1
|
Yang Y, Xu Z, He S, Wang C, Li R, Zhang R, Li J, Yang Z, Li H, Liu S, Guo Q. Developmental dynamics mimicking inversely engineered pericellular matrix for articular cartilage regeneration. Biomaterials 2024; 317:123066. [PMID: 39742841 DOI: 10.1016/j.biomaterials.2024.123066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
The mechanical mismatch of scaffold matrix-mesenchymal stem cells (MSCs) has been a longstanding issue in the clinical application of MSC-based therapy for articular cartilage (AC) regeneration. Existing tissue-engineered scaffolds underestimate the importance of the natural chondrocyte pericellular matrix (PCM). Here, we reveal the temporal and spatial characteristics of collagen distribution around the chondrocytes. Next, we demonstrate a rationally designed layer-by-layer single-cell encapsulation system which can mimic PCM mechanical responses and enhance MSC chondrogenesis via reestablished the mechanical coupling of PCM-like primitive matrix and chondrocytes. This successfully simulates the temporal and spatial characteristics of collagen secretion. Through investigation of the micromechanical environment of the cells and full-atom simulation analysis of TRPV4, we determine the specific mechanisms by which cellular mechanical forces near the cell are converted into biological signals. The TRPV4-YAP/TAZ-PI3K-Akt signaling pathway is involved in MSC cartilage formation through a joint analysis of the mRNA sequencing and spatial transcriptome results. In a rat model of articular cartilage defects, our inversely engineered pericellular matrix-encapsulated MSC-loaded scaffolds show regenerative performance that are superior to those of scaffolds loaded with only MSCs. These results demonstrate the feasibility of using a PCM-mimicking system to improve MSC chondrogenesis and the efficacy of AC repair.
Collapse
Affiliation(s)
- Yongkang Yang
- School of Medicine, Nankai University, Tianjin, 300071, PR China; Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Ziheng Xu
- School of Medicine, Nankai University, Tianjin, 300071, PR China; Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Songlin He
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China; Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, 999077, PR China
| | - Chao Wang
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Runmeng Li
- School of Medicine, Nankai University, Tianjin, 300071, PR China; Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Ruiyang Zhang
- School of Medicine, Nankai University, Tianjin, 300071, PR China; Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Jianwei Li
- School of Medicine, Nankai University, Tianjin, 300071, PR China; Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Zhen Yang
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, PR China; Arthritis Institute, Peking University, Beijing, 100044, PR China
| | - Hao Li
- School of Medicine, Nankai University, Tianjin, 300071, PR China; Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China; National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, 100853, PR China.
| | - Shuyun Liu
- School of Medicine, Nankai University, Tianjin, 300071, PR China; Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China.
| | - Quanyi Guo
- School of Medicine, Nankai University, Tianjin, 300071, PR China; Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China; National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, 100853, PR China.
| |
Collapse
|
2
|
Binkle-Ladisch L, Pironet A, Zaliani A, Alcouffe C, Mensching D, Haferkamp U, Willing A, Woo MS, Erdmann A, Jessen T, Hess SD, Gribbon P, Pless O, Vennekens R, Friese MA. Identification and development of TRPM4 antagonists to counteract neuronal excitotoxicity. iScience 2024; 27:111425. [PMID: 39687019 PMCID: PMC11648915 DOI: 10.1016/j.isci.2024.111425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/21/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegeneration in central nervous system disorders is linked to dysregulated neuronal calcium. Direct inhibition of glutamate-induced neuronal calcium influx, particularly via N-methyl-D-aspartate receptors (NMDAR), has led to adverse effects and clinical trial failures. A more feasible approach is to modulate NMDAR activity or calcium signaling indirectly. In this respect, the calcium-activated non-selective cation channel transient receptor potential melastatin 4 (TRPM4) has been identified as a promising target. However, high affinity and specific antagonists are lacking. Here, we conducted high-throughput screening of a compound library to identify high affinity TRPM4 antagonists. This yielded five lead compound series with nanomolar half-maximal inhibitory concentration values. Through medicinal chemistry optimization of two series, we established detailed structure-activity relationships and inhibition of excitotoxicity in neurons. Moreover, we identified their potential binding site supported by electrophysiological measurements. These potent TRPM4 antagonists are promising drugs for treating neurodegenerative disorders and TRPM4-related pathologies, potentially overcoming previous therapeutic challenges.
