1
|
Wolf F, Dietrich-Ntoukas T, Reinach PS, Pleyer U, Mergler S. Nerve Growth Factor Modulates Regulatory Cell Volume Behavior via Stimulating TRPV1, TRPM8 Channels and Inducing Ca 2+ Signaling in Human Conjunctival Epithelial Cells. Cells 2025; 14:719. [PMID: 40422222 DOI: 10.3390/cells14100719] [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: 04/04/2025] [Revised: 05/06/2025] [Accepted: 05/09/2025] [Indexed: 05/28/2025] Open
Abstract
NGF plays important roles in ocular surface homeostasis and different pathological conditions. One effect includes promoting conjunctival epithelial cell differentiation and mucin secretion. This study characterizes the individual roles of TRPV1 and TRPM8 channel activity in mediating the effects of NGF on intracellular Ca2+ regulation and its alteration of regulatory cell volume responses to anisosmotic challenges in human conjunctival epithelial cells (IOBA-NHC). With fura-2/AM-loaded cells, the effects of 40 µM capsaicin and 20 µM AMG 9810 on Ca2+ regulation confirm functional TRPV1 expression. TRPM8 expression is evident since 500 µM menthol and 20 µM AMTB have opposing effects on [Ca2+]i. AMG 9810 and AMTB (both 20 µM) suppress the responses to NGF (100 ng/mL). With calcein/AM-loaded cells, the effects of these mediators are evaluated on apparent cell volume responses induced by an anisosmotic challenge. NGF decreases the apparent cell volume that AMG 9810 suppresses, whereas AMTB (both 20 µM) augments this response. Therefore, NGF interacts with TRPV1 and TRPM8 to induce opposing effects on cell volume regulatory behavior. These opposing effects suggest that the signaling pathways and effectors that mediate responses to TRPV1 and TRPM8 activation are not the same.
Collapse
Affiliation(s)
- Friedrich Wolf
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Tina Dietrich-Ntoukas
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Peter S Reinach
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou 325015, China
| | - Uwe Pleyer
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Stefan Mergler
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
2
|
Rech L, Dietrich-Ntoukas T, Reinach PS, Brockmann T, Pleyer U, Mergler S. Complement Component C5a and Fungal Pathogen Induce Diverse Responses through Crosstalk between Transient Receptor Potential Channel (TRPs) Subtypes in Human Conjunctival Epithelial Cells. Cells 2024; 13:1329. [PMID: 39195219 PMCID: PMC11352353 DOI: 10.3390/cells13161329] [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: 06/28/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024] Open
Abstract
The conjunctiva has immune-responsive properties to protect the eye from infections. Its innate immune system reacts against external pathogens, such as fungi. The complement factor C5a is an important contributor to the initial immune response. It is known that activation of transient-receptor-potential-vanilloid 1 (TRPV1) and TRP-melastatin 8 (TRPM8) channels is involved in different immune reactions and inflammation in the human body. The aim of this study was to determine if C5a and mucor racemosus e voluminae cellulae (MR) modulate Ca2+-signaling through changes in TRPs activity in human conjunctival epithelial cells (HCjECs). Furthermore, crosstalk was examined between C5a and MR in mediating calcium regulation. Intracellular Ca2+-concentration ([Ca2+]i) was measured by fluorescence calcium imaging, and whole-cell currents were recorded using the planar-patch-clamp technique. MR was used as a purified extract. Application of C5a (0.05-50 ng/mL) increased both [Ca2+]i and whole-cell currents, which were suppressed by either the TRPV1-blocker AMG 9810 or the TRPM8-blocker AMTB (both 20 µM). The N-terminal peptide C5L2p (20-50 ng/mL) blocked rises in [Ca2+]i induced by C5a. Moreover, the MR-induced rise in Ca2+-influx was suppressed by AMG 9810 and AMTB, as well as 0.05 ng/mL C5a. In conclusion, crosstalk between C5a and MR controls human conjunctival cell function through modulating interactions between TRPV1 and TRPM8 channel activity.
Collapse
Affiliation(s)
- Loreena Rech
- Department of Ophthalmology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (L.R.); (T.D.-N.); (U.P.)
| | - Tina Dietrich-Ntoukas
- Department of Ophthalmology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (L.R.); (T.D.-N.); (U.P.)
| | - Peter S. Reinach
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou 325015, China;
| | - Tobias Brockmann
- Department of Ophthalmology, Universitätsmedizin Rostock, 18057 Rostock, Germany;
- SciTec Department, University of Applied Sciences Jena, 07745 Jena, Germany
| | - Uwe Pleyer
- Department of Ophthalmology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (L.R.); (T.D.-N.); (U.P.)
| | - Stefan Mergler
- Department of Ophthalmology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (L.R.); (T.D.-N.); (U.P.)
| |
Collapse
|
3
|
Hu XQ, Zhang L. Role of transient receptor potential channels in the regulation of vascular tone. Drug Discov Today 2024; 29:104051. [PMID: 38838960 PMCID: PMC11938208 DOI: 10.1016/j.drudis.2024.104051] [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: 01/16/2024] [Revised: 05/17/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024]
Abstract
Vascular tone is a major element in the control of hemodynamics. Transient receptor potential (TRP) channels conducting monovalent and/or divalent cations (e.g. Na+ and Ca2+) are expressed in the vasculature. Accumulating evidence suggests that TRP channels participate in regulating vascular tone by regulating intracellular Ca2+ signaling in both vascular smooth muscle cells (VSMCs) and endothelial cells (ECs). Aberrant expression/function of TRP channels in the vasculature is associated with vascular dysfunction in systemic/pulmonary hypertension and metabolic syndromes. This review intends to summarize our current knowledge of TRP-mediated regulation of vascular tone in both physiological and pathophysiological conditions and to discuss potential therapeutic approaches to tackle abnormal vascular tone due to TRP dysfunction.
Collapse
Affiliation(s)
- Xiang-Qun Hu
- Lawrence D. Longo MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| | - Lubo Zhang
- Lawrence D. Longo MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| |
Collapse
|
4
|
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
|
5
|
Stinson RJ, Morice AH, Sadofsky LR. Modulation of transient receptor potential (TRP) channels by plant derived substances used in over-the-counter cough and cold remedies. Respir Res 2023; 24:45. [PMID: 36755306 PMCID: PMC9907891 DOI: 10.1186/s12931-023-02347-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 01/27/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Upper respiratory tract infections (URTIs) impact all age groups and have a significant economic and social burden on society, worldwide. Most URTIs are mild and self-limiting, but due to the wide range of possible causative agents, including Rhinovirus (hRV), Adenovirus, Respiratory Syncytial Virus (RSV), Coronavirus and Influenza, there is no single and effective treatment. Over-the-counter (OTC) remedies, including traditional medicines and those containing plant derived substances, help to alleviate symptoms including inflammation, pain, fever and cough. PURPOSE This systematic review focuses on the role of the major plant derived substances in several OTC remedies used to treat cold symptoms, with a particular focus on the transient receptor potential (TRP) channels involved in pain and cough. METHODS Literature searches were done using Pubmed and Web of Science, with no date limitations, using the principles of the PRISMA statement. The search terms used were 'TRP channel AND plant compound', 'cough AND plant compound', 'cough AND TRP channels AND plant compound', 'cough AND P2X3 AND plant compound' and 'P2X3 AND plant compound' where plant compound represents menthol or camphor or eucalyptus or turpentine or thymol. RESULTS The literature reviewed showed that menthol activates TRPM8 and may inhibit respiratory reflexes reducing irritation and cough. Menthol has a bimodal action on TRPA1, but inhibition may have an analgesic effect. Eucalyptus also activates TRPM8 and inhibits TRPA1 whilst down regulating P2X3, aiding in the reduction of cough, pain and airway irritation. Camphor inhibits TRPA1 and the activation of TRPM8 may add to the effects of menthol. Activation of TRPV1 by camphor, may also have an analgesic effect. CONCLUSIONS The literature suggests that these plant derived substances have multifaceted actions and can interact with the TRP 'cough' receptors. The plant derived substances used in cough and cold medicines have the potential to target multiple symptoms experienced during a cold.
