1
|
Rocereta JA, Sturhahn T, Pumroy RA, Fricke TC, Herzog C, Leffler A, Moiseenkova-Bell V. Structural insights into TRPV2 modulation by probenecid. Nat Struct Mol Biol 2025:10.1038/s41594-025-01494-9. [PMID: 39972168 DOI: 10.1038/s41594-025-01494-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 01/22/2025] [Indexed: 02/21/2025]
Abstract
The transient receptor potential vanilloid 2 (TRPV2) cation channel is a key player in cardiovascular physiology and pathophysiology. Probenecid (PBC), an FDA-approved uricosuric agent thought to activate TRPV2, has shown promise in enhancing cardiovascular function in both preclinical and clinical studies. Here our electrophysiological data reveal that PBC significantly potentiates rat TRPV2 to known stimuli, and cryo electron microscopy structures show that PBC directly interacts with rat TRPV2 in a previously unidentified intracellular binding pocket. PBC binding at a conserved TRPV2-specific histidine prevents the channel from taking on the inactivated carboxyl-terminal conformation. This effect extends to TRPV1 and TRPV3 channels when glutamine is substituted with histidine at the corresponding position, increasing their sensitivity to PBC. While PBC alone does not induce TRPV2 opening, its combination with 2-aminoethoxydiphenyl borate enables the channel to adopt an intermediate, potentiated state. Our results offer insights into potential therapeutic advancements for TRPV2 through this pocket.
Collapse
Affiliation(s)
- Julia A Rocereta
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Toni Sturhahn
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Ruth A Pumroy
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tabea C Fricke
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Christine Herzog
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Andreas Leffler
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany.
| | - Vera Moiseenkova-Bell
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Jiang D, Guo R, Dai R, Knoedler S, Tao J, Machens HG, Rinkevich Y. The Multifaceted Functions of TRPV4 and Calcium Oscillations in Tissue Repair. Int J Mol Sci 2024; 25:1179. [PMID: 38256251 PMCID: PMC10816018 DOI: 10.3390/ijms25021179] [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: 11/29/2023] [Revised: 01/09/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
The transient receptor potential vanilloid 4 (TRPV4) specifically functions as a mechanosensitive ion channel and is responsible for conveying changes in physical stimuli such as mechanical stress, osmotic pressure, and temperature. TRPV4 enables the entry of cation ions, particularly calcium ions, into the cell. Activation of TRPV4 channels initiates calcium oscillations, which trigger intracellular signaling pathways involved in a plethora of cellular processes, including tissue repair. Widely expressed throughout the body, TRPV4 can be activated by a wide array of physicochemical stimuli, thus contributing to sensory and physiological functions in multiple organs. This review focuses on how TRPV4 senses environmental cues and thereby initiates and maintains calcium oscillations, critical for responses to organ injury, tissue repair, and fibrosis. We provide a summary of TRPV4-induced calcium oscillations in distinct organ systems, along with the upstream and downstream signaling pathways involved. In addition, we delineate current animal and disease models supporting TRPV4 research and shed light on potential therapeutic targets for modulating TRPV4-induced calcium oscillation to promote tissue repair while reducing tissue fibrosis.
Collapse
Affiliation(s)
- Dongsheng Jiang
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, 81377 Munich, Germany; (R.G.); (R.D.); (S.K.)
| | - Ruiji Guo
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, 81377 Munich, Germany; (R.G.); (R.D.); (S.K.)
- Department of Plastic and Hand Surgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany;
| | - Ruoxuan Dai
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, 81377 Munich, Germany; (R.G.); (R.D.); (S.K.)
| | - Samuel Knoedler
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, 81377 Munich, Germany; (R.G.); (R.D.); (S.K.)
- Department of Plastic and Hand Surgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany;
- Division of Plastic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02152, USA
| | - Jin Tao
- Department of Physiology and Neurobiology and Centre for Ion Channelopathy, Medical College of Soochow University, Suzhou 215123, China;
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou 215123, China
| | - Hans-Günther Machens
- Department of Plastic and Hand Surgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany;
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, 81377 Munich, Germany; (R.G.); (R.D.); (S.K.)
| |
Collapse
|
3
|
Rhee YH, Choi YH, Hu AC, Lee MY, Ahn JC, Kim S, Mo JH, Woo SH, Chung PS. Role of Transient Receptor Potential Vanilloid 1 in Sonic Hedgehog-Dependent Taste Bud Differentiation. LIFE (BASEL, SWITZERLAND) 2022; 13:life13010075. [PMID: 36676024 PMCID: PMC9862146 DOI: 10.3390/life13010075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/19/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022]
Abstract
Taste bud cell differentiation is extremely important for taste sensation. Immature taste bud cells cannot function during taste perception transmission to the nerve. In this study, we investigated whether hedgehog signaling affected taste bud cell differentiation and whether transient receptor potential vanilloid 1 (TRPV1) played a key role in dry mouth. The induction of dry mouth due to salivary gland resection (SGR) was confirmed on the basis of reduced salivation and disrupted fungiform papillae. The expression of keratin 8 (K8) of taste bud cells, neurofilament (NF), sonic hedgehog (Shh), and glioma-associated oncogene homolog 1 (Gli1) around taste bud cells was downregulated; however, the expression of TRPV1, P2X purinoceptor 3 (P2X3), and hematopoietic stem cell factor (c-Kit) was upregulated at the NF ends in the dry mouth group. To investigate the effect of TRPV1 defect on dry mouth, we induced dry mouth in the TRPV-/- group. The K8, NF, and P2X3 expression patterns were the same in the TRPV1 wild-type and TRPV1-/- dry mouth groups. However, Shh and c-Kit expression decreased regardless of dry mouth in the case of TRPV1 deficiency. These results indicated that TRPV1 positively regulated proliferation during taste bud cell injury by blocking the Shh/Gli1 pathway. In addition, not only cell proliferation but also differentiation of taste bud cells could not be regulated under TRPV1-deficiency conditions. Thus, TRPV1 positively regulates taste bud cell innervation and differentiation; this finding could be valuable in the clinical treatment of dry mouth-related taste dysfunction.