Collapse
Affiliation(s)
- Lars Binkle-Ladisch
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Andy Pironet
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Campus Gasthuisberg O/N1, Herestraat 49-Bus 802, 3000 Leuven, Belgium
| | - Andrea Zaliani
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Chantal Alcouffe
- Department of Chemistry, Evotec SE, 195 Route D'Espagne, 31036 Toulouse, France
| | - Daniel Mensching
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Undine Haferkamp
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Anne Willing
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Marcel S. Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Alexandre Erdmann
- Department of Chemistry, Evotec SE, 195 Route D'Espagne, 31036 Toulouse, France
| | | | - Stephen D. Hess
- Evotec Asia Pte Ltd, 79 Science Park Drive, #04-05 Cintech IV, Singapore 118264, Singapore
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Ole Pless
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Rudi Vennekens
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Campus Gasthuisberg O/N1, Herestraat 49-Bus 802, 3000 Leuven, Belgium
| | - Manuel A. Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
3
|
Hernández-Vega AM, García-Villegas R, Rosenbaum T. Roles for TRPV4 in disease: A discussion of possible mechanisms. Cell Calcium 2024; 124:102972. [PMID: 39609180 DOI: 10.1016/j.ceca.2024.102972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024]
Abstract
The transient receptor potential vanilloid 4 (TRPV4) ion channel is a ubiquitously expressed Ca2+-permeable ion channel that controls intracellular calcium ([Ca2+]i) homeostasis in various types of cells. The physiological roles for TRPV4 are tissue specific and the mechanisms behind this specificity remain mostly unclarified. It is noteworthy that mutations in the TRPV4 channel have been associated to a broad spectrum of congenital diseases, with most of these mutations mainly resulting in gain-of-function. Mutations have been identified in human patients showing a variety of phenotypes and symptoms, mostly related to skeletal and neuromuscular disorders. Since TRPV4 is so widely expressed throughout the body, it comes as no surprise that the literature is growing in evidence linking this protein to malfunction in systems other than the skeletal and neuromuscular. In this review, we summarize the expression patterns of TRPV4 in several tissues and highlight findings of recent studies that address critical structural and functional features of this channel, particularly focusing on its interactions and signaling pathways related to Ca2+ entry. Moreover, we discuss the roles of TRPV4 mutations in some diseases and pinpoint some of the mechanisms underlying pathological states where TRPV4's malfunction is prominent.
Collapse
Affiliation(s)
- Ana M Hernández-Vega
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - Refugio García-Villegas
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Ciudad de México, 07360, México
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico.
| |
Collapse
|
4
|
Sánchez-Hernández R, Benítez-Angeles M, Hernández-Vega AM, Rosenbaum T. Recent advances on the structure and the function relationships of the TRPV4 ion channel. Channels (Austin) 2024; 18:2313323. [PMID: 38354101 PMCID: PMC10868539 DOI: 10.1080/19336950.2024.2313323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024] Open
Abstract
The members of the superfamily of Transient Receptor Potential (TRP) ion channels are physiologically important molecules that have been studied for many years and are still being intensively researched. Among the vanilloid TRP subfamily, the TRPV4 ion channel is an interesting protein due to its involvement in several essential physiological processes and in the development of various diseases. As in other proteins, changes in its function that lead to the development of pathological states, have been closely associated with modification of its regulation by different molecules, but also by the appearance of mutations which affect the structure and gating of the channel. In the last few years, some structures for the TRPV4 channel have been solved. Due to the importance of this protein in physiology, here we discuss the recent progress in determining the structure of the TRPV4 channel, which has been achieved in three species of animals (Xenopus tropicalis, Mus musculus, and Homo sapiens), highlighting conserved features as well as key differences among them and emphasizing the binding sites for some ligands that play crucial roles in its regulation.
Collapse
Affiliation(s)
- Raúl Sánchez-Hernández
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Miguel Benítez-Angeles
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Ana M. Hernández-Vega
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| |
Collapse
|
5
|
Talyzina IA, Nadezhdin KD, Sobolevsky AI. Forty sites of TRP channel regulation. Curr Opin Chem Biol 2024; 84:102550. [PMID: 39615427 DOI: 10.1016/j.cbpa.2024.102550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 12/14/2024]
Abstract
Transient receptor potential (TRP) channels are polymodal molecular sensors that integrate chemical, thermal, mechanical and electrical stimuli and convert them into ionic currents that regulate senses of taste, smell, vision, hearing, touch and contribute to perception of temperature and pain. TRP channels are implicated in the pathogenesis of numerous human diseases, including cancers, and represent one of the most ardently pursued drug targets. Recent advances in structural biology, particularly associated with the cryo-EM "resolution revolution", yielded numerous TRP channel structures in complex with ligands that might have therapeutic potential. In this review, we describe the recent progress in TRP channel structural biology, focusing on the description of identified binding sites for small molecules, their relationship to membrane lipids, and interaction of TRP channels with other proteins. The characterized binding sites and interfaces create a diversity of druggable targets and provide a roadmap to aid in the design of new molecules for tuning TRP channel function in disease conditions.
Collapse
Affiliation(s)
- Irina A Talyzina
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Kirill D Nadezhdin
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Alexander I Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA.
| |
Collapse
|
6
|
Zhao L, Xu K, Talyzina I, Shi J, Li S, Yang Y, Zhang S, Zheng J, Sobolevsky AI, Chen H, Cui J. Human TRPV4 engineering yields an ultrasound-sensitive actuator for sonogenetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.16.618766. [PMID: 39464052 PMCID: PMC11507911 DOI: 10.1101/2024.10.16.618766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Sonogenetics offers non-invasive and cell-type specific modulation of cells genetically engineered to express ultrasound-sensitive actuators. Finding an ion channel to serve as sonogenetic actuator it critical for advancing this promising technique. Here, we show that ultrasound can activate human TRP channel hTRPV4. By screening different hTRPV4 variants, we identify a mutation F617L that increases mechano-sensitivity of this channel to ultrasound, while reduces its sensitivity to hypo-osmolarity, elevated temperature, and agonist. This altered sensitivity profile correlates with structural differences in hTRPV4-F617L compared to wild-type channels revealed by our cryo-electron microscopy analysis. We also show that hTRPV4-F617L can serve as a sonogenetic actuator for neuromodulation in freely moving mice. Our findings demonstrate the use of structure-guided mutagenesis to engineer ion channels with tailored properties of ideal sonogenetic actuators.
