests that altered calcium signalling is a crucial component of the molecular re-programming that drives prostate cancer progression. Evidence points to calcium signalling re-modelling, commonly involving crosstalk between calcium and other cellular signalling pathways, underpinning the onset and temporal progression of this disease. Discrete alterations in calcium signalling have been implicated in hormone-sensitive, castration-resistant and aggressive variant forms of prostate cancer. Hence, modulation of calcium signals and downstream effector molecules is a plausible therapeutic strategy for both early and late stages of prostate cancer. Based on this premise, clinical trials have been undertaken to establish the feasibility of targeting calcium signalling specifically for prostate cancer.
Collapse
Affiliation(s)
| | - Vanessa Nicolì
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | | | - Francesco Crea
- Cancer Research Group, School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Martin D Bootman
- Cancer Research Group, School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK.
| |
Collapse
|
20
|
Van-Duyne G, Blair IA, Sprenger C, Moiseenkova-Bell V, Plymate S, Penning TM. The androgen receptor. VITAMINS AND HORMONES 2023; 123:439-481. [PMID: 37717994 DOI: 10.1016/bs.vh.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
The Androgen Receptor (AR) is a ligand (androgen) activated transcription factor and a member of the nuclear receptor (NR) superfamily. It is required for male sex hormone function. AR-FL (full-length) has the domain structure of NRs, an N-terminal domain (NTD) required for transactivation, a DNA-binding domain (DBD), a nuclear localization signal (NLS) and a ligand-binding domain (LBD). Paradoxes exist in that endogenous ligands testosterone (T) and 5α-dihydrotestosterone (DHT) have differential effects on male sexual development while binding to the same receptor and transcriptional specificity is achieved even though the androgen response elements (AREs) are identical to those seen for the progesterone, glucocorticoid and mineralocorticoid receptors. A high resolution 3-dimensional structure of AR-FL by either cryo-EM or X-ray crystallography has remained elusive largely due to the intrinsic disorder of the NTD. AR function is regulated by post-translational modification leading to a large number of proteoforms. The interaction of these proteoforms in multiprotein complexes with co-activators and co-repressors driven by interdomain coupling mediates the AR transcriptional output. The AR is a drug target for selective androgen receptor modulators (SARMS) that either have anabolic or androgenic effects. Protstate cancer is treated with androgen deprivation therapy or by the use of AR antagonists that bind to the LBD. Drug resistance occurs due to adaptive AR upregulation and the appearance of splice variants that lack the LBD and become constitutively active. Bipolar T treatment and NTD-antagonists could surmount these resistance mechanisms, respectively. These recent advances in AR signaling are described.
Collapse
Affiliation(s)
- Greg Van-Duyne
- Department of Biophysics & Biochemistry, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, United States
| | - Ian A Blair
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, United States
| | - Cynthia Sprenger
- Division of Gerontology & Geriatric Medicine, Department of Medicine, University of Washington and GRECC, Seattle, WA, United States
| | - Vera Moiseenkova-Bell
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, United States
| | - Stephen Plymate
- Division of Gerontology & Geriatric Medicine, Department of Medicine, University of Washington and GRECC, Seattle, WA, United States
| | - Trevor M Penning
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
21
|
Greenfield AM, Alba BK, Giersch GEW, Seeley AD. Sex differences in thermal sensitivity and perception: Implications for behavioral and autonomic thermoregulation. Physiol Behav 2023; 263:114126. [PMID: 36787810 DOI: 10.1016/j.physbeh.2023.114126] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/20/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023]
Abstract
Temperature sensitive receptors in the skin and deep body enable the detection of the external and internal environment, including the perception of thermal stimuli. Changes in heat balance require autonomic (e.g., sweating) and behavioral (e.g., seeking shade) thermoeffector initiation to maintain thermal homeostasis. Sex differences in body morphology can largely, but not entirely, account for divergent responses in thermoeffector and perceptual responses to environmental stress between men and women. Thus, it has been suggested that innate differences in thermosensation may exist between men and women. Our goal in this review is to summarize the existing literature that investigates localized and whole-body cold and heat exposure pertaining to sex differences in thermal sensitivity and perception, and the interplay between autonomic and behavioral thermoeffector responses. Overall, it appears that local differences in thermal sensitivity and perception are minimized, yet still apparent, when morphological characteristics are well-controlled. Sex differences in the early vasomotor response to environmental stress and subsequent changes in blood flow likely contribute to the heightened thermal awareness observed in women. However, the contribution of thermoreceptors to observed sex differences in thermal perception and thermoeffector function is unclear, as human studies investigating these questions have not been performed.
Collapse
Affiliation(s)
- Andrew M Greenfield
- Thermal and Mountain Medicine Division, US Army Research Institute of Environmental Medicine, Natick, MA, United States of America; Oak Ridge Institute for Science and Education, Belcamp, MD, United States of America.
| | - Billie K Alba
- Thermal and Mountain Medicine Division, US Army Research Institute of Environmental Medicine, Natick, MA, United States of America
| | - Gabrielle E W Giersch
- Thermal and Mountain Medicine Division, US Army Research Institute of Environmental Medicine, Natick, MA, United States of America
| | - Afton D Seeley
- Thermal and Mountain Medicine Division, US Army Research Institute of Environmental Medicine, Natick, MA, United States of America
| |
Collapse
|
22
|
Cohen CF, Roh J, Lee SH, Park CK, Berta T. Targeting Nociceptive Neurons and Transient Receptor Potential Channels for the Treatment of Migraine. Int J Mol Sci 2023; 24:ijms24097897. [PMID: 37175602 PMCID: PMC10177956 DOI: 10.3390/ijms24097897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Migraine is a neurovascular disorder that affects approximately 12% of the global population. While its exact causes are still being studied, researchers believe that nociceptive neurons in the trigeminal ganglia play a key role in the pain signals of migraine. These nociceptive neurons innervate the intracranial meninges and convey pain signals from the meninges to the thalamus. Targeting nociceptive neurons is considered promising due to their accessibility and distinct molecular profile, which includes the expression of several transient receptor potential (TRP) channels. These channels have been linked to various pain conditions, including migraine. This review discusses the role and mechanisms of nociceptive neurons in migraine, the challenges of current anti-migraine drugs, and the evidence for well-studied and emerging TRP channels, particularly TRPC4, as novel targets for migraine prevention and treatment.
Collapse
Affiliation(s)
- Cinder Faith Cohen
- Pain Research Center, Department of Anesthesiology, Medical Center, University of Cincinnati, Cincinnati, OH 45219, USA
- Neuroscience Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Jueun Roh
- Pain Research Center, Department of Anesthesiology, Medical Center, University of Cincinnati, Cincinnati, OH 45219, USA
- Department of Physiology, Gachon Pain Center, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
| | - Sang Hoon Lee
- Pain Research Center, Department of Anesthesiology, Medical Center, University of Cincinnati, Cincinnati, OH 45219, USA
- Neuroscience Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Chul-Kyu Park
- Department of Physiology, Gachon Pain Center, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, Medical Center, University of Cincinnati, Cincinnati, OH 45219, USA
| |
Collapse
|
23
|
Ye L, Zhu M, Ju J, Yang H. Effects of Dietary Cholesterol Regulation on Spermatogenesis of Gobiocypris rarus Rare Minnow. Int J Mol Sci 2023; 24:ijms24087492. [PMID: 37108655 PMCID: PMC10141657 DOI: 10.3390/ijms24087492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Cholesterol is an important component of cell membranes, and also a precursor for the synthesis of sex hormones, playing an important role in reproduction. However, few studies have focused on cholesterol and reproductive health. To investigate the toxic effects of different cholesterol levels on the spermatogenesis of rare minnows, we regulate the cholesterol content in fish by feeding them a high-cholesterol diet and cholesterol inhibitor pravastatin, and cholesterol levels, sex hormone (T and 11KT) levels, testis histology, sperm morphology and function, and the expression of genes related to sex hormone synthesis were investigated. The research findings indicate that increasing cholesterol levels significantly increases the liver weight and hepatic-somatic index, as well as the total cholesterol and free cholesterol levels in the testis, liver, and plasma of rare minnow, while inhibiting cholesterol has the opposite effect (p < 0.05). However, both increasing and decreasing cholesterol levels can suppress rare minnow testicular development, as evidenced by a decrease in testis weight, lowered gonadosomatic index, suppressed sex hormone levels, and reduced mature sperm count. Further exploration revealed that the expression of sex hormone synthesis-related genes, including star, cyp19a1a, and hsd11b2, was significantly affected (p < 0.05), which may be an important reason for the decrease in sex hormone synthesis and consequent inhibition of testicular development. At the same time, the fertilization ability of mature sperm in both treatment groups significantly decreased. Scanning electron microscopy and fluorescence polarization tests showed that reducing cholesterol levels significantly increased the rate of sperm head cell membrane damage, while both increasing and decreasing cholesterol levels led to a reduction in sperm cell membrane fluidity, which may be the main reason for the decrease in sperm fertilization ability. This study demonstrates that both increasing and decreasing the levels of cholesterol are detrimental to the fish spermatogenesis, providing fundamental information for the study of fish reproduction and also a reference for the causes of male reproductive dysfunction.