Collapse
Affiliation(s)
- Yun-Hee Rhee
- Beckman Laser Institute Korea, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Medical Laser Research Center, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Laser Translational Clinical Trial Center, Dankook University Hospital, Cheonan 31116, Republic of Korea
| | - Young-Hoon Choi
- Laser Translational Clinical Trial Center, Dankook University Hospital, Cheonan 31116, Republic of Korea
| | - Allison C. Hu
- Beckman Laser Institute and Medical Clinic, University of California Irvine, 1002 Health Sciences Rd., Irvine, CA 92697, USA
| | - Min Young Lee
- Beckman Laser Institute Korea, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Medical Laser Research Center, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Jin-Chul Ahn
- Beckman Laser Institute Korea, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Medical Laser Research Center, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
| | - Sehwan Kim
- Beckman Laser Institute Korea, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
| | - Ji-Hun Mo
- Beckman Laser Institute Korea, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Medical Laser Research Center, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Laser Translational Clinical Trial Center, Dankook University Hospital, Cheonan 31116, Republic of Korea
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Seung Hoon Woo
- Beckman Laser Institute Korea, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Medical Laser Research Center, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Laser Translational Clinical Trial Center, Dankook University Hospital, Cheonan 31116, Republic of Korea
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Phil-Sang Chung
- Beckman Laser Institute Korea, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Medical Laser Research Center, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Laser Translational Clinical Trial Center, Dankook University Hospital, Cheonan 31116, Republic of Korea
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Correspondence:
| |
Collapse
|
4
|
Van den Eynde C, Held K, Ciprietti M, De Clercq K, Kerselaers S, Marchand A, Chaltin P, Voets T, Vriens J. Loratadine, an antihistaminic drug, suppresses the proliferation of endometrial stromal cells by inhibition of TRPV2. Eur J Pharmacol 2022; 928:175086. [PMID: 35714693 DOI: 10.1016/j.ejphar.2022.175086] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 06/01/2022] [Accepted: 06/01/2022] [Indexed: 11/03/2022]
Abstract
The transient receptor potential (TRP) channel TRPV2 is widely expressed in a variety of different cell types and tissues. However, elucidating the exact biological functions of TRPV2 is significantly hampered by the lack of selective pharmacological tools to modulate channel activity in vitro and in vivo. This study aimed to identify new compounds that modify TRPV2 activity via the use of a plate-based calcium imaging approach to screen a drug repurposing library. Three antihistaminic drugs, loratadine, astemizole and clemizole were identified to reduce calcium-influx evoked by the TRPV2 agonist tetrahydrocannabivarin in HEK293 cells expressing murine TRPV2. Using single-cell calcium-microfluorimetry and whole-cell patch clamp recordings, we further confirmed that all three compounds induced a concentration-dependent block of TRPV2-mediated Ca2+ influx and whole-cell currents, with loratadine being the most potent antagonist of TRPV2. Moreover, this study demonstrated that loratadine was able to block both the human and mouse TRPV2 orthologs, without inhibiting the activity of other closely related members of the TRPV superfamily. Finally, loratadine inhibited TRPV2-dependent responses in a primary culture of mouse endometrial stromal cells and attenuated cell proliferation and migration in in vitro cell proliferation and wound healing assays. Taken together, our study revealed that the antihistaminic drugs loratadine, astemizole and clemizole target TRPV2 in a concentration-dependent manner. The identification of these antihistaminic drugs as blockers of TRPV2 may form a new starting point for the synthesis of more potent and selective TRPV2 antagonists, which could further lead to the unravelling of the physiological role of the channel.
Collapse
Affiliation(s)
- Charlotte Van den Eynde
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 box 611, 3000, Leuven, Belgium; 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
| | - Katharina Held
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 box 611, 3000, Leuven, Belgium; 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
| | - Martina Ciprietti
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 box 611, 3000, Leuven, Belgium; 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
| | - Katrien De Clercq
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 box 611, 3000, Leuven, Belgium; 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
| | - 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
| | - Arnaud Marchand
- CISTIM Leuven vzw, Gaston Geenslaan 2, 3001, Leuven, Heverlee, Belgium
| | - Patrick Chaltin
- CISTIM Leuven vzw, Gaston Geenslaan 2, 3001, Leuven, Heverlee, Belgium; Centre for Drug Design and Discovery (CD3), KU Leuven, Gaston Geenslaan 2, 3001, Leuven, Heverlee, Belgium
| | - 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
| | - Joris Vriens
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 box 611, 3000, Leuven, Belgium.
| |
Collapse
|
5
|
Oxidative Stress-Induced TRPV2 Expression Increase Is Involved in Diabetic Cataracts and Apoptosis of Lens Epithelial Cells in a High-Glucose Environment. Cells 2022; 11:cells11071196. [PMID: 35406761 PMCID: PMC8998065 DOI: 10.3390/cells11071196] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022] Open
Abstract
Cataracts are a serious complication of diabetes. In long-term hyperglycemia, intracellular Ca2+ concentration ([Ca2+]i) and reactive oxygen species (ROS) are increased. The apoptosis of lens epithelial cells plays a key role in the development of cataract. We investigated a potential role for transient receptor potential vanilloid 2 (TRPV2) in the development of diabetic cataracts. Immunohistochemical and Western blotting analyses showed that TRPV2 expression levels were significantly increased in the lens epithelial cells of patients with diabetic cataracts as compared with senile cataract, as well as in both a human lens epithelial cell line (HLEpiC) and primary rat lens epithelial cells (RLEpiCs) cultured under high-glucose conditions. The [Ca2+]i increase evoked by a TRPV2 channel agonist was significantly enhanced in both HLEpiCs and RLEpiCs cultured in high-glucose media. This enhancement was blocked by the TRPV2 nonspecific inhibitor ruthenium red and by TRPV2-specific small interfering (si)RNA transfection. Culturing HLEpiCs or RLEpiCs for seven days in high glucose significantly increased apoptosis, which was inhibited by TRPV2-specific siRNA transfection. In addition, ROS inhibitor significantly suppressed the ROS-induced increase of TRPV2-mediated Ca2+ signal and apoptosis under high-glucose conditions. These findings suggest a mechanism underlying high-glucose–induced apoptosis of lens epithelial cells, and offer a potential target for developing new therapeutic options for diabetes-related cataracts.