Collapse
|
7
|
Orfali R, AlFaiz A, Alanazi M, Alabdulsalam R, Alharbi M, Alromaih Y, Dallak I, Alrahal M, Alwatban A, Saud R. TRPV4 Channel Modulators as Potential Drug Candidates for Cystic Fibrosis. Int J Mol Sci 2024; 25:10551. [PMID: 39408877 PMCID: PMC11476765 DOI: 10.3390/ijms251910551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Cystic fibrosis (CF) is a genetic disorder caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, resulting in defective chloride ion channels. This leads to thick, dehydrated mucus that severely disrupts mucociliary clearance in the respiratory system and triggers infection that eventually is the cause of death of CF patients. Current therapeutic strategies primarily focus on restoring CFTR function, blocking epithelial sodium channels to prevent mucus dehydration, or directly targeting mucus to reduce its viscosity. Among the ion channels expressed in ciliated bronchial epithelial cells, the transient receptor potential vanilloid 4 (TRPV4) channel emerges as a significant channel in CF pathogenesis. Activation of TRPV4 channels affects the regulation of airway surface liquid by modulating sodium absorption and intracellular calcium levels, which indirectly influences CFTR activity. TRPV4 is also involved in the regulatory volume decrease (RVD) process and enhances inflammatory responses in CF patients. Here, we combine current findings on TRPV4 channel modulation as a promising therapeutic approach for CF. Although limited studies have directly explored TRPV4 in CF, emerging evidence indicates that TRPV4 activation can significantly impact key pathological processes in the disease. Further investigation into TRPV4 modulators could lead to innovative treatments that alleviate severe respiratory complications and improve outcomes for CF patients.
Collapse
Affiliation(s)
- Razan Orfali
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia (M.A.)
| | - Ali AlFaiz
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia (M.A.)
| | - Madhawi Alanazi
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia (M.A.)
| | - Rahaf Alabdulsalam
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia (M.A.)
| | - Meaad Alharbi
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia (M.A.)
| | - Yara Alromaih
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia (M.A.)
| | - Ismail Dallak
- King Abdulaziz Medical City, Jeddah 9515, Saudi Arabia
| | - Marah Alrahal
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia (M.A.)
| | - Abdulaziz Alwatban
- College of Medicine, Imam Mohammad Ibn Saud Islamic University, Riyadh 13317, Saudi Arabia
| | - Reem Saud
- General Education Department, Dar Al-Hikmah University, Jeddah 22246, Saudi Arabia
| |
Collapse
|
8
|
Zhang F, Mehta H, Choudhary HH, Islam R, Hanafy KA. TRPV4 Channel in Neurological Disease: from Molecular Mechanisms to Therapeutic Potential. Mol Neurobiol 2024:10.1007/s12035-024-04518-5. [PMID: 39333347 DOI: 10.1007/s12035-024-04518-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/20/2024] [Indexed: 09/29/2024]
Abstract
Transient Receptor Potential Vanilloid 4 (TRPV4) is a non-selective cation channel with pivotal roles in various physiological processes, including osmosensitivity, mechanosensation, neuronal development, vascular tone regulation, and bone homeostasis in human bodies. Recent studies have made significant progress in understanding the structure and functional role of TRPV4, shedding light on its involvement in pathological processes, particularly in the realm of neurological diseases. Here, we aim to provide a comprehensive exploration of the multifaceted contributions of TRPV4 to neurological diseases, spanning its intricate molecular mechanisms to its potential as a target for therapeutic interventions. We delve into the structural and functional attributes of TRPV4, scrutinize its expression profile, and elucidate the possible mechanisms through which it participates in the pathogenesis of neurological disorders. Furthermore, we discussed recent years' progress in therapeutic strategies aimed at harnessing TRPV4 for the treatment of these diseases. These insights will provide a basis for understanding and designing modality-specific pharmacological agents to treat TRPV4-associated disorders.
Collapse
Affiliation(s)
- Feng Zhang
- Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper University Health Care, Camden, NJ, USA
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Hritik Mehta
- Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper University Health Care, Camden, NJ, USA
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Hadi Hasan Choudhary
- Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper University Health Care, Camden, NJ, USA
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Rezwanul Islam
- Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper University Health Care, Camden, NJ, USA
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Khalid A Hanafy
- Cooper Medical School at Rowan University, Camden, NJ, USA.
- Cooper University Health Care, Camden, NJ, USA.
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA.