Collapse
Affiliation(s)
- Lv Ye
- College of Physical Education, Yangzhou University, Yangzhou 225009, China
| | - Mingzhen Zhu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Jian Ju
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Hui Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
24
|
Čonkaš J, Sabol M, Ozretić P. 'Toxic Masculinity': What Is Known about the Role of Androgen Receptors in Head and Neck Squamous Cell Carcinoma. Int J Mol Sci 2023; 24:3766. [PMID: 36835177 PMCID: PMC9965076 DOI: 10.3390/ijms24043766] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/03/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC), the most prevalent cancer in the head and neck region, develops from the mucosal epithelium of the upper aerodigestive tract. Its development directly correlates with alcohol and/or tobacco consumption and infection with human papillomavirus. Interestingly, the relative risk for HNSCC is up to five times higher in males, so it is considered that the endocrine microenvironment is another risk factor. A gender-specific risk for HNSCC suggests either the existence of specific risk factors that affect only males or that females have defensive hormonal and metabolic features. In this review, we summarized the current knowledge about the role of both nuclear and membrane androgen receptors (nAR and mARs, respectively) in HNSCC. As expected, the significance of nAR is much better known; it was shown that increased nAR expression was observed in HNSCC, while treatment with dihydrotestosterone increased proliferation, migration, and invasion of HNSCC cells. For only three out of five currently known mARs-TRPM8, CaV1.2, and OXER1-it was shown either their increased expression in various types of HNSCC or that their increased activity enhanced the migration and invasion of HNSCC cells. The primary treatments for HNSCC are surgery and radiotherapy, but targeted immunotherapies are on the rise. On the other hand, given the evidence of elevated nAR expression in HNSCC, this receptor represents a potential target for antiandrogen therapy. Moreover, there is still plenty of room for further examination of mARs' role in HNSCC diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
| | | | - Petar Ozretić
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| |
Collapse
|
25
|
Pereira ÉR, Pinheiro LCL, Francelino AL, Miqueloto CA, Guembarovski AFML, de Oliveira KB, Fuganti PE, de Syllos Cólus IM, Guembarovski RL. Tissue immunostaining of candidate prognostic proteins in metastatic and non-metastatic prostate cancer. J Cancer Res Clin Oncol 2023; 149:567-577. [PMID: 36008689 DOI: 10.1007/s00432-022-04274-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 08/06/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE Prostate cancer (PCa) lacks specific markers capable of distinguishing aggressive tumors from those with indolent behavior. Therefore, the aim of this study was to evaluate the immunostaining of candidate proteins (PTEN, AKT, TRPM8, and NKX3.1) through the immunohistochemistry technique (IHC) on patients with metastatic and non-metastatic PCa. METHODS Tissues from 60 patients were divided into three groups categorized according to prognostic parameters: better prognosis (n = 20), worse prognosis (n = 23), and metastatic (n = 17). Immunostaining was analyzed by a pathologist and staining classifications were considered according to signal intensity: (0) no staining, (+) weak, and (++ and +++) intermediate to strong. RESULTS AKT protein was associated (p = 0.012) and correlated (p = 0.014; Tau = - 0.288) with the prognostic groups. The immunostaining for TRPM8 (p = 0.010) and NKX3.1 (p = 0.003) proteins differed between malignant tumor and non-tumoral adjacent tissue as well as for proteins in cellular locations (nucleus and cytoplasm). TRPM8 was independently associated with the ISUP grade ≥ 4 (p = 0.024; OR = 8.373; 95% CI = 1.319-53.164). The NKX3.1 showed positive and predominantly strong immunostaining in all patients in both tumoral and non-tumoral adjacent tissues. All metastatic samples had positive immunostaining, with strong intensity for NKX3.1 (p = 0.021; Tau = - 0.302). In the non-metastatic group, this strong protein staining was not observed in any patients. CONCLUSION This study confirmed that NKX3.1 is highly specific for prostate tissue and indicated that NKX3.1, AKT, and TRPM8 may be candidate markers for prostate cancer prognosis.
Collapse
Affiliation(s)
- Érica Romão Pereira
- Laboratory of Mutagenesis and Oncogenetics, Department of General Biology, Londrina State University, Londrina, PR, Brazil
| | - Laís Capelasso Lucas Pinheiro
- Laboratory of Mutagenesis and Oncogenetics, Department of General Biology, Londrina State University, Londrina, PR, Brazil
| | - Amanda Letícia Francelino
- Laboratory of Mutagenesis and Oncogenetics, Department of General Biology, Londrina State University, Londrina, PR, Brazil
| | - Carlos Alberto Miqueloto
- Laboratory of Extracellular Matrix, Department of General Biology, Londrina State University, Londrina, PR, Brazil
| | | | - Karen Brajão de Oliveira
- Laboratory of Molecular Genetics and Immunology, Department of Pathological Science, Londrina State University, Londrina, PR, Brazil
| | | | - Ilce Mara de Syllos Cólus
- Laboratory of Mutagenesis and Oncogenetics, Department of General Biology, Londrina State University, Londrina, PR, Brazil
| | - Roberta Losi Guembarovski
- Laboratory of Mutagenesis and Oncogenetics, Department of General Biology, Londrina State University, Londrina, PR, Brazil.
| |
Collapse
|
26
|
Progress in the Structural Basis of thermoTRP Channel Polymodal Gating. Int J Mol Sci 2023; 24:ijms24010743. [PMID: 36614186 PMCID: PMC9821180 DOI: 10.3390/ijms24010743] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
The thermosensory transient receptor potential (thermoTRP) family of ion channels is constituted by several nonselective cation channels that are activated by physical and chemical stimuli functioning as paradigmatic polymodal receptors. Gating of these ion channels is achieved through changes in temperature, osmolarity, voltage, pH, pressure, and by natural or synthetic chemical compounds that directly bind to these proteins to regulate their activity. Given that thermoTRP channels integrate diverse physical and chemical stimuli, a thorough understanding of the molecular mechanisms underlying polymodal gating has been pursued, including the interplay between stimuli and differences between family members. Despite its complexity, recent advances in cryo-electron microscopy techniques are facilitating this endeavor by providing high-resolution structures of these channels in different conformational states induced by ligand binding or temperature that, along with structure-function and molecular dynamics, are starting to shed light on the underlying allosteric gating mechanisms. Because dysfunctional thermoTRP channels play a pivotal role in human diseases such as chronic pain, unveiling the intricacies of allosteric channel gating should facilitate the development of novel drug-based resolving therapies for these disorders.