Collapse
|
6
|
Phillips KG, Turnbull IC, Hajjar RJ, Costa KD, Mayourian J. In silico Cell Therapy Model Restores Failing Human Myocyte Electrophysiology and Calcium Cycling in Fibrotic Myocardium. Front Physiol 2022; 12:755881. [PMID: 35046835 PMCID: PMC8762340 DOI: 10.3389/fphys.2021.755881] [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: 08/09/2021] [Accepted: 11/22/2021] [Indexed: 11/28/2022] Open
Abstract
Myocardial delivery of human c-kit+ cardiac interstitial cells (hCICs) and human mesenchymal stem cells (hMSCs), an emerging approach for treating the failing heart, has been limited by an incomplete understanding of the effects on host myocardium. This computational study aims to model hCIC and hMSC effects on electrophysiology and calcium cycling of healthy and diseased human cardiomyocytes (hCM), and reveals a possible cardiotherapeutic benefit independent of putative regeneration processes. First, we developed an original hCIC mathematical model with an electrical profile comprised of distinct experimentally identified ion currents. Next, we verified the model by confirming it is representative of published experiments on hCIC whole-cell electrophysiology and on hCIC co-cultures with rodent cardiomyocytes. We then used our model to compare electrophysiological effects of hCICs to other non-excitable cells, as well as clinically relevant hCIC-hMSC combination therapies and fused hCIC-hMSC CardioChimeras. Simulation of direct coupling of hCICs to healthy or failing hCMs through gap junctions led to greater increases in calcium cycling with lesser reductions in action potential duration (APD) compared with hMSCs. Combined coupling of hCICs and hMSCs to healthy or diseased hCMs led to intermediate effects on electrophysiology and calcium cycling compared to individually coupled hCICs or hMSCs. Fused hCIC-hMSC CardioChimeras decreased healthy and diseased hCM APD and calcium transient amplitude compared to individual or combined cell treatments. Finally, to provide a theoretical basis for optimizing cell-based therapies, we randomized populations of 2,500 models incorporating variable hMSC and hCIC interventions and simulated their effects on restoring diseased cardiomyocyte electrophysiology and calcium handling. The permutation simulation predicted the ability to correct abnormal properties of heart failure hCMs in fibrotic, but not non-fibrotic, myocardium. This permutation experiment also predicted paracrine signaling to be a necessary and sufficient mechanism for this correction, counteracting the fibrotic effects while also restoring arrhythmia-related metrics such as upstroke velocity and resting membrane potential. Altogether, our in silico findings suggest anti-fibrotic effects of paracrine signaling are critical to abrogating pathological cardiomyocyte electrophysiology and calcium cycling in fibrotic heart failure, and support further investigation of delivering an optimized cellular secretome as a potential strategy for improving heart failure therapy.
Collapse
Affiliation(s)
- Katherine G. Phillips
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Irene C. Turnbull
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Kevin D. Costa
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Joshua Mayourian
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
- Department of Pediatrics, Boston University, Boston, MA, United States
- Department of Pediatrics, Boston Medical Center, Boston, MA, United States
| |
Collapse
|
7
|
Canales Coutiño B, Mayor R. Reprint of: Mechanosensitive ion channels in cell migration. Cells Dev 2021; 168:203730. [PMID: 34456177 DOI: 10.1016/j.cdev.2021.203730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 11/18/2022]
Abstract
Cellular processes are initiated and regulated by different stimuli, including mechanical forces. Cell membrane mechanosensors represent the first step towards the conversion of mechanical stimuli to a biochemical or electrical response. Mechanosensitive (MS) ion channels form a growing family of ion gating channels that respond to direct physical force or plasma membrane deformations. A number of calcium (Ca2+) permeable MS channels are known to regulate the initiation, direction, and persistence of cell migration during development and tumour progression. While the evidence that links individual MS ion channels to cell migration is growing, a unified analysis of the molecular mechanisms regulated downstream of MS ion channel activation is lacking. In this review, we describe the MS ion channel families known to regulate cell migration. We discuss the molecular mechanisms that act downstream of MS ion channels with an emphasis on Ca2+ mediated processes. Finally, we propose the future directions and impact of MS ion channel activity in the field of cell migration.
Collapse
Affiliation(s)
- Brenda Canales Coutiño
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
8
|
Wu C, Zhou XX, Li JZ, Qiang HF, Wang Y, Li G. Pretreatment of cardiac progenitor cells with bradykinin attenuates H 2O 2-induced cell apoptosis and improves cardiac function in rats by regulating autophagy. Stem Cell Res Ther 2021; 12:437. [PMID: 34353364 PMCID: PMC8340370 DOI: 10.1186/s13287-021-02503-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 07/01/2021] [Indexed: 02/07/2023] Open
Abstract
Background Previous studies have demonstrated that human cardiac c-Kit+ progenitor cells (hCPCs) can effectively improve ischemic heart disease. However, the major challenge in applying hCPCs to clinical therapy is the low survival rate of graft hCPCs in the host heart, which limited the benefit of transplanted hCPCs. Bradykinin (BK) is a principal active agent of the tissue kinin-kallikrein system. Our previous studies have highlighted that BK mediated the growth and migration of CPCs by regulating Ca2+ influx. However, the protective effect of BK on CPCs, improvement in the survival rate of BK-pretreated hCPCs in the infarcted heart, and the related mechanism remain elusive. Methods HCPCs were treated with H2O2 to induce cell apoptosis and autophagy, and different concentration of BK was applied to rescue the H2O2-induced injury detected by MTT assay, TUNEL staining, flow cytometry, western blotting, and mitoSOX assays. The role of autophagy in the anti-apoptotic effect of BK was chemically activated or inhibited using the autophagy inducer, rapamycin, or the inhibitor, 3-methyladenine (3-MA). To explore the protective effect of BK on hCPCs, 3-MA or BK-pretreated hCPCs were transplanted into the myocardial infarcted rats. An echocardiogram was used to determine cardiac function, H&E and Masson staining were employed to assess pathological characteristics, HLA gene expression was quantified by qRT-PCR, and immunostaining was applied to examine neovascularization using confocal microscopy. Results The in vitro results showed that BK suppressed H2O2-induced hCPCs apoptosis and ROS production in a concentration-dependent manner by promoting pAkt and Bcl-2 expression and reducing cleaved caspase 3 and Bax expression. Moreover, BK restrained the H2O2-induced cell autophagy by decreasing LC3II/I, Beclin1, and ATG5 expression and increasing P62 expression. In the in vivo experiment, the transplanted BK- or 3-MA-treated hCPCs were found to be more effectively improved cardiac function by decreasing cardiomyocyte apoptosis, inflammatory infiltration, and myocardial fibrosis, and promoting neovascularization in the infarcted heart, compared to untreated-hCPCs or c-kit- cardiomyocytes (CPC- cells). Conclusions Our present study established a new method to rescue transplanted hCPCs in the infarcted cardiac area via regulating cell apoptosis and autophagy of hCPCs by pretreatment with BK, providing a new therapeutic option for heart failure. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02503-6.