- Cooper Neurological Institute Center for Neuroinflammation, Cooper Medical School at Rowan University, Camden, NJ, USA.
| |
Collapse
|
9
|
Hernández-Vega AM, Llorente I, Sánchez-Hernández R, Segura Y, Tusié-Luna T, Morales-Buenrostro LE, García-Villegas R, Islas LD, Rosenbaum T. Identification and Properties of TRPV4 Mutant Channels Present in Polycystic Kidney Disease Patients. FUNCTION 2024; 5:zqae031. [PMID: 38984987 PMCID: PMC11384909 DOI: 10.1093/function/zqae031] [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: 04/21/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 07/11/2024] Open
Abstract
Polycystic kidney disease (PKD), a disease characterized by the enlargement of the kidney through cystic growth is the fourth leading cause of end-stage kidney disease world-wide. Transient receptor potential Vanilloid 4 (TRPV4), a calcium-permeable TRP, channel participates in kidney cell physiology and since TRPV4 forms complexes with another channel whose malfunction is associated to PKD, TRPP2 (or PKD2), we sought to determine whether patients with PKD, exhibit previously unknown mutations in TRPV4. Here, we report the presence of mutations in the TRPV4 gene in patients diagnosed with PKD and determine that they produce gain-of-function (GOF). Mutations in the sequence of the TRPV4 gene have been associated to a broad spectrum of neuropathies and skeletal dysplasias but not PKD, and their biophysical effects on channel function have not been elucidated. We identified and examined the functional behavior of a novel E6K mutant and of the previously known S94L and A217S mutant TRVP4 channels. The A217S mutation has been associated to mixed neuropathy and/or skeletal dysplasia phenotypes, however, the PKD carriers of these variants had not been diagnosed with these reported clinical manifestations. The presence of certain mutations in TRPV4 may influence the progression and severity of PKD through GOF mechanisms. PKD patients carrying TRVP4 mutations are putatively more likely to require dialysis or renal transplant as compared to those without these mutations.
Collapse
Affiliation(s)
- Ana M Hernández-Vega
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Itzel Llorente
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Raúl Sánchez-Hernández
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Yayoi Segura
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México14080, Mexico
| | - Teresa Tusié-Luna
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México14080, Mexico
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Luis E Morales-Buenrostro
- Departmento de Nefrología y Metabolismo Mineral, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, México
| | - Refugio García-Villegas
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Ciudad de México 07360, Mexico
| | - León D Islas
- Departamento de Fisiología, Facultad de Medicina. Universidad Nacional Autónoma de México,Ciudad de México 04510, Mexico
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| |
Collapse
|
10
|
Abdul Halim MS, Dyson JM, Gong MM, O'Bryan MK, Nosrati R. Fallopian tube rheology regulates epithelial cell differentiation and function to enhance cilia formation and coordination. Nat Commun 2024; 15:7411. [PMID: 39198453 PMCID: PMC11358425 DOI: 10.1038/s41467-024-51481-9] [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: 06/14/2023] [Accepted: 08/08/2024] [Indexed: 09/01/2024] Open
Abstract
The rheological properties of the extracellular fluid in the female reproductive tract vary spatiotemporally, however, the effect on the behaviour of epithelial cells that line the tract is unexplored. Here, we reveal that epithelial cells respond to the elevated viscosity of culture media by modulating their development and functionality to enhance cilia formation and coordination. Specifically, ciliation increases by 4-fold and cilia beating frequency decreases by 30% when cells are cultured at 100 mPa·s. Further, cilia manifest a coordinated beating pattern that can facilitate the formation of metachronal waves. At the cellular level, viscous loading activates the TRPV4 channel in the epithelial cells to increase intracellular Ca2+, subsequently decreasing the mitochondrial membrane potential level for ATP production to maintain cell viability and function. Our findings provide additional insights into the role of elevated tubal fluid viscosity in promoting ciliation and coordinating their beating-a potential mechanism to facilitate the transport of egg and embryo, suggesting possible therapeutic opportunities for infertility treatment.
Collapse
Affiliation(s)
- Melati S Abdul Halim
- Department of Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria, Australia
| | - Jennifer M Dyson
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Max M Gong
- Department of Biomedical Engineering, Trine University, Angola, IN, USA
| | - Moira K O'Bryan
- School of BioSciences and Bio21 Molecular Science and Biotechnology Institute, Faculty of Science, University of Melbourne, Parkville, Victoria, Australia
| | - Reza Nosrati
- Department of Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
11
|
Cox CD, Poole K, Martinac B. Re-evaluating TRP channel mechanosensitivity. Trends Biochem Sci 2024; 49:693-702. [PMID: 38851904 DOI: 10.1016/j.tibs.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 06/10/2024]
Abstract
Transient receptor potential (TRP) channels are implicated in a wide array of mechanotransduction processes. However, a question remains whether TRP channels directly sense mechanical force, thus acting as primary mechanotransducers. We use several recent examples to demonstrate the difficulty in definitively ascribing mechanosensitivity to TRP channel subfamilies. Ultimately, despite being implicated in an ever-growing list of mechanosignalling events in most cases limited robust or reproducible evidence supports the contention that TRP channels act as primary transducers of mechanical forces. They either (i) possess unique and as yet unspecified structural or local requirements for mechanosensitivity; or (ii) act as mechanoamplifiers responding downstream of the activation of a primary mechanotransducer that could include Ca2+-permeable mechanosensitive (MS) channels or other potentially unidentified mechanosensors.
Collapse
Affiliation(s)
- Charles D Cox
- School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW, 2052, Australia; Victor Chang Cardiac Research Institute, Sydney, Darlinghurst, NSW, 2010, Australia
| | - Kate Poole
- School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW, 2052, Australia.