Collapse
|
27
|
Fernández-Peña C, Reimúndez A, Viana F, Arce VM, Señarís R. Sex differences in thermoregulation in mammals: Implications for energy homeostasis. Front Endocrinol (Lausanne) 2023; 14:1093376. [PMID: 36967809 PMCID: PMC10030879 DOI: 10.3389/fendo.2023.1093376] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/06/2023] [Indexed: 03/10/2023] Open
Abstract
Thermal homeostasis is a fundamental process in mammals, which allows the maintenance of a constant internal body temperature to ensure an efficient function of cells despite changes in ambient temperature. Increasing evidence has revealed the great impact of thermoregulation on energy homeostasis. Homeothermy requires a fine regulation of food intake, heat production, conservation and dissipation and energy expenditure. A great interest on this field of research has re-emerged following the discovery of thermogenic brown adipose tissue and browning of white fat in adult humans, with a potential clinical relevance on obesity and metabolic comorbidities. However, most of our knowledge comes from male animal models or men, which introduces unwanted biases on the findings. In this review, we discuss how differences in sex-dependent characteristics (anthropometry, body composition, hormonal regulation, and other sexual factors) influence numerous aspects of thermal regulation, which impact on energy homeostasis. Individuals of both sexes should be used in the experimental paradigms, considering the ovarian cycles and sexual hormonal regulation as influential factors in these studies. Only by collecting data in both sexes on molecular, functional, and clinical aspects, we will be able to establish in a rigorous way the real impact of thermoregulation on energy homeostasis, opening new avenues in the understanding and treatment of obesity and metabolic associated diseases.
Collapse
Affiliation(s)
| | - Alfonso Reimúndez
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Félix Viana
- Institute of Neuroscience, University Miguel Hernández (UMH)-CSIC, Alicante, Spain
| | - Victor M. Arce
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- *Correspondence: Rosa Señarís, ; Victor M. Arce,
| | - Rosa Señarís
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- *Correspondence: Rosa Señarís, ; Victor M. Arce,
| |
Collapse
|
28
|
Cabañero D, Villalba-Riquelme E, Fernández-Ballester G, Fernández-Carvajal A, Ferrer-Montiel A. ThermoTRP channels in pain sexual dimorphism: new insights for drug intervention. Pharmacol Ther 2022; 240:108297. [PMID: 36202261 DOI: 10.1016/j.pharmthera.2022.108297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/25/2022] [Accepted: 09/29/2022] [Indexed: 11/30/2022]
Abstract
Chronic pain is a major burden for the society and remains more prevalent and severe in females. The presence of chronic pain is linked to persistent alterations in the peripheral and the central nervous system. One of the main types of peripheral pain transducers are the transient receptor potential channels (TRP), also known as thermoTRP channels, which intervene in the perception of hot and cold external stimuli. These channels, and especially TRPV1, TRPA1 and TRPM8, have been subjected to profound investigation because of their role as thermosensors and also because of their implication in acute and chronic pain. Surprisingly, their sensitivity to endogenous signaling has been far less studied. Cumulative evidence suggests that the function of these channels may be differently modulated in males and females, in part through sexual hormones, and this could constitute a significant contributor to the sex differences in chronic pain. Here, we review the exciting advances in thermoTRP pharmacology for males and females in two paradigmatic types of chronic pain with a strong peripheral component: chronic migraine and chemotherapy-induced peripheral neuropathy (CIPN). The possibilities of peripheral druggability offered by these channels and the differential exploitation for men and women represent a development opportunity that will lead to a significant increment of the armamentarium of analgesic medicines for personalized chronic pain treatment.
Collapse
Affiliation(s)
- David Cabañero
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, 03202 Elche, Spain
| | - Eva Villalba-Riquelme
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, 03202 Elche, Spain
| | - Gregorio Fernández-Ballester
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, 03202 Elche, Spain
| | - Asia Fernández-Carvajal
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, 03202 Elche, Spain
| | - Antonio Ferrer-Montiel
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, 03202 Elche, Spain.
| |
Collapse
|
29
|
Alarcón-Alarcón D, Cabañero D, de Andrés-López J, Nikolaeva-Koleva M, Giorgi S, Fernández-Ballester G, Fernández-Carvajal A, Ferrer-Montiel A. TRPM8 contributes to sex dimorphism by promoting recovery of normal sensitivity in a mouse model of chronic migraine. Nat Commun 2022; 13:6304. [PMID: 36272975 PMCID: PMC9588003 DOI: 10.1038/s41467-022-33835-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 09/30/2022] [Indexed: 12/25/2022] Open
Abstract
TRPA1 and TRPM8 are transient receptor potential channels expressed in trigeminal neurons that are related to pathophysiology in migraine models. Here we use a mouse model of nitroglycerine-induced chronic migraine that displays a sexually dimorphic phenotype, characterized by mechanical hypersensitivity that develops in males and females, and is persistent up to day 20 in female mice, but disappears by day 18 in male mice. TRPA1 is required for development of hypersensitivity in males and females, whereas TRPM8 contributes to the faster recovery from hypersensitivity in males. TRPM8-mediated antinociception effects required the presence of endogenous testosterone in males. Administration of exogenous testosterone to females and orchidectomized males led to recovery from hypersensitivity. Calcium imaging and electrophysiological recordings in in vitro systems confirmed testosterone activity on murine and human TRPM8, independent of androgen receptor expression. Our findings suggest a protective function of TRPM8 in shortening the time frame of hypersensitivity in a mouse model of migraine.
Collapse
Affiliation(s)
- David Alarcón-Alarcón
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
| | - David Cabañero
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain.
| | - Jorge de Andrés-López
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
| | - Magdalena Nikolaeva-Koleva
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
| | - Simona Giorgi
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
| | - Gregorio Fernández-Ballester
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
| | - Asia Fernández-Carvajal
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain.
| | - Antonio Ferrer-Montiel
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain.
| |
Collapse
|
30
|
Rosenbaum T, Morales-Lázaro SL, Islas LD. TRP channels: a journey towards a molecular understanding of pain. Nat Rev Neurosci 2022; 23:596-610. [PMID: 35831443 DOI: 10.1038/s41583-022-00611-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2022] [Indexed: 12/18/2022]
Abstract
The perception of nociceptive signals, which are translated into pain, plays a fundamental role in the survival of organisms. Because pain is linked to a negative sensation, animals learn to avoid noxious signals. These signals are detected by receptors, which include some members of the transient receptor potential (TRP) family of ion channels that act as transducers of exogenous and endogenous noxious cues. These proteins have been in the focus of the field of physiology for several years, and much knowledge of how they regulate the function of the cell types and organs where they are expressed has been acquired. The last decade has been especially exciting because the 'resolution revolution' has allowed us to learn the molecular intimacies of TRP channels using cryogenic electron microscopy. These findings, in combination with functional studies, have provided insights into the role played by these channels in the generation and maintenance of pain.