Collapse
Affiliation(s)
- Chan Wu
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, 361015, Fujian, China
| | - Xiao-Xia Zhou
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, 361015, Fujian, China
| | - Jing-Zhou Li
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, 361015, Fujian, China
| | - Hai-Feng Qiang
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, 361015, Fujian, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, 361015, Fujian, China.
| | - Gang Li
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, 361015, Fujian, China.
| |
Collapse
|
9
|
Canales Coutiño B, Mayor R. Mechanosensitive ion channels in cell migration. Cells Dev 2021; 166:203683. [PMID: 33994356 PMCID: PMC8240554 DOI: 10.1016/j.cdev.2021.203683] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 01/05/2023]
Abstract
Cellular processes are initiated and regulated by different stimuli, including mechanical forces. Cell membrane mechanosensors represent the first step towards the conversion of mechanical stimuli to a biochemical or electrical response. Mechanosensitive (MS) ion channels form a growing family of ion gating channels that respond to direct physical force or plasma membrane deformations. A number of calcium (Ca2+) permeable MS channels are known to regulate the initiation, direction, and persistence of cell migration during development and tumour progression. While the evidence that links individual MS ion channels to cell migration is growing, a unified analysis of the molecular mechanisms regulated downstream of MS ion channel activation is lacking. In this review, we describe the MS ion channel families known to regulate cell migration. We discuss the molecular mechanisms that act downstream of MS ion channels with an emphasis on Ca2+ mediated processes. Finally, we propose the future directions and impact of MS ion channel activity in the field of cell migration.
Collapse
Affiliation(s)
- Brenda Canales Coutiño
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
10
|
O'Connor B, Robbins N, Koch SE, Rubinstein J. TRPV2 channel-based therapies in the cardiovascular field. Molecular underpinnings of clinically relevant therapies. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2020; 159:118-125. [PMID: 32565182 DOI: 10.1016/j.pbiomolbio.2020.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 12/11/2022]
Abstract
The transient receptor potential (TRP) ion channel family is composed of twenty-seven channel proteins that are ubiquitously expressed in the human body. The TRPV (vanilloid) subfamily has been a recent target of investigation within the cardiovascular field. TRPV1, which is sensitive to heat as well as vanilloids, is the best characterized TRPV channel and is the namesake for the subfamily that includes six members. Research into the function of TRPV2 has suggested that it plays an important role in cardiovascular function. Over the last twenty years a greater understanding of the differences among the TRPV channels has allowed for more precise experimentation and has opened various translational opportunities. TRPV2 has been found to be a both a mechanosensor and a mediator of calcium handling and has been found to play important roles in healthy and diseased cardiomyocytes. These roles have been translated into clinical studies in patients with muscular dystrophy (both agonism and antagonism) as well as in patients with cardiomyopathy and heart failure with reduced ejection fraction. Its role as a structural protein has also been elucidated, though the clinical significance of this finding has yet to be established. Despite the clinical progress that has been made there is still a need for large, prospective randomized studies with TRPV2 channel agonists and antagonists in order to bring these basic and translational science findings to the bedside.
Collapse
Affiliation(s)
- Brian O'Connor
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Nathan Robbins
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Sheryl E Koch
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Jack Rubinstein
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
11
|
Iwamoto M, Takashima M, Ohtubo Y. A subset of taste receptor cells express biocytin-permeable channels activated by reducing extracellular Ca 2+ concentration. Eur J Neurosci 2020; 51:1605-1623. [PMID: 31912931 DOI: 10.1111/ejn.14672] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 12/03/2019] [Accepted: 01/02/2020] [Indexed: 12/11/2022]
Abstract
Taste receptor cells (type II cells) transmit taste information to taste nerve fibres via ATP-permeable channels, including calcium homeostasis modulator (CALHM), connexin and/or pannexin1 channels, via the paracrine release of adenosine triphosphate (ATP) as a predominant transmitter. In the present study, we demonstrate that extracellular Ca2+ -dependent biocytin-permeable channels are present in a subset of type II cells in mouse fungiform taste buds using biocytin uptake, immunohistochemistry and in situ whole-cell recordings. Type II cells were labelled with biocytin in an extracellular Ca2+ concentration ([Ca2+ ]out )-sensitive manner. We found that the ratio of biocytin-labelled type II cells to type II cells per taste bud was approximately 20% in 2 mM Ca2+ saline, and this ratio increased to approximately 50% in nominally Ca2+ -free saline. The addition of 300 µM GdCl3 , which inhibits various channels including CALHM1 channels, significantly inhibited biocytin labelling in nominally Ca2+ -free saline, whereas the addition of 20 µM ruthenium red did not. Moreover, Cs+ -insensitive currents increased in nominally Ca2+ -free saline in approximately 40% of type II cells. These increased currents appeared at a potential of above -35 mV, reversed at approximately +10 mV and increased with depolarization. These results suggest that biocytin labels type II cells via ion channels activated by [Ca2+ ]out reduction, probably "CALHM-like" channels, on the basolateral membrane and that taste receptor cells can be categorized into two groups based on differences in the expression levels of [Ca2+ ]out -dependent biocytin-permeable channels. These data indicate electrophysiological and pharmacologically relevant properties of biocytin-permeable channels and suggest their contributions to taste signal transduction.