Collapse
Affiliation(s)
- Rebecca J. Stinson
- grid.9481.40000 0004 0412 8669Centre for Biomedicine, Hull York Medical School, The University of Hull, Cottingham Road, Hull, HU6 7RX UK
| | - Alyn H. Morice
- grid.413631.20000 0000 9468 0801Clinical Sciences Centre, Hull York Medical School, Castle Hill Hospital, Cottingham, Hull, HU16 5JQ UK
| | - Laura R. Sadofsky
- grid.9481.40000 0004 0412 8669Centre for Biomedicine, Hull York Medical School, The University of Hull, Cottingham Road, Hull, HU6 7RX UK
| |
Collapse
|
6
|
Zhang Z, Kang L, Yan X, Leng Z, Fang K, Chen T, Xu M. Global Trends and Hotspots of Transient Receptor Potential Melastatin 8 Research from 2002 to 2021: A Bibliometric Analysis. J Pain Res 2022; 15:3881-3892. [PMID: 36536695 PMCID: PMC9759117 DOI: 10.2147/jpr.s393582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/06/2022] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Transient receptor potential channels are the major temperature and nociceptive receptors in the human body and transient receptor potential melastatin 8 (TRPM8) is the cold-sensitive and non-selective cation channel. In our study, we performed a bibliometric analysis of TRPM8 from 2002 to 2021 to summarize the current research status and potential research direction in the future. METHODS The TRPM8-related publications were selected from the Web of Science Core Collection SCI-EXPANDED database from 2002 to 2021. The publication details, such as authors, titles, and author keywords, were used for bibliometric analysis and network visualization to present the current state of TRPM8 research. RESULTS A total of 1035 articles met the inclusion criteria. The number of TRPM8-related articles has grown rapidly over the past two decades. The USA has the largest number of publications, citations, and international collaborations. The TRPM8-related articles are mainly published and cited in neurological journals, such as the Journal of Neuroscience (41 publications and 2171 local citations). Prevarskaya N. has the most publications (26), and Patapoutian A. has been cited the most (1414 local citations). The popular disciplines in TRPM8 research include Neurosciences and Neurology, Pharmacology and Pharmacy, Biochemistry, and Molecular Biology. Research hotspots are mainly TRP channel, calcium, prostate cancer, proliferation, pain, cold, nociception, and inflammation. CONCLUSION Our bibliometric analysis demonstrates that the number of TRPM8 studies has increased from 2002 to 2021. The global research trends and hotspots include the activation mechanism of TRPM8 in neurons, the role of TRPM8 in neuronal and non-neuronal diseases, and therapeutic target research.
Collapse
Affiliation(s)
- Zehua Zhang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Le Kang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Xiaohan Yan
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Zhuyun Leng
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Kang Fang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Tao Chen
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Meidong Xu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| |
Collapse
|
7
|
Donau J, Luo H, Virta I, Skupin A, Pushina M, Loeffler J, Haertel FV, Das A, Kurth T, Gerlach M, Lindemann D, Reinach PS, Mergler S, Valtink M. TRPV4 Stimulation Level Regulates Ca2+-Dependent Control of Human Corneal Endothelial Cell Viability and Survival. MEMBRANES 2022; 12:membranes12030281. [PMID: 35323756 PMCID: PMC8952823 DOI: 10.3390/membranes12030281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/18/2022] [Accepted: 02/24/2022] [Indexed: 02/04/2023]
Abstract
The functional contribution of transient receptor potential vanilloid 4 (TRPV4) expression in maintaining human corneal endothelial cells (HCEC) homeostasis is unclear. Accordingly, we determined the effects of TRPV4 gene and protein overexpression on responses modulating the viability and survival of HCEC. Q-PCR, Western blot, FACS analyses and fluorescence single-cell calcium imaging confirmed TRPV4 gene and protein overexpression in lentivirally transduced 12V4 cells derived from their parent HCEC-12 line. Although TRPV4 overexpression did not alter the baseline transendothelial electrical resistance (TEER), its cellular capacitance (Ccl) was larger than that in its parent. Scanning electron microscopy revealed that only the 12V4 cells developed densely packed villus-like protrusions. Stimulation of TRPV4 activity with GSK1016790A (GSK101, 10 µmol/L) induced larger Ca2+ transients in the 12V4 cells than those in the parental HCEC-12. One to ten nmol/L GSK101 decreased 12V4 viability, increased cell death rates and reduced the TEER, whereas 1 µmol/L GSK101 was required to induce similar effects in the HCEC-12. However, the TRPV4 channel blocker RN1734 (1 to 30 µmol/L) failed to alter HCEC-12 and 12V4 morphology, cell viability and metabolic activity. Taken together, TRPV4 overexpression altered both the HCEC morphology and markedly lowered the GSK101 dosages required to stimulate its channel activity.
Collapse
Affiliation(s)
- Jennifer Donau
- Institute of Anatomy, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; (J.D.); (A.S.); (M.P.); (J.L.)
- Institute of Medical Microbiology and Virology, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany;
| | - Huan Luo
- Klinik für Augenheilkunde, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany; (H.L.); (I.V.)
| | - Iiris Virta
- Klinik für Augenheilkunde, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany; (H.L.); (I.V.)
| | - Annett Skupin
- Institute of Anatomy, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; (J.D.); (A.S.); (M.P.); (J.L.)
- Institute of Medical Microbiology and Virology, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany;
| | - Margarita Pushina
- Institute of Anatomy, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; (J.D.); (A.S.); (M.P.); (J.L.)
| | - Jana Loeffler
- Institute of Anatomy, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; (J.D.); (A.S.); (M.P.); (J.L.)
| | - Frauke V. Haertel
- Institute of Physiology, Faculty of Medicine, University Giessen, 35392 Giessen, Germany;
- Institute of Physiology, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany;
| | - Anupam Das
- Institute of Physiology, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany;
| | - Thomas Kurth
- Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform, TU Dresden, 01307 Dresden, Germany;
| | - Michael Gerlach
- Core Facility Cellular Imaging, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany;
| | - Dirk Lindemann
- Institute of Medical Microbiology and Virology, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany;
| | - Peter S. Reinach
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou 325027, China;
| | - Stefan Mergler
- Klinik für Augenheilkunde, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany; (H.L.); (I.V.)
- Correspondence: (S.M.); (M.V.)
| | - Monika Valtink
- Institute of Anatomy, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; (J.D.); (A.S.); (M.P.); (J.L.)
- Equality and Diversity Unit, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
- Correspondence: (S.M.); (M.V.)
| |
Collapse
|
8
|
Wang G, Cao R, Qian K, Peng T, Yuan L, Chen L, Cheng S, Xiong Y, Ju L, Wang X, Xiao Y. TRPM8 Inhibition Regulates the Proliferation, Migration and ROS Metabolism of Bladder Cancer Cells. Onco Targets Ther 2020; 13:8825-8835. [PMID: 32943886 PMCID: PMC7481304 DOI: 10.2147/ott.s257056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/11/2020] [Indexed: 12/20/2022] Open
Abstract
Introduction Based on accumulating evidence, transient receptor potential (TRP) ion channels may play important roles in the occurrence and the progression of cancer. TRP melastatin 8 (TRPM8), a member of the TRP family, functions as a Ca2+-permeable channel and regulates various physiological and pathological processes. However, the effects of TRPM8 on bladder cancer (BCa) and its underlying mechanisms have not been elucidated. Methods BCa tissues and matched noncancerous tissues were collected to examine the expression of the TRPM8 mRNA and protein using qRT-PCR, Western blotting and immunofluorescence staining. Meanwhile, the effect of knockdown or inhibition of the activity of the TRPM8 protein on the proliferation, migration and ROS metabolism of bladder cancer cells was detected using the MTT assay, clonogenic survival assay, Transwell chamber migration assay, and reactive oxygen species (ROS) detection, respectively. Furthermore, a mouse model transplanted with BCa cells was established to assess tumor growth after TRPM8 expression was inhibited in vivo. Results Compared with the noncancerous tissues, the levels of TRPM8 in BCa tissues were significantly increased. Knockdown or inhibition of the activity of the TRPM8 protein in BCa cells reduced cell proliferation and migration. Moreover, the production of ROS was increased in cells treated with siTRPM8, which was accompanied by increased levels of Catalase, HO-1 and SOD2. Furthermore, a mouse model transplanted with the stable TRPM8-deficient T24 cell line was established, demonstrating that knockdown of TRPM8 delayed tumor growth in vivo. Discussion TRPM8 might play an essential for BCa tumor progression and metastasis by interfering with BCa cell proliferation, motility, ROS metabolism and migration.