Collapse
Affiliation(s)
- Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, UNAM, Mexico City, Mexico.
| | - Sara L Morales-Lázaro
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, UNAM, Mexico City, Mexico
| | - León D Islas
- Departamento de Fisiología, Facultad de Medicina, UNAM, Mexico City, Mexico
| |
Collapse
|
31
|
Mauvais-Jarvis F, Lange CA, Levin ER. Membrane-Initiated Estrogen, Androgen, and Progesterone Receptor Signaling in Health and Disease. Endocr Rev 2022; 43:720-742. [PMID: 34791092 PMCID: PMC9277649 DOI: 10.1210/endrev/bnab041] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Indexed: 12/15/2022]
Abstract
Rapid effects of steroid hormones were discovered in the early 1950s, but the subject was dominated in the 1970s by discoveries of estradiol and progesterone stimulating protein synthesis. This led to the paradigm that steroid hormones regulate growth, differentiation, and metabolism via binding a receptor in the nucleus. It took 30 years to appreciate not only that some cellular functions arise solely from membrane-localized steroid receptor (SR) actions, but that rapid sex steroid signaling from membrane-localized SRs is a prerequisite for the phosphorylation, nuclear import, and potentiation of the transcriptional activity of nuclear SR counterparts. Here, we provide a review and update on the current state of knowledge of membrane-initiated estrogen (ER), androgen (AR) and progesterone (PR) receptor signaling, the mechanisms of membrane-associated SR potentiation of their nuclear SR homologues, and the importance of this membrane-nuclear SR collaboration in physiology and disease. We also highlight potential clinical implications of pathway-selective modulation of membrane-associated SR.
Collapse
Affiliation(s)
- Franck Mauvais-Jarvis
- Department of Medicine, Section of Endocrinology and Metabolism, Tulane University School of Medicine, New Orleans, LA, 70112, USA.,Tulane Center of Excellence in Sex-Based Biology & Medicine, New Orleans, LA, 70112, USA.,Southeast Louisiana Veterans Affairs Medical Center, New Orleans, LA, 70119, USA
| | - Carol A Lange
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.,Department of Medicine (Division of Hematology, Oncology, and Transplantation), University of Minnesota, Minneapolis, MN 55455, USA.,Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ellis R Levin
- Division of Endocrinology, Department of Medicine, University of California, Irvine, Irvine, CA, 92697, USA.,Department of Veterans Affairs Medical Center, Long Beach, Long Beach, CA, 90822, USA
| |
Collapse
|
32
|
Hornsby E, King HW, Peiris M, Buccafusca R, Lee WYJ, Wing ES, Blackshaw LA, Lindsay JO, Stagg AJ. The cation channel TRPM8 influences the differentiation and function of human monocytes. J Leukoc Biol 2022; 112:365-381. [PMID: 35233801 PMCID: PMC9543907 DOI: 10.1002/jlb.1hi0421-181r] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 01/20/2022] [Indexed: 12/24/2022] Open
Abstract
Monocytes are mononuclear phagocytes that can differentiate to a variety of cell fates under the influence of their microenvironment and hardwired commitment. We found that inhibition of TRPM8 in human blood CD14+ monocytes during a critical 3‐h window at the beginning of their differentiation into macrophages led to enhanced survival and LPS‐driven TNFα production after 24 h. TRPM8 antagonism also promoted LPS‐driven TNFα production in CD14+ monocytes derived from the intestinal mucosa. Macrophages that had been derived for 6 days under blockade of TRPM8 had impaired phagocytic capacity and were transcriptionally distinct. Most of the affected genes were altered in a way that opposed normal monocyte to macrophage differentiation indicating that TRPM8 activity promotes aspects of this differentiation programme. Thus, we reveal a novel role for TRPM8 in regulating human CD14+ monocyte fate and function.
Collapse
Affiliation(s)
- Eve Hornsby
- Centre for Immunobiology & Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Hamish W King
- Centre for Immunobiology & Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Madusha Peiris
- Centre for Neuroscience & Trauma, Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Roberto Buccafusca
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London, UK
| | - Wing-Yiu Jason Lee
- Centre for Immunobiology & Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Elinor S Wing
- Centre for Immunobiology & Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - L Ashley Blackshaw
- Centre for Neuroscience & Trauma, Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - James O Lindsay
- Centre for Immunobiology & Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK.,Department of Gastroenterology, Barts Health NHS Trust, The Royal London Hospital, Whitechapel, London, UK
| | - Andrew J Stagg
- Centre for Immunobiology & Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
33
|
Martín-Escura C, Medina-Peris A, Spear LA, de la Torre Martínez R, Olivos-Oré LA, Barahona MV, González-Rodríguez S, Fernández-Ballester G, Fernández-Carvajal A, Artalejo AR, Ferrer-Montiel A, González-Muñiz R. β-Lactam TRPM8 Antagonist RGM8-51 Displays Antinociceptive Activity in Different Animal Models. Int J Mol Sci 2022; 23:ijms23052692. [PMID: 35269831 PMCID: PMC8910920 DOI: 10.3390/ijms23052692] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 02/05/2023] Open
Abstract
Transient receptor potential melastatin subtype 8 (TRPM8) is a cation channel extensively expressed in sensory neurons and implicated in different painful states. However, the effectiveness of TRPM8 modulators for pain relief is still a matter of discussion, since structurally diverse modulators lead to different results, depending on the animal pain model. In this work, we described the antinociceptive activity of a β–lactam derivative, RGM8-51, showing good TRPM8 antagonist activity, and selectivity against related thermoTRP channels and other pain-mediating receptors. In primary cultures of rat dorsal root ganglion (DRG) neurons, RGM8-51 potently reduced menthol-evoked neuronal firing without affecting the major ion conductances responsible for action potential generation. This compound has in vivo antinociceptive activity in response to cold, in a mouse model of oxaliplatin-induced peripheral neuropathy. In addition, it reduces cold, mechanical and heat hypersensitivity in a rat model of neuropathic pain arising after chronic constriction of the sciatic nerve. Furthermore, RGM8-51 exhibits mechanical hypersensitivity-relieving activity, in a mouse model of NTG-induced hyperesthesia. Taken together, these preclinical results substantiate that this TRPM8 antagonist is a promising pharmacological tool to study TRPM8-related diseases.
Collapse
Affiliation(s)
- Cristina Martín-Escura
- Instituto de Química Médica (IQM-CSIC), 28006 Madrid, Spain; (C.M.-E.); (L.A.S.)
- Alodia Farmacéutica SL, 28108 Alcobendas, Spain
| | - Alicia Medina-Peris
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
| | - Luke A. Spear
- Instituto de Química Médica (IQM-CSIC), 28006 Madrid, Spain; (C.M.-E.); (L.A.S.)
| | - Roberto de la Torre Martínez
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
| | - Luis A. Olivos-Oré
- Departamento de Farmacología y Toxicología, Facultad de Veterinaria, Universidad Complutense de Madrid, 28040 Madrid, Spain; (L.A.O.-O.); (M.V.B.); (A.R.A.)
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - María Victoria Barahona
- Departamento de Farmacología y Toxicología, Facultad de Veterinaria, Universidad Complutense de Madrid, 28040 Madrid, Spain; (L.A.O.-O.); (M.V.B.); (A.R.A.)
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Sara González-Rodríguez
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
| | - Gregorio Fernández-Ballester
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
| | - Asia Fernández-Carvajal
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
- Correspondence: (A.F.-C.); (R.G.-M.); Tel.: +00-34-258-74-34 (R.G.-M.)
| | - Antonio R. Artalejo
- Departamento de Farmacología y Toxicología, Facultad de Veterinaria, Universidad Complutense de Madrid, 28040 Madrid, Spain; (L.A.O.-O.); (M.V.B.); (A.R.A.)
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Antonio Ferrer-Montiel
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
| | - Rosario González-Muñiz
- Instituto de Química Médica (IQM-CSIC), 28006 Madrid, Spain; (C.M.-E.); (L.A.S.)