Collapse
Affiliation(s)
- Masafumi Iwamoto
- Graduate school of Life Science and Systems Engineering, Kyushu Institute of Technology, Kitakyushu-shi, Japan
| | - Madoka Takashima
- Graduate school of Life Science and Systems Engineering, Kyushu Institute of Technology, Kitakyushu-shi, Japan
| | - Yoshitaka Ohtubo
- Graduate school of Life Science and Systems Engineering, Kyushu Institute of Technology, Kitakyushu-shi, Japan
| |
Collapse
|
12
|
O'Reilly D, Buchanan P. Calcium channels and cancer stem cells. Cell Calcium 2019; 81:21-28. [PMID: 31163289 DOI: 10.1016/j.ceca.2019.05.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/21/2019] [Accepted: 05/21/2019] [Indexed: 12/24/2022]
Abstract
Cancer stem cells (CSC's) have emerged as a key area of investigation due to associations with cancer development and treatment resistance, related to their ability to remain quiescent, self-renew and terminally differentiate. Targeting CSC's in addition to the tumour bulk could ensure complete removal of the cancer, lessening the risk of relapse and improving patient survival. Understanding the mechanisms supporting the functions of CSC's is essential to highlight targets for the development of therapeutic strategies. Changes in intracellular calcium through calcium channel activity is fundamental for integral cellular processes such as proliferation, migration, differentiation and survival in a range of cell types, under both normal and pathological conditions. Here in we highlight how calcium channels represent a key mechanism involved in CSC function. It is clear that expression and or function of a number of channels involved in calcium entry and intracellular store release are altered in CSC's. Correlating with aberrant proliferation, self-renewal and differentiation, which in turn promoted cancer progression and treatment resistance. Research outlined has demonstrated that targeting altered calcium channels in CSC populations can reduce their stem properties and induce terminal differentiation, sensitising them to existing cancer treatments. Overall this highlights calcium channels as emerging novel targets for CSC therapies.
Collapse
Affiliation(s)
- Debbie O'Reilly
- National Institute of Cellular Biotechnology, Dublin City University, Dublin, Ireland; School of Nursing and Human science, Dublin City University, Dublin, Ireland
| | - Paul Buchanan
- National Institute of Cellular Biotechnology, Dublin City University, Dublin, Ireland; School of Nursing and Human science, Dublin City University, Dublin, Ireland.
| |
Collapse
|
13
|
Luo H, Rossi E, Saubamea B, Chasseigneaux S, Cochois V, Choublier N, Smirnova M, Glacial F, Perrière N, Bourdoulous S, Smadja DM, Menet MC, Couraud PO, Cisternino S, Declèves X. Cannabidiol Increases Proliferation, Migration, Tubulogenesis, and Integrity of Human Brain Endothelial Cells through TRPV2 Activation. Mol Pharm 2019; 16:1312-1326. [PMID: 30721081 DOI: 10.1021/acs.molpharmaceut.8b01252] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The effect of cannabidiol (CBD), a high-affinity agonist of the transient receptor potential vanilloid-2 (TRPV2) channel, has been poorly investigated in human brain microvessel endothelial cells (BMEC) forming the blood-brain barrier (BBB). TRPV2 expression and its role on Ca2+ cellular dynamics, trans-endothelial electrical resistance (TEER), cell viability and growth, migration, and tubulogenesis were evaluated in human primary cultures of BMEC (hPBMEC) or in the human cerebral microvessel endothelial hCMEC/D3 cell line. Abundant TRPV2 expression was measured in hCMEC/D3 and hPBMEC by qRT-PCR, Western blotting, nontargeted proteomics, and cellular immunofluorescence studies. Intracellular Ca2+ levels were increased by heat and CBD and blocked by the nonspecific TRP antagonist ruthenium red (RR) and the selective TRPV2 inhibitor tranilast (TNL) or by silencing cells with TRPV2 siRNA. CBD dose-dependently induced the hCMEC/D3 cell number (EC50 0.3 ± 0.1 μM), and this effect was fully abolished by TNL or TRPV2 siRNA. A wound healing assay showed that CBD induced cell migration, which was also inhibited by TNL or TRPV2 siRNA. Tubulogenesis of hCMEC/D3 cells in 3D matrigel cultures was significantly increased by 41 and 73% after a 7 or 24 h CBD treatment, respectively, and abolished by TNL. CBD also increased the TEER of hPBMEC monolayers cultured in transwell, and this was blocked by TNL. Our results show that CBD, at extracellular concentrations close to those observed in plasma of patients treated by CBD, induces proliferation, migration, tubulogenesis, and TEER increase in human brain endothelial cells, suggesting CBD might be a potent target for modulating the human BBB.