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Sydney, Darlinghurst, NSW, 2010, Australia.
| |
Collapse
|
12
|
Fan J, Guo C, Liao D, Ke H, Lei J, Xie W, Tang Y, Tominaga M, Huang Z, Lei X. Structural Pharmacology of TRPV4 Antagonists. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401583. [PMID: 38659239 PMCID: PMC11220649 DOI: 10.1002/advs.202401583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/21/2024] [Indexed: 04/26/2024]
Abstract
The nonselective calcium-permeable Transient Receptor Potential Cation Channel Subfamily V Member4 (TRPV4) channel regulates various physiological activities. Dysfunction of TRPV4 is linked to many severe diseases, including edema, pain, gastrointestinal disorders, lung diseases, and inherited neurodegeneration. Emerging TRPV4 antagonists show potential clinical benefits. However, the molecular mechanisms of TRPV4 antagonism remain poorly understood. Here, cryo-electron microscopy (cryo-EM) structures of human TRPV4 are presented in-complex with two potent antagonists, revealing the detailed binding pockets and regulatory mechanisms of TRPV4 gating. Both antagonists bind to the voltage-sensing-like domain (VSLD) and stabilize the channel in closed states. These two antagonists induce TRPV4 to undergo an apparent fourfold to twofold symmetry transition. Moreover, it is demonstrated that one of the antagonists binds to the VSLD extended pocket, which differs from the canonical VSLD pocket. Complemented with functional and molecular dynamics simulation results, this study provides crucial mechanistic insights into TRPV4 regulation by small-molecule antagonists, which may facilitate future drug discovery targeting TRPV4.
Collapse
Affiliation(s)
- Junping Fan
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringPeking‐Tsinghua Center for Life SciencesPeking UniversityBeijing100871China
| | - Chang Guo
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking University Health Science CenterBeijing100191China
| | | | - Han Ke
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringPeking‐Tsinghua Center for Life SciencesPeking UniversityBeijing100871China
| | - Jing Lei
- Division of Cell SignalingNational Institute for Physiological SciencesThermal Biology GroupExploratory Research Center on Life and Living SystemsNational Institutes of Natural SciencesOkazaki444‐8787Japan
| | - Wenjun Xie
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringPeking‐Tsinghua Center for Life SciencesPeking UniversityBeijing100871China
| | - Yuliang Tang
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringPeking‐Tsinghua Center for Life SciencesPeking UniversityBeijing100871China
| | - Makoto Tominaga
- Division of Cell SignalingNational Institute for Physiological SciencesThermal Biology GroupExploratory Research Center on Life and Living SystemsNational Institutes of Natural SciencesOkazaki444‐8787Japan
- Nagoya Advanced Research and Developmet CenterNagoya City UniversityNagoya467‐8601Japan
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking University Health Science CenterBeijing100191China
| | - Xiaoguang Lei
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringPeking‐Tsinghua Center for Life SciencesPeking UniversityBeijing100871China
| |
Collapse
|
13
|
Trofimov YA, Krylov NA, Minakov AS, Nadezhdin KD, Neuberger A, Sobolevsky AI, Efremov RG. Dynamic molecular portraits of ion-conducting pores characterize functional states of TRPV channels. Commun Chem 2024; 7:119. [PMID: 38824263 PMCID: PMC11144267 DOI: 10.1038/s42004-024-01198-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 05/06/2024] [Indexed: 06/03/2024] Open
Abstract
Structural biology is solving an ever-increasing number of snapshots of ion channel conformational ensembles. Deciphering ion channel mechanisms, however, requires understanding the ensemble dynamics beyond the static structures. Here, we present a molecular modeling-based approach characterizing the ion channel structural intermediates, or their "dynamic molecular portraits", by assessing water and ion conductivity along with the detailed evaluation of pore hydrophobicity and residue packing. We illustrate the power of this approach by analyzing structures of few vanilloid-subfamily transient receptor potential (TRPV) channels. Based on the pore architecture, there are three major states that are common for TRPVs, which we call α-closed, π-closed, and π-open. We show that the pore hydrophobicity and residue packing for the open state is most favorable for the pore conductance. On the contrary, the α-closed state is the most hydrophobic and always non-conducting. Our approach can also be used for structural and functional classification of ion channels.