- Correspondence: (A.F.-C.); (R.G.-M.); Tel.: +00-34-258-74-34 (R.G.-M.)
| |
Collapse
|
34
|
Cheng WWL, Arcario MJ, Petroff JT. Druggable Lipid Binding Sites in Pentameric Ligand-Gated Ion Channels and Transient Receptor Potential Channels. Front Physiol 2022; 12:798102. [PMID: 35069257 PMCID: PMC8777383 DOI: 10.3389/fphys.2021.798102] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/02/2021] [Indexed: 12/17/2022] Open
Abstract
Lipids modulate the function of many ion channels, possibly through direct lipid-protein interactions. The recent outpouring of ion channel structures by cryo-EM has revealed many lipid binding sites. Whether these sites mediate lipid modulation of ion channel function is not firmly established in most cases. However, it is intriguing that many of these lipid binding sites are also known sites for other allosteric modulators or drugs, supporting the notion that lipids act as endogenous allosteric modulators through these sites. Here, we review such lipid-drug binding sites, focusing on pentameric ligand-gated ion channels and transient receptor potential channels. Notable examples include sites for phospholipids and sterols that are shared by anesthetics and vanilloids. We discuss some implications of lipid binding at these sites including the possibility that lipids can alter drug potency or that understanding protein-lipid interactions can guide drug design. Structures are only the first step toward understanding the mechanism of lipid modulation at these sites. Looking forward, we identify knowledge gaps in the field and approaches to address them. These include defining the effects of lipids on channel function in reconstituted systems using asymmetric membranes and measuring lipid binding affinities at specific sites using native mass spectrometry, fluorescence binding assays, and computational approaches.
Collapse
Affiliation(s)
- Wayland W L Cheng
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States
| | - Mark J Arcario
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States
| | - John T Petroff
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
35
|
Persoons E, Kerselaers S, Voets T, Vriens J, Held K. Partial Agonistic Actions of Sex Hormone Steroids on TRPM3 Function. Int J Mol Sci 2021; 22:13652. [PMID: 34948452 PMCID: PMC8708174 DOI: 10.3390/ijms222413652] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 11/23/2022] Open
Abstract
Sex hormone steroidal drugs were reported to have modulating actions on the ion channel TRPM3. Pregnenolone sulphate (PS) presents the most potent known endogenous chemical agonist of TRPM3 and affects several gating modes of the channel. These includes a synergistic action of PS and high temperatures on channel opening and the PS-induced opening of a noncanonical pore in the presence of other TRPM3 modulators. Moreover, human TRPM3 variants associated with neurodevelopmental disease exhibit an increased sensitivity for PS. However, other steroidal sex hormones were reported to influence TRPM3 functions with activating or inhibiting capacity. Here, we aimed to answer how DHEAS, estradiol, progesterone and testosterone act on the various modes of TRPM3 function in the wild-type channel and two-channel variants associated with human disease. By means of calcium imaging and whole-cell patch clamp experiments, we revealed that all four drugs are weak TRPM3 agonists that share a common steroidal interaction site. Furthermore, they exhibit increased activity on TRPM3 at physiological temperatures and in channels that carry disease-associated mutations. Finally, all steroids are able to open the noncanonical pore in wild-type and DHEAS also in mutant TRPM3. Collectively, our data provide new valuable insights in TRPM3 gating, structure-function relationships and ligand sensitivity.
Collapse
Affiliation(s)
- Eleonora Persoons
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 Box 611, 3000 Leuven, Belgium; (E.P.); (K.H.)
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49 Box 802, 3000 Leuven, Belgium; (S.K.); (T.V.)
| | - Sara Kerselaers
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49 Box 802, 3000 Leuven, Belgium; (S.K.); (T.V.)
| | - Thomas Voets
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49 Box 802, 3000 Leuven, Belgium; (S.K.); (T.V.)
| | - Joris Vriens
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 Box 611, 3000 Leuven, Belgium; (E.P.); (K.H.)
| | - Katharina Held
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 Box 611, 3000 Leuven, Belgium; (E.P.); (K.H.)
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49 Box 802, 3000 Leuven, Belgium; (S.K.); (T.V.)
| |
Collapse
|
36
|
Design, synthesis and biological evaluation of new thiazole scaffolds as potential TRPM8 antagonists. Bioorg Med Chem Lett 2021; 52:128392. [PMID: 34606996 DOI: 10.1016/j.bmcl.2021.128392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 11/21/2022]
Abstract
The preliminary results on the development of a viable methodology for the further functionalization of 4-hydroxythiazole derivatives to afford target TRPM8 antagonists are reported. The combined Sonogashira coupling/annulation reactions of the ethyl 2-(3-fluorophenyl)-4-tifluoromethylsulfonyloxy-1,3-thiazole-5-carboxylate have been applied to the synthesis of analogues of the selective blocker of TRPM8 DFL23448. Among all the synthetised derivatives, the most promising compound resulted to be active as TRPM8 blocker (IC50 = 4.06 µM), showing an excellent metabolic stability and no cytotoxic effects. Finally, in silico characterisation of the derivatives showed no violation of the drug-likeness rules.
Collapse
|
37
|
Molecular Characterization of Membrane Steroid Receptors in Hormone-Sensitive Cancers. Cells 2021; 10:cells10112999. [PMID: 34831222 PMCID: PMC8616056 DOI: 10.3390/cells10112999] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 12/20/2022] Open
Abstract
Cancer is one of the most common causes of death worldwide, and its development is a result of the complex interaction of genetic factors, environmental cues, and aging. Hormone-sensitive cancers depend on the action of one or more hormones for their development and progression. Sex steroids and corticosteroids can regulate different physiological functions, including metabolism, growth, and proliferation, through their interaction with specific nuclear receptors, that can transcriptionally regulate target genes via their genomic actions. Therefore, interference with hormones’ activities, e.g., deregulation of their production and downstream pathways or the exposition to exogenous hormone-active substances such as endocrine-disrupting chemicals (EDCs), can affect the regulation of their correlated pathways and trigger the neoplastic transformation. Although nuclear receptors account for most hormone-related biologic effects and their slow genomic responses are well-studied, less-known membrane receptors are emerging for their ability to mediate steroid hormones effects through the activation of rapid non-genomic responses also involved in the development of hormone-sensitive cancers. This review aims to collect pre-clinical and clinical data on these extranuclear receptors not only to draw attention to their emerging role in cancer development and progression but also to highlight their dual role as tumor microenvironment players and potential candidate drug targets.
Collapse
|
38
|
Abstract
Steroid hormones bind receptors in the cell nucleus and in the cell membrane. The most widely studied class of steroid hormone receptors are the nuclear receptors, named for their function as ligand-dependent transcription factors in the cell nucleus. Nuclear receptors, such as estrogen receptor alpha, can also be anchored to the plasma membrane, where they respond to steroids by activating signaling pathways independent of their function as transcription factors. Steroids can also bind integral membrane proteins, such as the G protein-coupled estrogen receptor. Membrane estrogen and progestin receptors have been cloned and characterized in vitro and influence the development and function of many organ systems. Membrane androgen receptors were cloned and characterized in vitro, but their function as androgen receptors in vivo is unresolved. We review the identity and function of membrane proteins that bind estrogens, progestins, and androgens. We discuss evidence that membrane glucocorticoid and mineralocorticoid receptors exist, and whether glucocorticoid and mineralocorticoid nuclear receptors act at the cell membrane. In many cases, integral membrane steroid receptors act independently of nuclear steroid receptors, even though they may share a ligand.
Collapse
Affiliation(s)
- Lindsey S Treviño
- Department of Population Sciences, Division of Health Equities, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Daniel A Gorelick
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence: Daniel A Gorelick, PhD, One Baylor Plaza, Alkek Building N1317.07, Houston, TX, 77030-3411, USA.
| |
Collapse
|
39
|
Abstract
Transient receptor potential melastatin 8 (TRPM8) channels play a central role in the detection of environmental cold temperatures in the somatosensory system. TRPM8 is found in a subset of unmyelinated (C-type) afferents located in the dorsal root (DRG) and trigeminal ganglion (TG). Cold hypersensitivity is a common symptom of neuropathic pain conditions caused by cancer therapy, spinal cord injury, viral infection, multiple sclerosis, diabetes, or withdrawal symptoms associated with chronic morphine treatment. Therefore, TRPM8 has received great attention as a therapeutic target. However, as the activity of TRPM8 is unique in sensing cool temperature as well as warming, it is critical to understand the signaling transduction pathways that control modality-specific activity of TRPM8 in healthy versus pathological settings. This review summarizes recent advances in our understanding of the mechanisms involved in the regulation of the TRPM8 activity.