Collapse
Affiliation(s)
- Gang Wang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China.,Human Genetics Resource Preservation Center of Wuhan University, Wuhan, People's Republic of China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, People's Republic of China.,Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Wuhan, People's Republic of China
| | - Rui Cao
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Kaiyu Qian
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China.,Human Genetics Resource Preservation Center of Wuhan University, Wuhan, People's Republic of China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, People's Republic of China.,Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Wuhan, People's Republic of China
| | - Tianchen Peng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Lushun Yuan
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Liang Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Songtao Cheng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Yaoyi Xiong
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Lingao Ju
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China.,Human Genetics Resource Preservation Center of Wuhan University, Wuhan, People's Republic of China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, People's Republic of China.,Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Wuhan, People's Republic of China
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Yu Xiao
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China.,Human Genetics Resource Preservation Center of Wuhan University, Wuhan, People's Republic of China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, People's Republic of China.,Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Wuhan, People's Republic of China.,Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
9
|
Developmental change in the gene expression of transient receptor potential melastatin channel 3 (TRPM3) in murine lacrimal gland. Ann Anat 2020; 231:151551. [PMID: 32512204 DOI: 10.1016/j.aanat.2020.151551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 05/19/2020] [Indexed: 11/20/2022]
Abstract
Transient receptor potential (TRP) channels are cation channels with ubiquitous expression. Various TRP channels are functionally active at the ocular surface and are involved in tear secretion and multiple inflammatory processes. So far, the impact of TRP channels regarding the development of the lacrimal gland (LG) is unclear. While investigating TRP channels in the LG, the TRPM3 channel presented itself as a promising candidate to play a role in the development and functioning of the LG. Therefore, Trpm3 expression was analyzed in different embryonic and postembryonic LGs. Thus, gene expression of TRPM channels including Trpm2, Trpm3, Trpm4 and Trpm6 was analyzed by quantitative RT-PCR in murine LGs at different developmental stages. Localization of TRPM3 in LGs was examined by immunohistochemistry. Primary LG epithelial cells (LGEC) and mesenchymal cells (MC) from newborn mice were cultured (either separately or collectively) for three days, and Trpm3 expression was analyzed in LGEC and MC. As a result, gene expression of Trpm2, Trpm4 and Trpm6 showed no significant difference in LGs in the different stages of development. However, Trpm3 gene expression was significantly higher in the embryonic stage than in the postnatal stage with the peak at E18. Postnatal, Trpm3 expression significantly decreased up to 28-fold until two years of age. Immunohistochemistry for TRPM3 revealed apical membranous expression in the excretory ducts, as well as in the acini of up to P7 old mice. Trpm3 expression in LGEC were significantly higher than that of MC. Our results indicate that Trpm3 expression in murine LG is age-dependent and peaks at age E18. Its expression is localized in the apical membrane of the glandular epithelium. However, its functional role still requires additional study in the LG.
Collapse
|
10
|
Fiorio Pla A, Gkika D. Ca2+ Channel Toolkit in Neuroendocrine Tumors. Neuroendocrinology 2020; 110:147-154. [PMID: 31177261 DOI: 10.1159/000501397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 06/06/2019] [Indexed: 11/19/2022]
Abstract
Neuroendocrine tumors (NET) constitute a heterogeneous group of malignancies with various clinical presentations and growth rates but a common origin in neuroendocrine cells located all over the body. NET are a relatively low-frequency disease mostly represented by gastroenteropancreatic (GEP) and bronchopulmonary tumors (pNET); on the other hand, an increasing frequency and prevalence have been associated with NET. Despite great efforts in recent years, the management of NET is still a critical unmet need due to the lack of knowledge of the biology of the disease, the lack of adequate biomarkers, late presentation, the relative insensitivity of imaging modalities, and a paucity of predictably effective treatment options. In this context Ca2+ signals, being pivotal molecular devices in sensing and integrating signals from the microenvironment, are emerging to be particularly relevant in cancer, where they mediate interactions between tumor cells and the tumor microenvironment to drive different aspects of neoplastic progression (e.g., cell proliferation and survival, cell invasiveness, and proangiogenetic programs). Indeed, ion channels represent good potential pharmacological targets due to their location on the plasma membrane, where they can be easily accessed by drugs. The present review aims to provide a critical and up-to-date overview of NET development integrating Ca2+ signal involvement. In this perspective, we first give an introduction to NET and Ca2+ channels and then describe the different families of Ca2+ channels implicated in NET, i.e., ionotropic receptors, voltage-dependent Ca2+ channels, and transient receptor potential channels, as well as intracellular Ca2+ channels and their signaling molecules.
Collapse
Affiliation(s)
- Alessandra Fiorio Pla
- Department of Life Science and Systems Biology, University of Torino, Turin, Italy,
- Inserm, U1003 - PHYCEL (Physiologie Cellulaire), Université de Lille, Lille, France,
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France,
| | - Dimitra Gkika
- Inserm, U1003 - PHYCEL (Physiologie Cellulaire), Université de Lille, Lille, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| |
Collapse
|
11
|
|
12
|
Walcher L, Budde C, Böhm A, Reinach PS, Dhandapani P, Ljubojevic N, Schweiger MW, von der Waydbrink H, Reimers I, Köhrle J, Mergler S. TRPM8 Activation via 3-Iodothyronamine Blunts VEGF-Induced Transactivation of TRPV1 in Human Uveal Melanoma Cells. Front Pharmacol 2018. [DOI: 10.3389/fphar.2018.01234 ecollection 2018.pmid: 30483120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2022] Open
|
13
|
Walcher L, Budde C, Böhm A, Reinach PS, Dhandapani P, Ljubojevic N, Schweiger MW, von der Waydbrink H, Reimers I, Köhrle J, Mergler S. TRPM8 Activation via 3-Iodothyronamine Blunts VEGF-Induced Transactivation of TRPV1 in Human Uveal Melanoma Cells. Front Pharmacol 2018; 9:1234. [PMID: 30483120 PMCID: PMC6243059 DOI: 10.3389/fphar.2018.01234] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 10/11/2018] [Indexed: 01/17/2023] Open
Abstract
In human uveal melanoma (UM), tumor enlargement is associated with increases in aqueous humor vascular endothelial growth factor-A (VEGF-A) content that induce neovascularization. 3-Iodothyronamine (3-T1AM), an endogenous thyroid hormone metabolite, activates TRP melastatin 8 (TRPM8), which blunts TRP vanilloid 1 (TRPV1) activation by capsaicin (CAP) in human corneal, conjunctival epithelial cells, and stromal cells. We compare here the effects of TRPM8 activation on VEGF-induced transactivation of TRPV1 in an UM cell line (92.1) with those in normal primary porcine melanocytes (PM) since TRPM8 is upregulated in melanoma. Fluorescence Ca2+-imaging and planar patch-clamping characterized functional channel activities. CAP (20 μM) induced Ca2+ transients and increased whole-cell currents in both the UM cell line and PM whereas TRPM8 agonists, 100 μM menthol and 20 μM icilin, blunted such responses in the UM cells. VEGF (10 ng/ml) elicited Ca2+ transients and augmented whole-cell currents, which were blocked by capsazepine (CPZ; 20 μM) but not by a highly selective TRPM8 blocker, AMTB (20 μM). The VEGF-induced current increases were not augmented by CAP. Both 3-T1AM (1 μM) and menthol (100 μM) increased the whole-cell currents, whereas 20 μM AMTB blocked them. 3-T1AM exposure suppressed both VEGF-induced Ca2+ transients and increases in underlying whole-cell currents. Taken together, functional TRPM8 upregulation in UM 92.1 cells suggests that TRPM8 is a potential drug target for suppressing VEGF induced increases in neovascularization and UM tumor growth since TRPM8 activation blocked VEGF transactivation of TRPV1.