Collapse
Affiliation(s)
- Yury A Trofimov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Nikolay A Krylov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | | | - Kirill D Nadezhdin
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Arthur Neuberger
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Alexander I Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Roman G Efremov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
14
|
Sullivan JM, Bagnell AM, Alevy J, Avila EM, Mihaljević L, Saavedra-Rivera PC, Kong L, Huh JS, McCray BA, Aisenberg WH, Zuberi AR, Bogdanik L, Lutz CM, Qiu Z, Quinlan KA, Searson PC, Sumner CJ. Gain-of-function mutations of TRPV4 acting in endothelial cells drive blood-CNS barrier breakdown and motor neuron degeneration in mice. Sci Transl Med 2024; 16:eadk1358. [PMID: 38776392 PMCID: PMC11316273 DOI: 10.1126/scitranslmed.adk1358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 05/01/2024] [Indexed: 05/25/2024]
Abstract
Blood-CNS barrier disruption is a hallmark of numerous neurological disorders, yet whether barrier breakdown is sufficient to trigger neurodegenerative disease remains unresolved. Therapeutic strategies to mitigate barrier hyperpermeability are also limited. Dominant missense mutations of the cation channel transient receptor potential vanilloid 4 (TRPV4) cause forms of hereditary motor neuron disease. To gain insights into the cellular basis of these disorders, we generated knock-in mouse models of TRPV4 channelopathy by introducing two disease-causing mutations (R269C and R232C) into the endogenous mouse Trpv4 gene. TRPV4 mutant mice exhibited weakness, early lethality, and regional motor neuron loss. Genetic deletion of the mutant Trpv4 allele from endothelial cells (but not neurons, glia, or muscle) rescued these phenotypes. Symptomatic mutant mice exhibited focal disruptions of blood-spinal cord barrier (BSCB) integrity, associated with a gain of function of mutant TRPV4 channel activity in neural vascular endothelial cells (NVECs) and alterations of NVEC tight junction structure. Systemic administration of a TRPV4-specific antagonist abrogated channel-mediated BSCB impairments and provided a marked phenotypic rescue of symptomatic mutant mice. Together, our findings show that mutant TRPV4 channels can drive motor neuron degeneration in a non-cell autonomous manner by precipitating focal breakdown of the BSCB. Further, these data highlight the reversibility of TRPV4-mediated BSCB impairments and identify a potential therapeutic strategy for patients with TRPV4 mutations.
Collapse
Affiliation(s)
- Jeremy M. Sullivan
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Anna M. Bagnell
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Jonathan Alevy
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Elvia Mena Avila
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island; Kingston, RI 02881, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island; Kingston, RI 02881, USA
| | - Ljubica Mihaljević
- Department of Physiology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | | | - Lingling Kong
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Jennifer S. Huh
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Brett A. McCray
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - William H. Aisenberg
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | | | | | | | - Zhaozhu Qiu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
- Department of Physiology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Katharina A. Quinlan
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island; Kingston, RI 02881, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island; Kingston, RI 02881, USA
| | - Peter C. Searson
- Institute for Nanobiotechnology, Johns Hopkins University; Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University; Baltimore, MD 21218, USA
| | - Charlotte J. Sumner
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| |
Collapse
|
15
|
Babaniamansour P, Jacho D, Niedzielski S, Rabino A, Garcia-Mata R, Yildirim-Ayan E. Modulating TRPV4 Channel Activity in Pro-Inflammatory Macrophages within the 3D Tissue Analog. Biomedicines 2024; 12:230. [PMID: 38275401 PMCID: PMC10813551 DOI: 10.3390/biomedicines12010230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
Investigating macrophage plasticity emerges as a promising strategy for promoting tissue regeneration and can be exploited by regulating the transient receptor potential vanilloid 4 (TRPV4) channel. The TRPV4 channel responds to various stimuli including mechanical, chemical, and selective pharmacological compounds. It is well documented that treating cells such as epithelial cells and fibroblasts with a TRPV4 agonist enhances the Ca2+ influx to the cells, which leads to secretion of pro-inflammatory cytokines, while a TRPV4 antagonist reduces both Ca2+ influx and pro-inflammatory cytokine secretion. In this work, we investigated the effect of selective TRPV4 modulator compounds on U937-differentiated macrophages encapsulated within three-dimensional (3D) matrices. Despite offering a more physiologically relevant model than 2D cultures, pharmacological treatment of macrophages within 3D collagen matrices is largely overlooked in the literature. In this study, pro-inflammatory macrophages were treated with an agonist, 500 nM of GSK1016790A (TRPV4(+)), and an antagonist, 10 mM of RN-1734 (TRPV4(-)), to elucidate the modulation of the TRPV4 channel at both cellular and extracellular levels. To evaluate macrophage phenotypic alterations within 3D collagen matrices following TRPV4 modulator treatment, we employed structural techniques (SEM, Masson's trichrome, and collagen hybridizing peptide (CHP) staining), quantitative morphological measures for phenotypic assessment, and genotypic methods such as quantitative real-time PCR (qRT-PCR) and immunohistochemistry (IHC). Our data reveal that pharmacological modulation of the macrophage TRPV4 channel alters the cytoskeletal structure of macrophages and influences the 3D structure encapsulating them. Moreover, we proved that treating macrophages with a TRPV4 agonist and antagonist enhances the expression of pro- and anti-inflammatory genes, respectively, leading to the upregulation of surface markers CD80 and CD206. In the TRPV4(-) group, the CD206 gene and CD206 surface marker were significantly upregulated by 9- and 2.5-fold, respectively, compared to the control group. These findings demonstrate that TRPV4 modulation can be utilized to shift macrophage phenotype within the 3D matrix toward a desired state. This is an innovative approach to addressing inflammation in musculoskeletal tissues.