Collapse
Affiliation(s)
- Huilong Luo
- Inserm , U1144 , Paris F-75006 , France.,Université Paris Descartes , UMR-S 1144 , Paris F-75006 , France.,Université Paris Descartes , Sorbonne Paris Cité , Paris F-75006 , France
| | - Elisa Rossi
- Université Paris Descartes , Sorbonne Paris Cité , Paris F-75006 , France.,Université Paris Descartes , UMR-S 1140 , Paris F-75006 , France
| | - Bruno Saubamea
- Inserm , U1144 , Paris F-75006 , France.,Université Paris Descartes , UMR-S 1144 , Paris F-75006 , France.,Université Paris Descartes , Sorbonne Paris Cité , Paris F-75006 , France
| | - Stéphanie Chasseigneaux
- Inserm , U1144 , Paris F-75006 , France.,Université Paris Descartes , UMR-S 1144 , Paris F-75006 , France.,Université Paris Descartes , Sorbonne Paris Cité , Paris F-75006 , France
| | - Véronique Cochois
- Inserm , U1144 , Paris F-75006 , France.,Université Paris Descartes , UMR-S 1144 , Paris F-75006 , France.,Université Paris Descartes , Sorbonne Paris Cité , Paris F-75006 , France
| | - Nina Choublier
- Inserm , U1144 , Paris F-75006 , France.,Université Paris Descartes , UMR-S 1144 , Paris F-75006 , France.,Université Paris Descartes , Sorbonne Paris Cité , Paris F-75006 , France
| | - Maria Smirnova
- Inserm , U1144 , Paris F-75006 , France.,Université Paris Descartes , UMR-S 1144 , Paris F-75006 , France.,Université Paris Descartes , Sorbonne Paris Cité , Paris F-75006 , France
| | | | | | - Sandrine Bourdoulous
- Université Paris Descartes , Sorbonne Paris Cité , Paris F-75006 , France.,Department of Infection, Institut Cochin , Inserm, U1016 , Paris F-75014 , France.,CNRS, UMR 8104 , Paris F-75014 , France
| | - David M Smadja
- Université Paris Descartes , Sorbonne Paris Cité , Paris F-75006 , France.,Université Paris Descartes , UMR-S 1140 , Paris F-75006 , France.,Hematology Department , AP-HP, Hôpital Européen Georges Pompidou , INSERM UMR-S 1140 , Paris F-75015 , France
| | - Marie-Claude Menet
- Inserm , U1144 , Paris F-75006 , France.,Université Paris Descartes , UMR-S 1144 , Paris F-75006 , France.,Université Paris Descartes , Sorbonne Paris Cité , Paris F-75006 , France
| | - Pierre-Olivier Couraud
- Université Paris Descartes , Sorbonne Paris Cité , Paris F-75006 , France.,Department of Infection, Institut Cochin , Inserm, U1016 , Paris F-75014 , France.,CNRS, UMR 8104 , Paris F-75014 , France
| | - Salvatore Cisternino
- Inserm , U1144 , Paris F-75006 , France.,Université Paris Descartes , UMR-S 1144 , Paris F-75006 , France.,Université Paris Descartes , Sorbonne Paris Cité , Paris F-75006 , France
| | - Xavier Declèves
- Inserm , U1144 , Paris F-75006 , France.,Université Paris Descartes , UMR-S 1144 , Paris F-75006 , France.,Université Paris Descartes , Sorbonne Paris Cité , Paris F-75006 , France
| |
Collapse
|
14
|
Li G, Che H, Wu WY, Jie LJ, Xiao GS, Wang Y, Li GR. Bradykinin-mediated Ca 2+ signalling regulates cell growth and mobility in human cardiac c-Kit + progenitor cells. J Cell Mol Med 2018; 22:4688-4699. [PMID: 30117680 PMCID: PMC6156395 DOI: 10.1111/jcmm.13706] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/27/2018] [Indexed: 01/05/2023] Open
Abstract
Our recent study showed that bradykinin increases cell cycling progression and migration of human cardiac c‐Kit+ progenitor cells by activating pAkt and pERK1/2 signals. This study investigated whether bradykinin‐mediated Ca2+ signalling participates in regulating cellular functions in cultured human cardiac c‐Kit+ progenitor cells using laser scanning confocal microscopy and biochemical approaches. It was found that bradykinin increased cytosolic free Ca2+ (Cai2+) by triggering a transient Ca2+ release from ER IP3Rs followed by sustained Ca2+ influx through store‐operated Ca2+ entry (SOCE) channel. Blockade of B2 receptor with HOE140 or IP3Rs with araguspongin B or silencing IP3R3 with siRNA abolished both Ca2+ release and Ca2+ influx. It is interesting to note that the bradykinin‐induced cell cycle progression and migration were not observed in cells with siRNA‐silenced IP3R3 or the SOCE component TRPC1, Orai1 or STIM1. Also the bradykinin‐induced increase in pAkt and pERK1/2 as well as cyclin D1 was reduced in these cells. These results demonstrate for the first time that bradykinin‐mediated increase in free Cai2+ via ER‐IP3R3 Ca2+ release followed by Ca2+ influx through SOCE channel plays a crucial role in regulating cell growth and migration via activating pAkt, pERK1/2 and cyclin D1 in human cardiac c‐Kit+ progenitor cells.