Collapse
Affiliation(s)
- Lia Walcher
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Clara Budde
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Arina Böhm
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Peter S Reinach
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | | | - Nina Ljubojevic
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Markus W Schweiger
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Henriette von der Waydbrink
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ilka Reimers
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Josef Köhrle
- Institut für Experimentelle Endokrinologie, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefan Mergler
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
14
|
Türker E, Garreis F, Khajavi N, Reinach PS, Joshi P, Brockmann T, Lucius A, Ljubojevic N, Turan E, Cooper D, Schick F, Reinholz R, Pleyer U, Köhrle J, Mergler S. Vascular Endothelial Growth Factor (VEGF) Induced Downstream Responses to Transient Receptor Potential Vanilloid 1 (TRPV1) and 3-Iodothyronamine (3-T 1AM) in Human Corneal Keratocytes. Front Endocrinol (Lausanne) 2018; 9:670. [PMID: 30524369 PMCID: PMC6262029 DOI: 10.3389/fendo.2018.00670] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 10/26/2018] [Indexed: 12/30/2022] Open
Abstract
This study was undertaken to determine if crosstalk among the transient receptor potential (TRP) melastatin 8 (TRPM8), TRP vanilloid 1 (TRPV1), and vascular endothelial growth factor (VEGF) receptor triad modulates VEGF-induced Ca2+ signaling in human corneal keratocytes. Using RT-PCR, qPCR and immunohistochemistry, we determined TRPV1 and TRPM8 gene and protein coexpression in a human corneal keratocyte cell line (HCK) and human corneal cross sections. Fluorescence Ca2+ imaging using both a photomultiplier and a single cell digital imaging system as well as planar patch-clamping measured relative intracellular Ca2+ levels and underlying whole-cell currents. The TRPV1 agonist capsaicin increased both intracellular Ca2+ levels and whole-cell currents, while the antagonist capsazepine (CPZ) inhibited them. VEGF-induced Ca2+ transients and rises in whole-cell currents were suppressed by CPZ, whereas a selective TRPM8 antagonist, AMTB, increased VEGF signaling. In contrast, an endogenous thyroid hormone-derived metabolite 3-Iodothyronamine (3-T1AM) suppressed increases in the VEGF-induced current. The TRPM8 agonist menthol increased the currents, while AMTB suppressed this response. The VEGF-induced increases in Ca2+ influx and their underlying ionic currents stem from crosstalk between VEGFR and TRPV1, which can be impeded by 3-T1AM-induced TRPM8 activation. Such suppression in turn blocks VEGF-induced TRPV1 activation. Therefore, crosstalk between TRPM8 and TRPV1 inhibits VEGFR-induced activation of TRPV1.
Collapse
Affiliation(s)
- Ersal Türker
- Klinik für Augenheilkunde, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Fabian Garreis
- Department of Functional and Clinical Anatomy, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Noushafarin Khajavi
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Walter Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, Munich, Germany
| | - Peter S. Reinach
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Pooja Joshi
- Klinik für Augenheilkunde, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Tobias Brockmann
- Klinik für Augenheilkunde, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Alexander Lucius
- Klinik für Augenheilkunde, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nina Ljubojevic
- Klinik für Augenheilkunde, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Elizabeth Turan
- Klinik für Augenheilkunde, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Drew Cooper
- Klinik für Augenheilkunde, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Felix Schick
- Klinik für Augenheilkunde, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Rob Reinholz
- Klinik für Augenheilkunde, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Uwe Pleyer
- Klinik für Augenheilkunde, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Josef Köhrle
- Institut für Experimentelle Endokrinologie, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Stefan Mergler
- Klinik für Augenheilkunde, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- *Correspondence: Stefan Mergler
| |
Collapse
|
15
|
Chiellini G, Bellusci L, Sabatini M, Zucchi R. Thyronamines and Analogues - The Route from Rediscovery to Translational Research on Thyronergic Amines. Mol Cell Endocrinol 2017; 458:149-155. [PMID: 28069535 DOI: 10.1016/j.mce.2017.01.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/07/2016] [Accepted: 01/02/2017] [Indexed: 11/18/2022]
Abstract
Thyronamines are a novel class of endogenous signaling compounds, structurally related to thyroid hormones (THs). Specific thyronamines, particularly 3-iodothyronamine (T1AM), stimulate with nanomolar affinity trace amine-associated receptor 1 (TAAR1), a G protein-coupled membrane receptor, and may also interact with other TAAR subtypes (particularly TAAR5), adrenergic receptors (particularly α2 receptors), amine transporters, and mitochondrial proteins. In addition to its structural similarities with THs, T1AM also contains the arylethylamine scaffold as in monoamine neurotransmitters, implicating an intriguing role for T1AM as both a neuromodulator and a hormone-like molecule constituting a part of thyroid hormone signaling. A large number of T1AM derivatives have already been synthesized. We discuss the different chemical strategies followed to obtain thyronamine analogues, their potency at TAAR1, and their structure-activity relationship. Preliminary characterization of the functional effects of these synthetic compounds is also provided.
Collapse
|
16
|
Genova T, Grolez GP, Camillo C, Bernardini M, Bokhobza A, Richard E, Scianna M, Lemonnier L, Valdembri D, Munaron L, Philips MR, Mattot V, Serini G, Prevarskaya N, Gkika D, Pla AF. TRPM8 inhibits endothelial cell migration via a non-channel function by trapping the small GTPase Rap1. J Cell Biol 2017; 216:2107-2130. [PMID: 28550110 PMCID: PMC5496606 DOI: 10.1083/jcb.201506024] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 06/26/2016] [Accepted: 04/12/2017] [Indexed: 01/30/2023] Open
Abstract
Endothelial cell adhesion and migration are critical steps of the angiogenic process, whose dysfunction is associated with tumor growth and metastasis. The TRPM8 channel has recently been proposed to play a protective role in prostate cancer by impairing cell motility. However, the mechanisms by which it could influence vascular behavior are unknown. Here, we reveal a novel non-channel function for TRPM8 that unexpectedly acts as a Rap1 GTPase inhibitor, thereby inhibiting endothelial cell motility, independently of pore function. TRPM8 retains Rap1 intracellularly through direct protein-protein interaction, thus preventing its cytoplasm-plasma membrane trafficking. In turn, this mechanism impairs the activation of a major inside-out signaling pathway that triggers the conformational activation of integrin and, consequently, cell adhesion, migration, in vitro endothelial tube formation, and spheroid sprouting. Our results bring to light a novel, pore-independent molecular mechanism by which endogenous TRPM8 expression inhibits Rap1 GTPase and thus plays a critical role in the behavior of vascular endothelial cells by inhibiting migration.