Collapse
Affiliation(s)
- Mircea Iftinca
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary , Calgary, Alberta, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary , Calgary, Alberta, Canada
| |
Collapse
|
40
|
Huang Y, Li S, Jia Z, Li S, He W, Zhou C, Zhang R, Xu R, Sun B, Ali DW, Michalak M, Chen XZ, Tang J. TRIM4 interacts with TRPM8 and regulates its channel function through K423-mediated ubiquitination. J Cell Physiol 2021; 236:2934-2949. [PMID: 33037615 DOI: 10.1002/jcp.30065] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/16/2022]
Abstract
Transient receptor potential melastatin member 8 (TRPM8), a Ca2+ -permeable nonselective cation channel activated by cold and cooling agents, mediates allodynia. Dysfunction or abnormal expression of TRPM8 has been found in several human cancers. The role of ubiquitination in the regulation of TRPM8 function remains poorly understood. Here, we identified the ubiquitin (Ub)-ligase E3, tripartite motif-containing 4 (TRIM4), as a novel interaction partner of TRPM8 and confirmed that the TRIM4-TRPM8 interaction was mediated through the SPRY domain of TRIM4. Patch-clamp assays showed that TRIM4 negatively regulates TRPM8-mediated currents in HEK293 cells. Moreover, TRIM4 reduced the expression of TRPM8 on the cell surface by promoting the K63-linked ubiquitination of TRPM8. Further analyses revealed that the TRPM8 N-terminal lysine residue at 423 was the major ubiquitination site that mediates its functional regulation by TRIM4. A Ub-activating enzyme E1, Ub-like modifier-activating enzyme 1 (UBA1), was also found to interact with TRPM8, thereby regulating its channel function and ubiquitination. In addition, knockdown of UBA1 impaired the regulation of TRPM8 ubiquitination and function by TRIM4. Thus, this study demonstrates that TRIM4 downregulates TRPM8 via K423-mediated TRPM8 ubiquitination and requires UBA1 to regulate TRPM8.
Collapse
Affiliation(s)
- Yuan Huang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Shunyao Li
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Zhenhua Jia
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Shi Li
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Wenzao He
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Cefan Zhou
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Rui Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Rui Xu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Bo Sun
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Declan William Ali
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry of Alberta, Edmonton, Alberta, Canada
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| |
Collapse
|
41
|
Sanders OD, Rajagopal JA, Rajagopal L. Menthol to Induce Non-shivering Thermogenesis via TRPM8/PKA Signaling for Treatment of Obesity. J Obes Metab Syndr 2021; 30:4-11. [PMID: 33071240 PMCID: PMC8017329 DOI: 10.7570/jomes20038] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/25/2020] [Accepted: 06/12/2020] [Indexed: 12/25/2022] Open
Abstract
Increasing basal energy expenditure via uncoupling protein 1 (UCP1)-dependent non-shivering thermogenesis is an attractive therapeutic strategy for treatment of obesity. Transient receptor potential melastatin 8 (TRPM8) channel activation by cold and cold mimetics induces UCP1 transcription and prevents obesity in animals, but the clinical relevance of this relationship remains incompletely understood. A review of TRPM8 channel agonism for treatment of obesity focusing on menthol was undertaken. Adipocyte TRPM8 activation results in Ca2+ influx and protein kinase A (PKA) activation, which induces mitochondrial elongation, mitochondrial localization to lipid droplets, lipolysis, β-oxidation, and UCP1 expression. Ca2+-induced mitochondrial reactive oxygen species activate UCP1. In animals, TRPM8 agonism increases basal metabolic rate, non-shivering thermogenesis, oxygen consumption, exercise endurance, and fatty acid oxidation and decreases abdominal fat percentage. Menthol prevents high-fat diet-induced obesity, glucose intolerance, insulin resistance, and liver triacylglycerol accumulation. Hypothalamic TRPM8 activation releases glucagon, which activates PKA and promotes catabolism. TRPM8 polymorphisms are associated with obesity. In humans, oral menthol and other TRPM8 agonists have little effect. However, topical menthol appears to increase core body temperature and metabolic rate. A randomized clinical control trial of topical menthol in obese patients is warranted.
Collapse
Affiliation(s)
| | | | - Lekshmy Rajagopal
- Oto-Rhino-Laryngology, College of Physicians and Surgeons, Mumbai, India
| |
Collapse
|
42
|
Abstract
Incorporation of ion channels in planar lipid bilayers allows detecting and measuring ion channel activity in a well-controlled system. This technique provides critical information about ion channel kinetics, ion selectivity, gating mechanism, open probability, unitary conductance, subconductance states, voltage dependence, and burst opening events, particularly at the single molecule level. Planar lipid bilayers provide a unique controllable environment that enables maintaining specific regulatory components, including lipids, ligands, inhibitors, particular ions, and proteins, as well as the temperature that can modulate activity of many ion channels. Thus, this system provides explicit details about ion channel gating mechanism and enables identifying its particular regulatory molecules or components. This chapter will describe the planar lipid bilayer method using the example of a transient receptor potential (TRP) ion channel family member. The planar lipid bilayer electrophysiological approach has proven to be useful in studying intrinsic properties of TRP channels. This method is particularly valuable for our understanding of intrinsic temperature sensitivity of thermoreceptors such as TRP channels and direct effects of TRP channels agonists, antagonists, co-factors, and other modifiers.
Collapse
Affiliation(s)
- Eleonora Zakharian
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, United States.
| |
Collapse
|
43
|
D’Arrigo G, Gianquinto E, Rossetti G, Cruciani G, Lorenzetti S, Spyrakis F. Binding of Androgen- and Estrogen-Like Flavonoids to Their Cognate (Non)Nuclear Receptors: A Comparison by Computational Prediction. Molecules 2021; 26:1613. [PMID: 33799482 PMCID: PMC8001607 DOI: 10.3390/molecules26061613] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/02/2021] [Accepted: 03/10/2021] [Indexed: 12/24/2022] Open
Abstract
Flavonoids are plant bioactives that are recognized as hormone-like polyphenols because of their similarity to the endogenous sex steroids 17β-estradiol and testosterone, and to their estrogen- and androgen-like activity. Most efforts to verify flavonoid binding to nuclear receptors (NRs) and explain their action have been focused on ERα, while less attention has been paid to other nuclear and non-nuclear membrane androgen and estrogen receptors. Here, we investigate six flavonoids (apigenin, genistein, luteolin, naringenin, quercetin, and resveratrol) that are widely present in fruits and vegetables, and often used as replacement therapy in menopause. We performed comparative computational docking simulations to predict their capability of binding nuclear receptors ERα, ERβ, ERRβ, ERRγ, androgen receptor (AR), and its variant ART877A and membrane receptors for androgens, i.e., ZIP9, GPRC6A, OXER1, TRPM8, and estrogens, i.e., G Protein-Coupled Estrogen Receptor (GPER). In agreement with data reported in literature, our results suggest that these flavonoids show a relevant degree of complementarity with both estrogen and androgen NR binding sites, likely triggering genomic-mediated effects. It is noteworthy that reliable protein-ligand complexes and estimated interaction energies were also obtained for some suggested estrogen and androgen membrane receptors, indicating that flavonoids could also exert non-genomic actions. Further investigations are needed to clarify flavonoid multiple genomic and non-genomic effects. Caution in their administration could be necessary, until the safe assumption of these natural molecules that are largely present in food is assured.