Collapse
Affiliation(s)
- Gang Li
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong Pokfulam, Hong Kong, China
| | - Hui Che
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong Pokfulam, Hong Kong, China
| | - Wei-Yin Wu
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Ling-Jun Jie
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Guo-Sheng Xiao
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Gui-Rong Li
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong Pokfulam, Hong Kong, China
| |
Collapse
|
15
|
Vigneault P, Naud P, Qi X, Xiao J, Villeneuve L, Davis DR, Nattel S. Calcium-dependent potassium channels control proliferation of cardiac progenitor cells and bone marrow-derived mesenchymal stem cells. J Physiol 2018; 596:2359-2379. [PMID: 29574723 DOI: 10.1113/jp275388] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 02/26/2018] [Indexed: 12/26/2022] Open
Abstract
KEY POINTS Ex vivo proliferated c-Kit+ endogenous cardiac progenitor cells (eCPCs) obtained from mouse and human cardiac tissues have been reported to express a wide range of functional ion channels. In contrast to previous reports in cultured c-Kit+ eCPCs, we found that ion currents were minimal in freshly isolated cells. However, inclusion of free Ca2+ intracellularly revealed a prominent inwardly rectifying current identified as the intermediate conductance Ca2+ -activated K+ current (KCa3.1) Electrical function of both c-Kit+ eCPCs and bone marrow-derived mesenchymal stem cells is critically governed by KCa3.1 calcium-dependent potassium channels. Ca2+ -induced increases in KCa3.1 conductance are necessary to optimize membrane potential during Ca2+ entry. Membrane hyperpolarization due to KCa3.1 activation maintains the driving force for Ca2+ entry that activates stem cell proliferation. Cardiac disease downregulates KCa3.1 channels in resident cardiac progenitor cells. Alterations in KCa3.1 may have pathophysiological and therapeutic significance in regenerative medicine. ABSTRACT Endogenous c-Kit+ cardiac progenitor cells (eCPCs) and bone marrow (BM)-derived mesenchymal stem cells (MSCs) are being developed for cardiac regenerative therapy, but a better understanding of their physiology is needed. Here, we addressed the unknown functional role of ion channels in freshly isolated eCPCs and expanded BM-MSCs using patch-clamp, microfluorometry and confocal microscopy. Isolated c-Kit+ eCPCs were purified from dog hearts by immunomagnetic selection. Ion currents were barely detectable in freshly isolated c-Kit+ eCPCs with buffering of intracellular calcium (Ca2+i ). Under conditions allowing free intracellular Ca2+ , freshly isolated c-Kit+ eCPCs and ex vivo proliferated BM-MSCs showed prominent voltage-independent conductances that were sensitive to intermediate-conductance K+ -channel (KCa3.1 current, IKCa3.1 ) blockers and corresponding gene (KCNN4)-expression knockdown. Depletion of Ca2+i induced membrane-potential (Vmem ) depolarization, while store-operated Ca2+ entry (SOCE) hyperpolarized Vmem in both cell types. The hyperpolarizing SOCE effect was substantially reduced by IKCa3.1 or SOCE blockade (TRAM-34, 2-APB), and IKCa3.1 blockade (TRAM-34) or KCNN4-knockdown decreased the Ca2+ entry resulting from SOCE. IKCa3.1 suppression reduced c-Kit+ eCPC and BM-MSC proliferation, while significantly altering the profile of cyclin expression. IKCa3.1 was reduced in c-Kit+ eCPCs isolated from dogs with congestive heart failure (CHF), along with corresponding KCNN4 mRNA. Under perforated-patch conditions to maintain physiological [Ca2+ ]i , c-Kit+ eCPCs from CHF dogs had less negative resting membrane potentials (-58 ± 7 mV) versus c-Kit+ eCPCs from control dogs (-73 ± 3 mV, P < 0.05), along with slower proliferation. Our study suggests that Ca2+ -induced increases in IKCa3.1 are necessary to optimize membrane potential during the Ca2+ entry that activates progenitor cell proliferation, and that alterations in KCa3.1 may have pathophysiological and therapeutic significance in regenerative medicine.
Collapse
Affiliation(s)
- Patrick Vigneault
- Research Center and Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Patrice Naud
- Research Center and Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Xiaoyan Qi
- Research Center and Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Jiening Xiao
- Research Center and Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Louis Villeneuve
- Research Center and Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Darryl R Davis
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Stanley Nattel
- Research Center and Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada.,Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
16
|
Dong X, Nakagomi H, Miyamoto T, Ihara T, Kira S, Sawada N, Mitsui T, Takeda M. Tadalafil attenuates hypotonicity-induced Ca 2+ influx via TRPV2 and TRPV4 in primary rat bladder urothelial cell cultures. Neurourol Urodyn 2018; 37:1541-1548. [PMID: 29566267 DOI: 10.1002/nau.23423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 09/06/2017] [Indexed: 12/24/2022]
Abstract
AIMS To investigate the localization of phosphodiesterase 5 (PDE5) and the molecular mechanism underlying the effect of the PDE5 inhibitor tadalafil in signal transduction in the bladder urothelium. METHODS PDE5 expression in rat bladder tissues and cultured primary rat bladder urothelial cells was evaluated using immunochemistry and western blot assays. Ca2+ influx in cells exposed to isotonic solution, hypotonic solution, a selective transient receptor potential vanilloid 2 (TRPV2) channel agonist (cannabidiol), a selective TRPV4 channel agonist (GSK1016790A), a TRP cation channel melastatin 7 (TRPM7) channel agonist (PIP2), or a purinergic receptor agonist (ATP) in the presence or absence of 10 µM tadalafil was evaluated using calcium imaging techniques. We also evaluated stretch-induced changes in ATP concentration in the mouse bladder in the presence or absence of 100 µM tadalafil. RESULTS Immunochemistry and western blot analyses demonstrated that PDE5 is abundantly expressed in the bladder urothelium and in primary rat urothelial cells. Ca2+ influx induced by hypotonic stimulation, GSK1016790A, or cannabidiol was significantly inhibited by tadalafil, whereas ATP-induced Ca2+ influx was unaffected by tadalafil. PIP2 did not induce Ca2+ influx. ATP release in tadalafil-pretreated bladders significantly decreased compared to control bladders. CONCLUSIONS Tadalafil attenuates Ca2+ influx via TRPV4 and TRPV2, and inhibits ATP release in the bladder urothelium. These findings indicate that tadalafil functions as an inhibitor of urothelial signal transduction.