Collapse
Affiliation(s)
- Tullio Genova
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy.,Department of Surgical Sciences, C.I.R. Dental School, University of Torino, Torino, Italy
| | - Guillaume P Grolez
- Laboratoire de Physiologie cellulaire, Institut National de la Santé et de la Recherche Médicale U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| | - Chiara Camillo
- Laboratory of Cell Adhesion Dynamics, Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia, Istituto di Ricovero e Cura a Carattere Scientifico, Department of Oncology, University of Torino School of Medicine, Candiolo, Italy
| | - Michela Bernardini
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy.,Laboratoire de Physiologie cellulaire, Institut National de la Santé et de la Recherche Médicale U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| | - Alexandre Bokhobza
- Laboratoire de Physiologie cellulaire, Institut National de la Santé et de la Recherche Médicale U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| | - Elodie Richard
- BICeL Campus Lille1, FR3688 FRABio, Université de Lille, Villeneuve d'Ascq, France
| | - Marco Scianna
- Department of Mathematical Sciences, Politecnico di Torino, Torino, Italy
| | - Loic Lemonnier
- Laboratoire de Physiologie cellulaire, Institut National de la Santé et de la Recherche Médicale U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| | - Donatella Valdembri
- Laboratory of Cell Adhesion Dynamics, Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia, Istituto di Ricovero e Cura a Carattere Scientifico, Department of Oncology, University of Torino School of Medicine, Candiolo, Italy
| | - Luca Munaron
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy.,Nanostructured Interfaces and Surfaces Centre of Excellence, University of Torino, Torino, Italy
| | - Mark R Philips
- Cancer Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY
| | - Virginie Mattot
- Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8161 - Mechanisms of Tumorigenesis and Target Therapies, Universite de Lille, Lille, France
| | - Guido Serini
- Laboratory of Cell Adhesion Dynamics, Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia, Istituto di Ricovero e Cura a Carattere Scientifico, Department of Oncology, University of Torino School of Medicine, Candiolo, Italy
| | - Natalia Prevarskaya
- Laboratoire de Physiologie cellulaire, Institut National de la Santé et de la Recherche Médicale U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| | - Dimitra Gkika
- Laboratoire de Physiologie cellulaire, Institut National de la Santé et de la Recherche Médicale U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| | - Alessandra Fiorio Pla
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy .,Nanostructured Interfaces and Surfaces Centre of Excellence, University of Torino, Torino, Italy.,Laboratoire de Physiologie cellulaire, Institut National de la Santé et de la Recherche Médicale U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| |
Collapse
|
17
|
Chen GL, Lei M, Zhou LP, Zeng B, Zou F. Borneol Is a TRPM8 Agonist that Increases Ocular Surface Wetness. PLoS One 2016; 11:e0158868. [PMID: 27448228 PMCID: PMC4957794 DOI: 10.1371/journal.pone.0158868] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 05/17/2016] [Indexed: 12/12/2022] Open
Abstract
Borneol is a compound widely used in ophthalmic preparations in China. Little is known about its exact role in treating eye diseases. Here we report that transient receptor potential melastatin 8 (TRPM8) channel is a pharmacological target of borneol and mediates its therapeutic effect in the eyes. Ca2+ measurement and electrophysiological recordings revealed that borneol activated TRPM8 channel in a temperature- and dose-dependent manner, which was similar to but less effective than the action of menthol, an established TRPM8 agonist. Borneol significantly increased tear production in guinea pigs without evoking nociceptive responses at 25°C, but failed to induce tear secretion at 35°C. In contrast, menthol evoked tearing response at both 25 and 35°C. TRPM8 channel blockers N-(3-Aminopropyl)-2-[(3-methylphenyl)methoxy]-N-(2-thienylmethyl)benzamide hydrochloride (AMTB) and N-(4-tert-butylphenyl)-4-(3-chloropyridin-2-yl)piperazine-1-carboxamide (BCTC) abolished borneol- and menthol-induced tear secretion. Borneol at micromolar concentrations did not affect the viability of human corneal epithelial cells. We conclude that borneol can activate the cold-sensing TRPM8 channel and modestly increase ocular surface wetness, which suggests it is an active compound in ophthalmic preparations and particularly useful in treating dry eye syndrome.
Collapse
Affiliation(s)
- Gui-Lan Chen
- Ministry of Education Key Laboratory of Bio-resources and Eco-environment, College of Life Sciences, Sichuan University, Chengdu, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, and Institute of Cardiovascular Research, Sichuan Medical University, Luzhou, China
| | - Ming Lei
- Key Laboratory of Medical Electrophysiology, Ministry of Education, and Institute of Cardiovascular Research, Sichuan Medical University, Luzhou, China
| | - Lu-Ping Zhou
- Key Laboratory of Medical Electrophysiology, Ministry of Education, and Institute of Cardiovascular Research, Sichuan Medical University, Luzhou, China
| | - Bo Zeng
- Key Laboratory of Medical Electrophysiology, Ministry of Education, and Institute of Cardiovascular Research, Sichuan Medical University, Luzhou, China
- * E-mail: (BZ); (FZ)
| | - Fangdong Zou
- Ministry of Education Key Laboratory of Bio-resources and Eco-environment, College of Life Sciences, Sichuan University, Chengdu, China
- * E-mail: (BZ); (FZ)
| |
Collapse
|
18
|
Giblin JP, Comes N, Strauss O, Gasull X. Ion Channels in the Eye: Involvement in Ocular Pathologies. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015; 104:157-231. [PMID: 27038375 DOI: 10.1016/bs.apcsb.2015.11.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The eye is the sensory organ of vision. There, the retina transforms photons into electrical signals that are sent to higher brain areas to produce visual sensations. In the light path to the retina, different types of cells and tissues are involved in maintaining the transparency of avascular structures like the cornea or lens, while others, like the retinal pigment epithelium, have a critical role in the maintenance of photoreceptor function by regenerating the visual pigment. Here, we have reviewed the roles of different ion channels expressed in ocular tissues (cornea, conjunctiva and neurons innervating the ocular surface, lens, retina, retinal pigment epithelium, and the inflow and outflow systems of the aqueous humor) that are involved in ocular disease pathophysiologies and those whose deletion or pharmacological modulation leads to specific diseases of the eye. These include pathologies such as retinitis pigmentosa, macular degeneration, achromatopsia, glaucoma, cataracts, dry eye, or keratoconjunctivitis among others. Several disease-associated ion channels are potential targets for pharmacological intervention or other therapeutic approaches, thus highlighting the importance of these channels in ocular physiology and pathophysiology.
Collapse
Affiliation(s)
- Jonathan P Giblin
- Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Nuria Comes
- Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Xavier Gasull
- Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
19
|
Reinach PS, Mergler S, Okada Y, Saika S. Ocular transient receptor potential channel function in health and disease. BMC Ophthalmol 2015; 15 Suppl 1:153. [PMID: 26818117 PMCID: PMC4895786 DOI: 10.1186/s12886-015-0135-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Transient receptor potential (TRP) channels sense and transduce environmental stimuli into Ca(2+) transients that in turn induce responses essential for cell function and adaptation. These non-selective channels with variable Ca(2+) selectivity are grouped into seven different subfamilies containing 28 subtypes based on differences in amino acid sequence homology. Many of these subtypes are expressed in the eye on both neuronal and non-neuronal cells where they affect a host of stress-induced regulatory responses essential for normal vision maintenance. This article reviews our current knowledge about the expression, function and regulation of TRPs in different eye tissues. We also describe how under certain conditions TRP activation can induce responses that are maladaptive to ocular function. Furthermore, the possibility of an association between TRP mutations and disease is considered. These findings contribute to evidence suggesting that drug targeting TRP channels may be of therapeutic benefit in a clinical setting. We point out issues that must be more extensively addressed before it will be possible to decide with certainty that this is a realistic endeavor. Another possible upshot of future studies is that disease process progression can be better evaluated by profiling changes in tissue specific functional TRP subtype activity as well as their gene and protein expression.
Collapse
Affiliation(s)
- Peter S Reinach
- Department of Ophthalmology and Optometry, Wenzhou Medical University, 270 Xuejuan Road, Wenzhou, Zhejiang, 325027, P. R. China.
| | - Stefan Mergler
- Department of Ophthalmology, Charité-University Medicine Berlin, Campus Virchow-Clinic, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Yuka Okada
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan.