Collapse
Affiliation(s)
- Giulia D’Arrigo
- Department of Drug Science and Technology, University of Turin, Via Giuria 9, 10125 Turin, Italy; (G.D.); (E.G.)
| | - Eleonora Gianquinto
- Department of Drug Science and Technology, University of Turin, Via Giuria 9, 10125 Turin, Italy; (G.D.); (E.G.)
| | - Giulia Rossetti
- Institute for Neuroscience and Medicine (INM-9) and Institute for Advanced Simulations (IAS-5) “Computational Biomedicine”, Forschungszentrum Jülich, 52425 Jülich, Germany
- Jülich Supercomputing Center (JSC), Forschungszentrum Jülich, 52425 Jülich, Germany
- Department of Neurology, RWTH, Aachen University, 52074 Aachen, Germany;
| | - Gabriele Cruciani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy;
| | - Stefano Lorenzetti
- Istituto Superiore di Sanità (ISS), Department of Food Safety, Nutrition and Veterinary Public Health, Viale Regina Elena 299, 00161 Rome, Italy
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, Via Giuria 9, 10125 Turin, Italy; (G.D.); (E.G.)
| |
Collapse
|
44
|
Phenylalanine-Derived β-Lactam TRPM8 Modulators. Configuration Effect on the Antagonist Activity. Int J Mol Sci 2021; 22:ijms22052370. [PMID: 33673444 PMCID: PMC7956626 DOI: 10.3390/ijms22052370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
Transient receptor potential cation channel subfamily M member 8 (TRPM8) is a Ca2+ non-selective ion channel implicated in a variety of pathological conditions, including cancer, inflammatory and neuropathic pain. In previous works we identified a family of chiral, highly hydrophobic β–lactam derivatives, and began to intuit a possible effect of the stereogenic centers on the antagonist activity. To investigate the influence of configuration on the TRPM8 antagonist properties, here we prepare and characterize four possible diastereoisomeric derivatives of 4-benzyl-1-[(3′-phenyl-2′-dibenzylamino)prop-1′-yl]-4-benzyloxycarbonyl-3-methyl-2-oxoazetidine. In microfluorography assays, all isomers were able to reduce the menthol-induced cell Ca2+ entry to larger or lesser extent. Potency follows the order 3R,4R,2′R > 3S,4S,2′R ≅ 3R,4R,2′S > 3S,4S,2′S, with the most potent diastereoisomer showing a half inhibitory concentration (IC50) in the low nanomolar range, confirmed by Patch-Clamp electrophysiology experiments. All four compounds display high receptor selectivity against other members of the TRP family. Furthermore, in primary cultures of rat dorsal root ganglion (DRG) neurons, the most potent diastereoisomers do not produce any alteration in neuronal excitability, indicating their high specificity for TRPM8 channels. Docking studies positioned these β-lactams at different subsites by the pore zone, suggesting a different mechanism than the known N-(3-aminopropyl)-2-[(3-methylphenyl)methoxy]-N-(2-thienylmethyl)-benzamide (AMTB) antagonist.
Collapse
|
45
|
Testosterone-loaded GM1 micelles targeted to the intracellular androgen receptor for the specific induction of genomic androgen signaling. Int J Pharm 2020; 591:119985. [DOI: 10.1016/j.ijpharm.2020.119985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/08/2020] [Accepted: 10/11/2020] [Indexed: 02/07/2023]
|
46
|
Martínez-Rojas VA, Jiménez-Garduño AM, Michelatti D, Tosatto L, Marchioretto M, Arosio D, Basso M, Pennuto M, Musio C. ClC-2-like Chloride Current Alterations in a Cell Model of Spinal and Bulbar Muscular Atrophy, a Polyglutamine Disease. J Mol Neurosci 2020; 71:662-674. [PMID: 32856205 DOI: 10.1007/s12031-020-01687-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/14/2020] [Indexed: 12/21/2022]
Abstract
Spinal and bulbar muscular atrophy (SBMA) is a neuromuscular disease caused by expansions of a polyglutamine (polyQ) tract in the androgen receptor (AR) gene. SBMA is associated with the progressive loss of lower motor neurons, together with muscle weakness and atrophy. PolyQ-AR is converted to a toxic species upon binding to its natural ligands, testosterone, and dihydrotestosterone (DHT). Our previous patch-clamp studies on a motor neuron-derived cell model of SBMA showed alterations in voltage-gated ion currents. Here, we identified and characterized chloride currents most likely belonging to the chloride channel-2 (ClC-2) subfamily, which showed significantly increased amplitudes in the SBMA cells. The treatment with the pituitary adenylyl cyclase-activating polypeptide (PACAP), a neuropeptide with a proven protective effect in a mouse model of SBMA, recovered chloride channel current alterations in SBMA cells. These observations suggest that the CIC-2 currents are affected in SBMA, an alteration that may contribute and potentially determine the pathophysiology of the disease.
Collapse
Affiliation(s)
- Vladimir A Martínez-Rojas
- Institute of Biophysics (IBF), Trento Unit, National Research Council (CNR) & LabSSAH, Bruno Kessler Foundation (FBK), Trento, Italy
| | - Aura M Jiménez-Garduño
- Institute of Biophysics (IBF), Trento Unit, National Research Council (CNR) & LabSSAH, Bruno Kessler Foundation (FBK), Trento, Italy.,Departamento de Ciencias de la Salud, Escuela de Ciencias, Universidad de las Américas Puebla (UDLAP), San Andrés Cholula, Puebla, Mexico
| | - Daniela Michelatti
- Institute of Biophysics (IBF), Trento Unit, National Research Council (CNR) & LabSSAH, Bruno Kessler Foundation (FBK), Trento, Italy.,CIBIO Department, Laboratory of Chromatin Biology and Epigenetics, University of Trento, Trento, Italy
| | - Laura Tosatto
- Institute of Biophysics (IBF), Trento Unit, National Research Council (CNR) & LabSSAH, Bruno Kessler Foundation (FBK), Trento, Italy
| | - Marta Marchioretto
- Institute of Biophysics (IBF), Trento Unit, National Research Council (CNR) & LabSSAH, Bruno Kessler Foundation (FBK), Trento, Italy
| | - Daniele Arosio
- Institute of Biophysics (IBF), Trento Unit, National Research Council (CNR) & LabSSAH, Bruno Kessler Foundation (FBK), Trento, Italy
| | - Manuela Basso
- CIBIO Department, Laboratory of Transcriptional Neurobiology, University of Trento, Trento, Italy
| | - Maria Pennuto
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Padova Neuroscience Center (PNC), University of Padova, Padova, Italy.,Veneto Institute of Molecular Medicine, Padova, Italy
| | - Carlo Musio
- Institute of Biophysics (IBF), Trento Unit, National Research Council (CNR) & LabSSAH, Bruno Kessler Foundation (FBK), Trento, Italy.
| |
Collapse
|
47
|
Xu L, Han Y, Chen X, Aierken A, Wen H, Zheng W, Wang H, Lu X, Zhao Z, Ma C, Liang P, Yang W, Yang S, Yang F. Molecular mechanisms underlying menthol binding and activation of TRPM8 ion channel. Nat Commun 2020; 11:3790. [PMID: 32728032 PMCID: PMC7391767 DOI: 10.1038/s41467-020-17582-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/06/2020] [Indexed: 02/07/2023] Open
Abstract
Menthol in mints elicits coolness sensation by selectively activating TRPM8 channel. Although structures of TRPM8 were determined in the apo and liganded states, the menthol-bounded state is unresolved. To understand how menthol activates the channel, we docked menthol to the channel and systematically validated our menthol binding models with thermodynamic mutant cycle analysis. We observed that menthol uses its hydroxyl group as a hand to specifically grab with R842, and its isopropyl group as legs to stand on I846 and L843. By imaging with fluorescent unnatural amino acid, we found that menthol binding induces wide-spread conformational rearrangements within the transmembrane domains. By Φ analysis based on single-channel recordings, we observed a temporal sequence of conformational changes in the S6 bundle crossing and the selectivity filter leading to channel activation. Therefore, our study suggested a 'grab and stand' mechanism of menthol binding and how menthol activates TRPM8 at the atomic level.