Collapse
Affiliation(s)
- Parto Babaniamansour
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43606, USA; (P.B.); (S.N.)
| | - Diego Jacho
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43606, USA; (P.B.); (S.N.)
| | - Skyler Niedzielski
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43606, USA; (P.B.); (S.N.)
| | - Agustin Rabino
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Rafael Garcia-Mata
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Eda Yildirim-Ayan
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43606, USA; (P.B.); (S.N.)
| |
Collapse
|
16
|
Huang J, Korsunsky A, Yazdani M, Chen J. Targeting TRP channels: recent advances in structure, ligand binding, and molecular mechanisms. Front Mol Neurosci 2024; 16:1334370. [PMID: 38273937 PMCID: PMC10808746 DOI: 10.3389/fnmol.2023.1334370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Transient receptor potential (TRP) channels are a large and diverse family of transmembrane ion channels that are widely expressed, have important physiological roles, and are associated with many human diseases. These proteins are actively pursued as promising drug targets, benefitting greatly from advances in structural and mechanistic studies of TRP channels. At the same time, the complex, polymodal activation and regulation of TRP channels have presented formidable challenges. In this short review, we summarize recent progresses toward understanding the structural basis of TRP channel function, as well as potential ligand binding sites that could be targeted for therapeutics. A particular focus is on the current understanding of the molecular mechanisms of TRP channel activation and regulation, where many fundamental questions remain unanswered. We believe that a deeper understanding of the functional mechanisms of TRP channels will be critical and likely transformative toward developing successful therapeutic strategies targeting these exciting proteins. This endeavor will require concerted efforts from computation, structural biology, medicinal chemistry, electrophysiology, pharmacology, drug safety and clinical studies.
Collapse
Affiliation(s)
- Jian Huang
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| | - Aron Korsunsky
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| | - Mahdieh Yazdani
- Modeling and Informatics, Merck & Co., Inc., West Point, PA, United States
| | - Jianhan Chen
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| |
Collapse
|
17
|
Huang Y, Ma D, Yang Z, Zhao Y, Guo J. Voltage-gated potassium channels KCNQs: Structures, mechanisms, and modulations. Biochem Biophys Res Commun 2023; 689:149218. [PMID: 37976835 DOI: 10.1016/j.bbrc.2023.149218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/19/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
KCNQ (Kv7) channels are voltage-gated, phosphatidylinositol 4,5-bisphosphate- (PIP2-) modulated potassium channels that play essential roles in regulating the activity of neurons and cardiac myocytes. Hundreds of mutations in KCNQ channels are closely related to various cardiac and neurological disorders, such as long QT syndrome, epilepsy, and deafness, which makes KCNQ channels important drug targets. During the past several years, the application of single-particle cryo-electron microscopy (cryo-EM) technique in the structure determination of KCNQ channels has greatly advanced our understanding of their molecular mechanisms. In this review, we summarize the currently available structures of KCNQ channels, analyze their special voltage gating mechanism, and discuss their activation mechanisms by both the endogenous membrane lipid and the exogenous synthetic ligands. These structural studies of KCNQ channels will guide the development of drugs targeting KCNQ channels.
Collapse
Affiliation(s)
- Yuan Huang
- Department of Cardiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Demin Ma
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zhenni Yang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yiwen Zhao
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050011, China
| | - Jiangtao Guo
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
18
|
Hu X, Hu H. Structural insights into the TRPV4-RhoA complex offer clues to solve the puzzle of TRPV4 channelopathies. Cell Calcium 2023; 116:102814. [PMID: 37839180 PMCID: PMC11192545 DOI: 10.1016/j.ceca.2023.102814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/17/2023]
Affiliation(s)
- Xueming Hu
- The Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hongzhen Hu
- The Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
19
|
Otero-Sobrino Á, Blanco-Carlón P, Navarro-Aguadero MÁ, Gallardo M, Martínez-López J, Velasco-Estévez M. Mechanosensitive Ion Channels: Their Physiological Importance and Potential Key Role in Cancer. Int J Mol Sci 2023; 24:13710. [PMID: 37762011 PMCID: PMC10530364 DOI: 10.3390/ijms241813710] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Mechanosensitive ion channels comprise a broad group of proteins that sense mechanical extracellular and intracellular changes, translating them into cation influx to adapt and respond to these physical cues. All cells in the organism are mechanosensitive, and these physical cues have proven to have an important role in regulating proliferation, cell fate and differentiation, migration and cellular stress, among other processes. Indeed, the mechanical properties of the extracellular matrix in cancer change drastically due to high cell proliferation and modification of extracellular protein secretion, suggesting an important contribution to tumor cell regulation. In this review, we describe the physiological significance of mechanosensitive ion channels, emphasizing their role in cancer and immunity, and providing compelling proof of the importance of continuing to explore their potential as new therapeutic targets in cancer research.
Collapse
Affiliation(s)
- Álvaro Otero-Sobrino
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Pablo Blanco-Carlón
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Miguel Ángel Navarro-Aguadero
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Miguel Gallardo
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Joaquín Martínez-López
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
| | - María Velasco-Estévez
- H12O-CNIO Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), 28029 Madrid, Spain; (Á.O.-S.); (P.B.-C.); (M.Á.N.-A.); (M.G.); (J.M.-L.)