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan.
| |
Collapse
|
20
|
Lucius A, Khajavi N, Reinach PS, Köhrle J, Dhandapani P, Huimann P, Ljubojevic N, Grötzinger C, Mergler S. 3-Iodothyronamine increases transient receptor potential melastatin channel 8 (TRPM8) activity in immortalized human corneal epithelial cells. Cell Signal 2015; 28:136-147. [PMID: 26689735 DOI: 10.1016/j.cellsig.2015.12.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 11/25/2015] [Accepted: 12/08/2015] [Indexed: 10/22/2022]
Abstract
3-Iodothyronamine (3T1AM) is an endogenous thyroid hormone metabolite that interacts with the human trace amine-associated receptor 1 (hTAAR1), a G-protein-coupled receptor, to induce numerous physiological responses including dose-dependent body temperature lowering in rodents. 3T1AM also directly activates cold-sensitive transient receptor potential melastatin 8 (TRPM8) channels in human conjunctival epithelial cells (HCjEC) at constant temperature as well as reducing rises in IL-6 release induced by transient receptor potential vanilloid 1 (TRPV1) activation by capsaicin (CAP). Here, we describe that 3T1AM-induced TRPM8 activation suppresses through crosstalk TRPV1 activation in immortalized human corneal epithelial cells (HCEC). RT-PCR and immunofluorescent staining identified TRPM8 gene and protein expression. Increases in Ca(2+) influx induced by the TRPM8 agonists either 3T1AM (0.1-10 μM), menthol (500 μM), icilin (15-60 μM) or temperature lowering (either <17°C or >17°C) were all blocked by 10-20 μM BCTC, a mixed TRPV1/TRPM8 antagonist. BCTC blocked 3T1AM-induced recombinant TRPM8 activation of Ca(2+) transients in an osteosarcoma heterologous expression system. The effects of BCTC in HCEC were attributable to selective TRPM8 inhibition since whole-cell patch-clamp currents underlying Ca(2+) rises induced by 20 μM CAP were BCTC insensitive. On the other hand, Ca(2+) transients induced by activating TRPV1 with either CAP or a hyperosmolar medium were suppressed during exposure to either 1 μM 3T1AM or 15 μM icilin. All of these modulatory effects on intracellular Ca(2+) regulation induced by the aforementioned agents were attributable to changes in underlying inward and outward current. Taken together, TRPM8 activation by 3T1AM markedly attenuates and even eliminates hyperosmolar and CAP induced TRPV1 activation through crosstalk.
Collapse
Affiliation(s)
- Alexander Lucius
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Noushafarin Khajavi
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Peter S Reinach
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou 325027, PR China
| | - Josef Köhrle
- Institut für Experimentelle Endokrinologie, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Priyavathi Dhandapani
- Gastroenterology, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Philipp Huimann
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Nina Ljubojevic
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Carsten Grötzinger
- Gastroenterology, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Stefan Mergler
- Klinik für Augenheilkunde, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
21
|
Jin Y. La(3+) Alters the Response Properties of Neurons in the Mouse Primary Somatosensory Cortex to Low-Temperature Noxious Stimulation of the Dental Pulp. BIOCHEMISTRY INSIGHTS 2015; 8:9-20. [PMID: 26604777 PMCID: PMC4640426 DOI: 10.4137/bci.s30752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 10/12/2015] [Accepted: 10/14/2015] [Indexed: 12/31/2022]
Abstract
Although dental pain is a serious health issue with high incidence among the human population, its cellular and molecular mechanisms are still unclear. Transient receptor potential (TRP) channels are assumed to be involved in the generation of dental pain. However, most of the studies were conducted with molecular biological or histological methods. In vivo functional studies on the role of TRP channels in the mechanisms of dental pain are lacking. This study uses in vivo cellular electrophysiological and neuropharmacological method to directly disclose the effect of LaCl3, a broad spectrum TRP channel blocker, on the response properties of neurons in the mouse primary somatosensory cortex to low-temperature noxious stimulation of the dental pulp. It was found that LaCl3 suppresses the high-firing-rate responses of all nociceptive neurons to noxious low-temperature stimulation and also inhibits the spontaneous activities in some nonnociceptive neurons. The effect of LaCl3 is reversible. Furthermore, this effect is persistent and stable unless LaCl3 is washed out. Washout of LaCl3 quickly revitalized the responsiveness of neurons to low-temperature noxious stimulation. This study adds direct evidence for the hypothesis that TRP channels are involved in the generation of dental pain and sensation. Blockade of TRP channels may provide a novel therapeutic treatment for dental pain.
Collapse
Affiliation(s)
- Yanjiao Jin
- Department of Stomatology, Tianjin Medical University General Hospital, Heping District, Tianjin, People's Republic of China
| |
Collapse
|
22
|
Liu T, Fang Z, Wang G, Shi M, Wang X, Jiang K, Yang Z, Cao R, Tao H, Wang X, Zhou J. Anti-tumor activity of the TRPM8 inhibitor BCTC in prostate cancer DU145 cells. Oncol Lett 2015; 11:182-188. [PMID: 26870186 PMCID: PMC4727066 DOI: 10.3892/ol.2015.3854] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 09/09/2015] [Indexed: 11/20/2022] Open
Abstract
The present study investigated the anti-tumor activity of N-(4-tertiarybutylphenyl)-4-(3-chloropyridin-2-yl)tetrahydropyrazine-1(2H)-carbox-amide (BCTC), a potent and specific inhibitor of transient receptor potential cation channel subfamily M member 8 (TRPM8) in prostate cancer (PCa) DU145 cells. TRPM8 expression in DU145 and normal prostate PNT1A cells was detected by reverse transcription polymerase chain reaction and western blot analysis. The effect of BCTC on DU145 cells was analyzed by flow cytometry analysis, and MTT, scratch motility and Transwell invasion assays. The molecular mechanism through which BCTC acts was investigated by western blot analysis. TRPM8 expression was increased in DU145 cells compared with PNT1A cells at the mRNA and protein levels. The present study provided evidence that inhibition of TRPM8 by BCTC reduced the viability of DU145 cells, but not PNT1A cells. In addition, BCTC inhibited cell cycle progression, migration and invasion in DU145 cells. Cell cycle-associated proteins, including phosphorylated protein kinase B, cyclin D1, cyclin dependent kinase (CDK) 2 and CDK6 were downregulated by BCTC, while phosphorylated glycogen synthase kinase 3β was upregulated. However, investigations in the present study revealed that BCTC failed to trigger apoptosis in DU145 cells. In addition, in BCTC-treated DU145 cells, phosphorylated extracellular signal-regulated kinase 1/2 was downregulated substantially while phosphorylated p38 (p-p38) and phosphorylated c-Jun N-terminal kinases (p-JNK) were upregulated. The anti-proliferative activity of BCTC on DU145 cells was attenuated by p38 and JNK-specific inhibitors, suggesting that MAPK pathways are involved. Overall, the TRPM8 specific antagonist BCTC demonstrated excellent anti-tumor activity in PCa DU145 cells, and therefore has the potential to become a targeted therapeutic strategy against PCa.