Collapse
Affiliation(s)
- Lizhen Xu
- Department of Biophysics, and Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang Province, China
| | - Yalan Han
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, 650223, Kunming, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoying Chen
- Department of Biophysics, and Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang Province, China
| | - Aerziguli Aierken
- Department of Biophysics, and Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang Province, China
| | - Han Wen
- Department of Physics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Wenjun Zheng
- Department of Physics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Hongkun Wang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
- Institute of Translational Medicine, Zhejiang University, Zhejiang, China
| | - Xiancui Lu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, 650223, Kunming, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhenye Zhao
- Department of Biophysics, and Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang Province, China
| | - Cheng Ma
- Protein facility, School of Medicine, Zhejiang University, Zhejiang, China
| | - Ping Liang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
- Institute of Translational Medicine, Zhejiang University, Zhejiang, China
| | - Wei Yang
- Department of Biophysics, and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang Province, China.
| | - Shilong Yang
- College of Wildlife and Protected Area, Northeast Forestry University, 150040, Harbin, China.
| | - Fan Yang
- Department of Biophysics, and Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
48
|
Mohandass A, Krishnan V, Gribkova ED, Asuthkar S, Baskaran P, Nersesyan Y, Hussain Z, Wise LM, George RE, Stokes N, Alexander BM, Cohen AM, Pavlov EV, Llano DA, Zhu MX, Thyagarajan B, Zakharian E. TRPM8 as the rapid testosterone signaling receptor: Implications in the regulation of dimorphic sexual and social behaviors. FASEB J 2020; 34:10887-10906. [PMID: 32609392 PMCID: PMC7496617 DOI: 10.1096/fj.202000794r] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/20/2020] [Accepted: 06/08/2020] [Indexed: 01/19/2023]
Abstract
Testosterone regulates dimorphic sexual behaviors in all vertebrates. However, the molecular mechanism underlying these behaviors remains unclear. Here, we report that a newly identified rapid testosterone signaling receptor, Transient Receptor Potential Melastatin 8 (TRPM8), regulates dimorphic sexual and social behaviors in mice. We found that, along with higher steroid levels in the circulation, TRPM8-/- male mice exhibit increased mounting frequency indiscriminate of sex, delayed sexual satiety, and increased aggression compared to wild-type controls, while TRPM8-/- females display an increased olfaction-exploratory behavior. Furthermore, neuronal responses to acute testosterone application onto the amygdala were attenuated in TRPM8-/- males but remained unchanged in females. Moreover, activation of dopaminergic neurons in the ventral tegmental area following mating was impaired in TRPM8-/- males. Together, these results demonstrate that TRPM8 regulates dimorphic sexual and social behaviors, and potentially constitutes a signalosome for mediation of sex-reward mechanism in males. Thus, deficiency of TRPM8 might lead to a delayed sexual satiety phenomenon.
Collapse
Affiliation(s)
- Adithya Mohandass
- College of Health Sciences, School of Pharmacy, University of Wyoming, Laramie, WY, USA
| | - Vivek Krishnan
- College of Health Sciences, School of Pharmacy, University of Wyoming, Laramie, WY, USA
| | - Ekaterina D Gribkova
- Department of Molecular and Integrative Physiology, Neuroscience Program and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Swapna Asuthkar
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| | - Padmamalini Baskaran
- College of Health Sciences, School of Pharmacy, University of Wyoming, Laramie, WY, USA
| | - Yelena Nersesyan
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| | - Zahir Hussain
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA.,Department of Physiology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Leslie M Wise
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| | - Robert E George
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| | - Nadarra Stokes
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| | | | - Alejandro M Cohen
- Biological Mass Spectrometry Core Facility, Life Sciences Research Institute, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Evgeny V Pavlov
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, USA
| | - Daniel A Llano
- Department of Molecular and Integrative Physiology, Neuroscience Program and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Baskaran Thyagarajan
- College of Health Sciences, School of Pharmacy, University of Wyoming, Laramie, WY, USA
| | - Eleonora Zakharian
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, IL, USA
| |
Collapse
|
49
|
Ramírez-Barrantes R, Carvajal-Zamorano K, Rodriguez B, Cordova C, Lozano C, Simon F, Díaz P, Muñoz P, Marchant I, Latorre R, Castillo K, Olivero P. TRPV1-Estradiol Stereospecific Relationship Underlies Cell Survival in Oxidative Cell Death. Front Physiol 2020; 11:444. [PMID: 32528302 PMCID: PMC7265966 DOI: 10.3389/fphys.2020.00444] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 04/09/2020] [Indexed: 12/31/2022] Open
Abstract
17β-estradiol is a neuronal survival factor against oxidative stress that triggers its protective effect even in the absence of classical estrogen receptors. The polymodal transient receptor potential vanilloid subtype 1 (TRPV1) channel has been proposed as a steroid receptor implied in tissue protection against oxidative damage. We show here that TRPV1 is sufficient condition for 17β-estradiol to enhance metabolic performance in injured cells. Specifically, in TRPV1 expressing cells, the application of 17β-estradiol within the first 3 h avoided H2O2-dependent mitochondrial depolarization and the activation of caspase 3/7 protecting against the irreversible damage triggered by H2O2. Furthermore, 17β-estradiol potentiates TRPV1 single channel activity associated with an increased open probability. This effect was not observed after the application of 17α-estradiol. We explored the TRPV1-Estrogen relationship also in primary culture of hippocampal-derived neurons and observed that 17β-estradiol cell protection against H2O2-induced damage was independent of estrogen receptors pathway activation, membrane started and stereospecific. These results support the role of TRPV1 as a 17β-estradiol-activated ionotropic membrane receptor coupling with mitochondrial function and cell survival.
Collapse
Affiliation(s)
- Ricardo Ramírez-Barrantes
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Escuela de Tecnología Médica, Universidad Andrés Bello, Viña del Mar, Chile
| | - Karina Carvajal-Zamorano
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Belen Rodriguez
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Claudio Cordova
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Carlo Lozano
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interoperativo en Ciencias Odontológicas y Médicas, Universidad de Valparaíso, Valparaíso, Chile
| | - Felipe Simon
- Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Chile, Santiago, Chile
| | - Paula Díaz
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Pablo Muñoz
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Ivanny Marchant
- Centro Interoperativo en Ciencias Odontológicas y Médicas, Universidad de Valparaíso, Valparaíso, Chile
| | - Ramón Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Karen Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Pablo Olivero
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interoperativo en Ciencias Odontológicas y Médicas, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
50
|
Steroids and TRP Channels: A Close Relationship. Int J Mol Sci 2020; 21:ijms21113819. [PMID: 32471309 PMCID: PMC7325571 DOI: 10.3390/ijms21113819] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 02/07/2023] Open
Abstract
Transient receptor potential (TRP) channels are remarkable transmembrane protein complexes that are essential for the physiology of the tissues in which they are expressed. They function as non-selective cation channels allowing for the signal transduction of several chemical, physical and thermal stimuli and modifying cell function. These channels play pivotal roles in the nervous and reproductive systems, kidney, pancreas, lung, bone, intestine, among others. TRP channels are finely modulated by different mechanisms: regulation of their function and/or by control of their expression or cellular/subcellular localization. These mechanisms are subject to being affected by several endogenously-produced compounds, some of which are of a lipidic nature such as steroids. Fascinatingly, steroids and TRP channels closely interplay to modulate several physiological events. Certain TRP channels are affected by the typical genomic long-term effects of steroids but others are also targets for non-genomic actions of some steroids that act as direct ligands of these receptors, as will be reviewed here.
Collapse
|