Collapse
Affiliation(s)
- Xiao Dong
- Department of Urology, the First Hospital of China Medical University, Shenyang, People's Republic of China.,Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi 1110 Shimokato, Chuo, Yamanashi, Japan
| | - Hiroshi Nakagomi
- Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi 1110 Shimokato, Chuo, Yamanashi, Japan
| | - Tatsuya Miyamoto
- Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi 1110 Shimokato, Chuo, Yamanashi, Japan
| | - Tatsuya Ihara
- Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi 1110 Shimokato, Chuo, Yamanashi, Japan
| | - Satoru Kira
- Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi 1110 Shimokato, Chuo, Yamanashi, Japan
| | - Norifumi Sawada
- Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi 1110 Shimokato, Chuo, Yamanashi, Japan
| | - Takahiko Mitsui
- Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi 1110 Shimokato, Chuo, Yamanashi, Japan
| | - Masayuki Takeda
- Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi 1110 Shimokato, Chuo, Yamanashi, Japan
| |
Collapse
|
17
|
Ishii T, Uchida K, Hata S, Hatta M, Kita T, Miyake Y, Okamura K, Tamaoki S, Ishikawa H, Yamazaki J. TRPV2 channel inhibitors attenuate fibroblast differentiation and contraction mediated by keratinocyte-derived TGF-β1 in an in vitro wound healing model of rats. J Dermatol Sci 2018; 90:332-342. [PMID: 29610016 DOI: 10.1016/j.jdermsci.2018.03.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/27/2018] [Accepted: 03/01/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Keratinocytes release several factors that are involved in wound contracture and scar formation. We previously reported that a three-dimensional reconstruction model derived from rat skin represents a good wound healing model. OBJECTIVE We characterized the role of transient receptor potential (TRP) channels in the release of transforming growth factor (TGF)-β1 from keratinocytes and the differentiation of fibroblasts to identify possible promising pharmacological approaches to prevent scar formation and contractures. METHODS The three-dimensional culture model was made from rat keratinocytes seeded on a collagen gel in which dermal fibroblasts had been embedded. RESULTS Among the TRP channel inhibitors tested, the TRPV2 inhibitors SKF96365 and tranilast attenuated most potently keratinocyte-dependent and - independent collagen gel contraction due to TGF-β signaling as well as TGF-β1 release from keratinocytes and α-smooth muscle actin production in myofibroblasts. Besides the low amounts detected in normal dermis, TRPV2 mRNA and protein levels were increased after fibroblasts were embedded in the gel. TRPV2 was also expressed in the epidermis and keratinocyte layers of the model. Both inhibitors and TRPV2 siRNA attenuated the intracellular increase of Ca2+ induced by the TRPV agonist 2-aminoethoxydiphenyl borate in TGF-β1-pretreated fibroblasts. CONCLUSION This is the first study to show that compounds targeting TRPV2 channels ameliorate wound contraction through the inhibition of TGF-β1 release and the differentiation of dermal fibroblasts in a culture model.
Collapse
Affiliation(s)
- Taro Ishii
- Department of Oral Growth & Development, Fukuoka 814-0193, Japan
| | - Kunitoshi Uchida
- Department of Physiological Science & Molecular Biology, Fukuoka 814-0193, Japan
| | - Shozaburo Hata
- Department of Oral Growth & Development, Fukuoka 814-0193, Japan
| | - Mitsutoki Hatta
- Department of Physiological Science & Molecular Biology, Fukuoka 814-0193, Japan
| | - Tomo Kita
- Department of Physiological Science & Molecular Biology, Fukuoka 814-0193, Japan
| | - Yuki Miyake
- Department of Oral Growth & Development, Fukuoka 814-0193, Japan
| | - Kazuhiko Okamura
- Department of Morphological Biology, Fukuoka Dental College, Fukuoka 814-0193, Japan
| | - Sachio Tamaoki
- Department of Oral Growth & Development, Fukuoka 814-0193, Japan
| | | | - Jun Yamazaki
- Department of Physiological Science & Molecular Biology, Fukuoka 814-0193, Japan.
| |
Collapse
|
18
|
|
19
|
Li G, Wang Y, Li GR. Bradykinin regulates cell growth and migration in cultured human cardiac c-Kit+ progenitor cells. Oncotarget 2017; 8:10822-10835. [PMID: 28099911 PMCID: PMC5355226 DOI: 10.18632/oncotarget.14609] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 01/06/2017] [Indexed: 01/04/2023] Open
Abstract
Bradykinin is a well-known endogenous vasoactive peptide. The present study investigated the bradykinin receptor expression in human cardiac c-Kit+ progenitor cells and the potential role of bradykinin in regulating cell cycling progression and mobility. It was found that mRNA and protein of bradykinin type 2 receptors, but not bradykinin type 1 receptors, were abundant in cultured human cardiac c-Kit+ progenitor cells. Bradykinin (1-10 nM) stimulated cell growth and migration in a concentration-dependent manner. The increase of cell proliferation was related to promoting G0/G1 transition into G2/M and S phase. Western blots revealed that bradykinin significantly increased pAkt and pERK1/2 as well as cyclin D1, which were countered by HOE140 (an antagonist of bradykinin type 2 receptors) or by silencing bradykinin type 2 receptors. The increase of pAkt, pERK1/2 and cyclin D1 by bradykinin was prevented by the PI3K inhibitor Ly294002, the PLC inhibitors U73122 and neomycin, and/or the PKC inhibitor chelerythrine and the MAPK inhibitor PD98059. Our results demonstrate the novel information that bradykinin promotes cell cycling progression and migration in human cardiac c-Kit+ progenitor cells via activating PI3K, PLC, PKC, cyclin D1, pERK1/2, and pAkt.
Collapse
Affiliation(s)
- Gang Li
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Gui-Rong Li
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| |
Collapse
|
20
|
Blockage of transient receptor potential vanilloid 4 alleviates myocardial ischemia/reperfusion injury in mice. Sci Rep 2017; 7:42678. [PMID: 28205608 PMCID: PMC5311718 DOI: 10.1038/srep42678] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 01/13/2017] [Indexed: 12/15/2022] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a Ca2+-permeable nonselective cation channel and can be activated during ischemia/reperfusion (I/R). This study tested whether blockade of TRPV4 can alleviate myocardial I/R injury in mice. TRPV4 expression began to increase at 1 h, reached statistically at 4 h, and peaked at 24–72 h. Treatment with the selective TRPV4 antagonist HC-067047 or TRPV4 knockout markedly ameliorated myocardial I/R injury as demonstrated by reduced infarct size, decreased troponin T levels and improved cardiac function at 24 h after reperfusion. Importantly, the therapeutic window for HC-067047 lasts for at least 12 h following reperfusion. Furthermore, treatment with HC-067047 reduced apoptosis, as evidenced by the decrease in TUNEL-positive myocytes, Bax/Bcl-2 ratio, and caspase-3 activation. Meanwhile, treatment with HC-067047 attenuated the decrease in the activation of reperfusion injury salvage kinase (RISK) pathway (phosphorylation of Akt, ERK1/2, and GSK-3β), while the activation of survival activating factor enhancement (SAFE) pathway (phosphorylation of STAT3) remained unchanged. In addition, the anti-apoptotic effects of HC-067047 were abolished by the RISK pathway inhibitors. We conclude that blockade of TRPV4 reduces apoptosis via the activation of RISK pathway, and therefore might be a promising strategy to prevent myocardial I/R injury.
Collapse
|