Collapse
Affiliation(s)
- Tao Liu
- Department of Urology, Jingzhou Central Hospital, Jingzhou, Hubei 434020, P.R. China
| | - Zhihai Fang
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Gang Wang
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Mingjun Shi
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Xiao Wang
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Kun Jiang
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zhonghua Yang
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Rui Cao
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Huangheng Tao
- Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Jiajie Zhou
- Department of Urology, Jingzhou Central Hospital, Jingzhou, Hubei 434020, P.R. China
| |
Collapse
|
23
|
Dinter J, Khajavi N, Mühlhaus J, Wienchol CL, Cöster M, Hermsdorf T, Stäubert C, Köhrle J, Schöneberg T, Kleinau G, Mergler S, Biebermann H. The Multitarget Ligand 3-Iodothyronamine Modulates β-Adrenergic Receptor 2 Signaling. Eur Thyroid J 2015; 4:21-9. [PMID: 26601070 PMCID: PMC4640289 DOI: 10.1159/000381801] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 03/19/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND 3-Iodothyronamine (3-T1AM), a signaling molecule with structural similarities to thyroid hormones, induces numerous physiological responses including reversible body temperature decline. One target of 3-T1AM is the trace amine-associated receptor 1 (TAAR1), which is a member of the rhodopsin-like family of G protein-coupled receptors (GPCRs). Interestingly, the effects of 3-T1AM remain detectable in TAAR1 knockout mice, suggesting further targets for 3-T1AM such as adrenergic receptors. Therefore, we evaluated whether β-adrenergic receptor 1 (ADRB1) and 2 (ADRB2) signaling is affected by 3-T1AM in HEK293 cells and in human conjunctival epithelial cells (IOBA-NHC), where these receptors are highly expressed endogenously. METHODS A label-free EPIC system for prescreening the 3-T1AM-induced effects on ADRB1 and ADRB2 in transfected HEK293 cells was used. In addition, ADRB1 and ADRB2 activation was analyzed using a cyclic AMP assay and a MAPK reporter gene assay. Finally, fluorescence Ca(2+) imaging was utilized to delineate 3-T1AM-induced Ca(2+) signaling. RESULTS 3-T1AM (10(-5)-10(-10)M) enhanced isoprenaline-induced ADRB2-mediated Gs signaling but not that of ADRB1-mediated signaling. MAPK signaling remained unaffected for both receptors. In IOBA-NHC cells, norepinephrine-induced Ca(2+) influxes were blocked by the nonselective ADRB blocker timolol (10 µM), indicating that ADRBs are most likely linked with Ca(2+) channels. Notably, timolol was also found to block 3-T1AM (10(-5)M)-induced Ca(2+) influx. CONCLUSIONS The presented data support that 3-T1AM directly modulates β-adrenergic receptor signaling. The relationship between 3-T1AM and β-adrenergic signaling also reveals a potential therapeutic value for suppressing Ca(2+) channel-mediated inflammation processes, occurring in eye diseases such as conjunctivitis.
Collapse
Affiliation(s)
- Juliane Dinter
- Institut für Experimentelle Pädiatrische Endokrinologie, Berlin, Germany
| | - Noushafarin Khajavi
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jessica Mühlhaus
- Institut für Experimentelle Pädiatrische Endokrinologie, Berlin, Germany
| | | | - Maxi Cöster
- Institut für Biochemie, Molekulare Biochemie, Medizinische Fakultät, University of Leipzig, Leipzig, Germany
| | - Thomas Hermsdorf
- Institut für Biochemie, Molekulare Biochemie, Medizinische Fakultät, University of Leipzig, Leipzig, Germany
| | - Claudia Stäubert
- Institut für Biochemie, Molekulare Biochemie, Medizinische Fakultät, University of Leipzig, Leipzig, Germany
| | - Josef Köhrle
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Torsten Schöneberg
- Institut für Biochemie, Molekulare Biochemie, Medizinische Fakultät, University of Leipzig, Leipzig, Germany
| | - Gunnar Kleinau
- Institut für Experimentelle Pädiatrische Endokrinologie, Berlin, Germany
| | - Stefan Mergler
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Heike Biebermann
- Institut für Experimentelle Pädiatrische Endokrinologie, Berlin, Germany
- *Heike Biebermann, Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, DE-13353 Berlin (Germany), E-Mail , Stefan Mergler, Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, DE-13353 Berlin (Germany), E-Mail
| |
Collapse
|
24
|
Teichmann J, Nitschke M, Pette D, Valtink M, Gramm S, Härtel FV, Noll T, Funk RHW, Engelmann K, Werner C. Thermo-responsive cell culture carriers based on poly(vinyl methyl ether)-the effect of biomolecular ligands to balance cell adhesion and stimulated detachment. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2015; 16:045003. [PMID: 27877823 PMCID: PMC5090182 DOI: 10.1088/1468-6996/16/4/045003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 06/19/2015] [Accepted: 06/22/2015] [Indexed: 06/06/2023]
Abstract
Two established material systems for thermally stimulated detachment of adherent cells were combined in a cross-linked polymer blend to merge favorable properties. Through this approach poly(N-isopropylacrylamide) (PNiPAAm) with its superior switching characteristic was paired with a poly(vinyl methyl ether)-based composition that allows adjusting physico-chemical and biomolecular properties in a wide range. Beyond pure PNiPAAm, the proposed thermo-responsive coating provides thickness, stiffness and swelling behavior, as well as an apposite density of reactive sites for biomolecular functionalization, as effective tuning parameters to meet specific requirements of a particular cell type regarding initial adhesion and ease of detachment. To illustrate the strength of this approach, the novel cell culture carrier was applied to generate transplantable sheets of human corneal endothelial cells (HCEC). Sheets were grown, detached, and transferred onto planar targets. Cell morphology, viability and functionality were analyzed by immunocytochemistry and determination of transepithelial electrical resistance (TEER) before and after sheet detachment and transfer. HCEC layers showed regular morphology with appropriate TEER. Cells were positive for function-associated marker proteins ZO-1, Na+/K+-ATPase, and paxillin, and extracellular matrix proteins fibronectin, laminin and collagen type IV before and after transfer. Sheet detachment and transfer did not impair cell viability. Subsequently, a potential application in ophthalmology was demonstrated by transplantation onto de-endothelialized porcine corneas in vitro. The novel thermo-responsive cell culture carrier facilitates the generation and transfer of functional HCEC sheets. This paves the way to generate tissue engineered human corneal endothelium as an alternative transplant source for endothelial keratoplasty.
Collapse
Affiliation(s)
- Juliane Teichmann
- Institute of Anatomy, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, D-01307 Dresden, Germany
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, Hohe Straße 6, D-01069 Dresden, Germany
| | - Mirko Nitschke
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, Hohe Straße 6, D-01069 Dresden, Germany
| | - Dagmar Pette
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, Hohe Straße 6, D-01069 Dresden, Germany
| | - Monika Valtink
- Institute of Anatomy, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, D-01307 Dresden, Germany
| | - Stefan Gramm
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, Hohe Straße 6, D-01069 Dresden, Germany
- Herlac Coswig GmbH, Industriestraße 28, D-01640 Coswig, Germany
| | - Frauke V Härtel
- Institute of Physiology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, D-01307 Dresden, Germany
| | - Thomas Noll
- Institute of Physiology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, D-01307 Dresden, Germany
| | - Richard H W Funk
- Institute of Anatomy, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, D-01307 Dresden, Germany
- CRTD/DFG-Center for Regenerative Therapies Dresden—Cluster of Excellence, Fetscherstraße 105, D-01307 Dresden, Germany
| | - Katrin Engelmann
- Institute of Anatomy, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, D-01307 Dresden, Germany
- CRTD/DFG-Center for Regenerative Therapies Dresden—Cluster of Excellence, Fetscherstraße 105, D-01307 Dresden, Germany
- Department of Ophthalmology, Klinikum Chemnitz gGmbH, Flemmingstraße 2, D-09116 Chemnitz, Germany
| | - Carsten Werner
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, Hohe Straße 6, D-01069 Dresden, Germany
- CRTD/DFG-Center for Regenerative Therapies Dresden—Cluster of Excellence, Fetscherstraße 105, D-01307 Dresden, Germany
| |
Collapse
|
25
|
Reinach PS, Chen W, Mergler S. Polymodal roles of transient receptor potential channels in the control of ocular function. EYE AND VISION 2015; 2:5. [PMID: 26605361 PMCID: PMC4655450 DOI: 10.1186/s40662-015-0016-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 02/15/2015] [Indexed: 12/05/2022]
Abstract
Maintenance of intracellular Ca2+ levels at orders of magnitude below those in the extracellular environment is a requisite for preserving cell viability. Membrane channels contribute to such control through modulating their time-dependent opening and closing behaviour. Such regulation requires Ca2+ to serve as a second messenger mediating receptor control of numerous life-sustaining responses. Transient receptor potential (TRP) channels signal transduce a wide variety of different sensory stimuli to induce responses modulating cellular function. These channels are non-selective cation channels with variable Ca2+ selectivity having extensive sequence homology. They constitute a superfamily made up of 28 different members that are subdivided into 7 different subfamilies based on differences in sequence homology. Some of these TRP channel isotypes are expressed in the eye and localized to both neuronal and non-neuronal cell membranes. Their activation generates intracellular Ca2+ transients and other downstream-linked signalling events that affect numerous responses required for visual function. As there is an association between changes in functional TRP expression in various ocular diseases, there are efforts underway to determine if these channels can be used as drug targets to reverse declines in ocular function. We review here our current knowledge about the expression, function and regulation of TRPs in different eye tissues in health and disease. Furthermore, some of the remaining hurdles are described to developing safe and efficacious TRP channel modulators for use in a clinical setting.
Collapse
Affiliation(s)
- Peter S Reinach
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang 325027 P.R. China
| | - Weiwei Chen
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang 325027 P.R. China
| | - Stefan Mergler
- Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Klinik für Augenheilkunde, Augustenburger Platz 1, D-13353 Berlin, Germany
| |
Collapse
|
26
|
The involvement of mitochondrial apoptotic pathway in eugenol-induced cell death in human glioblastoma cells. Toxicol Lett 2015; 232:122-32. [DOI: 10.1016/j.toxlet.2014.10.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 09/30/2014] [Accepted: 10/15/2014] [Indexed: 12/27/2022]
|
27
|
Khajavi N, Reinach PS, Slavi N, Skrzypski M, Lucius A, Strauß O, Köhrle J, Mergler S. Thyronamine induces TRPM8 channel activation in human conjunctival epithelial cells. Cell Signal 2014; 27:315-25. [PMID: 25460045 DOI: 10.1016/j.cellsig.2014.11.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 10/31/2014] [Accepted: 11/12/2014] [Indexed: 11/16/2022]
Abstract
3-Iodothyronamine (T1AM), an endogenous thyroid hormone (TH) metabolite, induces numerous responses including a spontaneously reversible body temperature decline. As such an effect is associated in the eye with increases in basal tear flow and thermosensitive transient receptor potential melastatin 8 (TRPM8) channel activation, we determined in human conjunctival epithelial cells (IOBA-NHC) if T1AM also acts as a cooling agent to directly affect TRPM8 activation at a constant temperature. RT-PCR and quantitative real-time PCR (qPCR) along with immunocytochemistry probed for TRPM8 gene and protein expression whereas functional activity was evaluated by comparing the effects of T1AM with those of TRPM8 mediators on intracellular Ca(2+) ([Ca(2+)]i) and whole-cell currents. TRPM8 gene and protein expression was evident and icilin (20μM), a TRPM8 agonist, increased Ca(2+) influx as well as whole-cell currents whereas BCTC (10μM), a TRPM8 antagonist, suppressed these effects. Similarly, either temperature lowering below 23°C or T1AM (1μM) induced Ca(2+) transients that were blocked by this antagonist. TRPM8 activation by both 1µM T1AM and 20μM icilin prevented capsaicin (CAP) (20μM) from inducing increases in Ca(2+) influx through TRP vanilloid 1 (TRPV1) activation, whereas BCTC did not block this response. CAP (20μM) induced a 2.5-fold increase in IL-6 release whereas during exposure to 20μM capsazepine this rise was completely blocked. Similarly, T1AM (1μM) prevented this response. Taken together, T1AM like icilin is a cooling agent since they both directly elicit TRPM8 activation at a constant temperature. Moreover, there is an inverse association between changes in TRPM8 and TRPV1 activity since these cooling agents blocked both CAP-induced TRPV1 activation and downstream rises in IL-6 release.
Collapse
Affiliation(s)
- Noushafarin Khajavi
- Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Klinik für Augenheilkunde, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Peter S Reinach
- Biological Sciences, SUNY College of Optometry, New York, NY 10036, USA; School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou 325027, PR China
| | - Nefeli Slavi
- Biological Sciences, SUNY College of Optometry, New York, NY 10036, USA
| | - Marek Skrzypski
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, 60-637 Poznań, Poland
| | - Alexander Lucius
- Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Klinik für Augenheilkunde, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Olaf Strauß
- Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Klinik für Augenheilkunde, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Josef Köhrle
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Stefan Mergler
- Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Klinik für Augenheilkunde, Augustenburger Platz 1, D-13353 Berlin, Germany.
| |
Collapse
|
28
|
Mergler S, Valtink M, Takayoshi S, Okada Y, Miyajima M, Saika S, Reinach PS. Temperature-sensitive transient receptor potential channels in corneal tissue layers and cells. Ophthalmic Res 2014; 52:151-9. [PMID: 25301091 DOI: 10.1159/000365334] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 06/16/2014] [Indexed: 11/19/2022]
Abstract
We here provide a brief summary of the characteristics of transient receptor potential channels (TRPs) identified in corneal tissue layers and cells. In general, TRPs are nonselective cation channels which are Ca(2+) permeable. Most TRPs serve as thermosensitive molecular sensors (thermo-TRPs). Based on their functional importance, the possibilities are described for drug-targeting TRP activity in a clinical setting. TRPs are expressed in various tissues of the eye including both human corneal epithelial and endothelial layers as well as stromal fibroblasts and stromal nerve fibers. TRP vanilloid type 1 (TRPV1) heat receptor, also known as capsaicin receptor, along with TRP melastatin type 8 (TRPM8) cold receptor, which is also known as menthol receptor, are prototypes of the thermo-TRP family. The TRPV1 functional channel is the most investigated TRP channel in these tissues, owing to its contribution to maintaining tissue homeostasis as well as eliciting wound healing responses to injury. Other thermo-TRP family members identified in these tissues are TRPV2, 3 and 4. Finally, there is the TRP ankyrin type 1 (TRPA1) cold receptor. All of these thermo-TRPs can be activated within specific temperature ranges and transduce such inputs into chemical and electrical signals. Although several recent studies have begun to unravel complex roles for thermo-TRPs such as TRPV1 in corneal layers and resident cells, additional studies are needed to further elucidate their roles in health and disease.
Collapse
Affiliation(s)
- Stefan Mergler
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
29
|
Permeation, regulation and control of expression of TRP channels by trace metal ions. Pflugers Arch 2014; 467:1143-64. [PMID: 25106481 PMCID: PMC4435931 DOI: 10.1007/s00424-014-1590-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/10/2014] [Accepted: 07/13/2014] [Indexed: 01/26/2023]
Abstract
Transient receptor potential (TRP) channels form a diverse family of cation channels comprising 28 members in mammals. Although some TRP proteins can only be found on intracellular membranes, most of the TRP protein isoforms reach the plasma membrane where they form ion channels and control a wide number of biological processes. There, their involvement in the transport of cations such as calcium and sodium has been well documented. However, a growing number of studies have started to expand our understanding of these proteins by showing that they also transport other biologically relevant metal ions like zinc, magnesium, manganese and cobalt. In addition to this newly recognized property, the activity and expression of TRP channels can be regulated by metal ions like magnesium, gadolinium, lanthanum or cisplatin. The aim of this review is to highlight the complex relationship between metal ions and TRP channels.
Collapse
|
30
|
Madrid R, Pertusa M. Intimacies and physiological role of the polymodal cold-sensitive ion channel TRPM8. CURRENT TOPICS IN MEMBRANES 2014; 74:293-324. [PMID: 25366241 DOI: 10.1016/b978-0-12-800181-3.00011-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The detection of environmental temperature is critical for the survival of the most diverse organisms. Thermosensitive transient receptor potential (thermoTRP) channels have evolved as a class of ion channels activated by a wide range of temperatures. These molecular thermal sensors are spread through the different TRP channel subfamilies. Among the Melastatin subfamily of TRP channels, the eighth member, TRPM8, is a calcium-permeable cationic ion channel activated by cold, by substances that evoke cold sensation such as menthol, and by voltage. This channel is considered the main molecular entity responsible for the sensitivity to cold of primary sensory neurons of the somatosensory system. Here we present to the readers a summary of some the most relevant biophysical properties, physiological role, and molecular intimacies of this polymodal thermoTRP channel.
Collapse
Affiliation(s)
- Rodolfo Madrid
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - María Pertusa
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|