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| |
Collapse
|
20
|
Wei BY, Hou JN, Yan CP, Wen SY, Shang XS, Guo YC, Feng T, Liu TF, Chen ZY, Chen XP. Shexiang Baoxin Pill treats acute myocardial infarction by promoting angiogenesis via GDF15-TRPV4 signaling. Biomed Pharmacother 2023; 165:115186. [PMID: 37481933 DOI: 10.1016/j.biopha.2023.115186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/12/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023] Open
Abstract
Angiogenesis has been considered a pivotal strategy for treating ischemic heart disease. One possible approach, the Shexiang Baoxin Pill (MUSKARDIA), has been noted to promote angiogenesis, but its underlying mechanism is still largely unknown. We aimed to determine the effects of MUSKARDIA on acute myocardial infarction (AMI), as well as the underlying mechanistic bases. AMI was induced in rats, using left anterior descending coronary arterial occlusion, and either 6 (low) or 12 (high-dose) mg/kg/day of MUSKARDIA was administered for 56 days. We found that MUSKARDIA improved cardiac function and counteracted against adverse remodeling among AMI rats, which most likely is due to it promoting angiogenesis. Transcriptome analysis by RNA-sequencing found that MUSKARDIA up-regulated cardiac pro-angiogenic genes, particularly growth differentiation factor 15 (GDF15), which was confirmed by RT-qPCR. This up-regulation was also correlated with elevated serum GDF15 levels. In vitro analyses with human umbilical vein endothelial cells found that increased GDF15, stimulated by MUSKARDIA, resulted in enhanced cell migration, proliferation, and tubular formation, all of which were reversed after GDF15 knockdown using a lentiviral vector. Gene Ontology, as well as Kyoto Genes and Genomes enrichment analyses identified calcium signaling pathway as a major contributor to these outcomes, which was verified by Western blot and Cal-590 AM loading showing that transient receptor potential cation channel subfamily V member 4 protein (TRPV4) and intracellular Ca2+ levels increased in accordance with MUSKARDIA-induced GDF15 up-regulation, and decreased with GDF15 knock-down. Therefore, MUSKARDIA may exert its cardioprotective effects via stimulating the GDF15/TRPV4/calcium signaling/angiogenesis axis.
Collapse
Affiliation(s)
- Bing-Yan Wei
- Shanxi Key Laboratory of Experimental Animals and Animal Models for Human Diseases, Laboratory Animal Center, Shanxi Medical University, Taiyuan 030001, China
| | - Jia-Nan Hou
- Shanxi Key Laboratory of Experimental Animals and Animal Models for Human Diseases, Laboratory Animal Center, Shanxi Medical University, Taiyuan 030001, China
| | - Chang-Ping Yan
- Department of gynecology of Shanxi Cancer Hospital, Taiyuan 030001, China
| | - Shi-Yuan Wen
- Basic Medical School, Shanxi Medical University, Taiyuan 030001, China
| | - Xiao-Sen Shang
- Department of Cardiology of Taiyuan Central Hospital, Taiyuan 030001, China
| | - Yong-Chang Guo
- Shanxi Key Laboratory of Experimental Animals and Animal Models for Human Diseases, Laboratory Animal Center, Shanxi Medical University, Taiyuan 030001, China
| | - Tao Feng
- Department of Cardiology of Taiyuan Central Hospital, Taiyuan 030001, China
| | - Tian-Fu Liu
- Shanxi Key Laboratory of Experimental Animals and Animal Models for Human Diseases, Laboratory Animal Center, Shanxi Medical University, Taiyuan 030001, China.
| | - Zhao-Yang Chen
- Shanxi Key Laboratory of Experimental Animals and Animal Models for Human Diseases, Laboratory Animal Center, Shanxi Medical University, Taiyuan 030001, China.
| | - Xiao-Ping Chen
- Department of Cardiology of Taiyuan Central Hospital, Taiyuan 030001, China.
| |
Collapse
|
21
|
Goretzki B, Wiedemann C, McCray BA, Schäfer SL, Jansen J, Tebbe F, Mitrovic SA, Nöth J, Cabezudo AC, Donohue JK, Jeffries CM, Steinchen W, Stengel F, Sumner CJ, Hummer G, Hellmich UA. Crosstalk between regulatory elements in disordered TRPV4 N-terminus modulates lipid-dependent channel activity. Nat Commun 2023; 14:4165. [PMID: 37443299 PMCID: PMC10344929 DOI: 10.1038/s41467-023-39808-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Intrinsically disordered regions (IDRs) are essential for membrane receptor regulation but often remain unresolved in structural studies. TRPV4, a member of the TRP vanilloid channel family involved in thermo- and osmosensation, has a large N-terminal IDR of approximately 150 amino acids. With an integrated structural biology approach, we analyze the structural ensemble of the TRPV4 IDR and the network of antagonistic regulatory elements it encodes. These modulate channel activity in a hierarchical lipid-dependent manner through transient long-range interactions. A highly conserved autoinhibitory patch acts as a master regulator by competing with PIP2 binding to attenuate channel activity. Molecular dynamics simulations show that loss of the interaction between the PIP2-binding site and the membrane reduces the force exerted by the IDR on the structured core of TRPV4. This work demonstrates that IDR structural dynamics are coupled to TRPV4 activity and highlights the importance of IDRs for TRP channel function and regulation.
Collapse
Affiliation(s)
- Benedikt Goretzki
- Friedrich Schiller University Jena, Faculty of Chemistry and Earth Sciences, Institute of Organic Chemistry and Macromolecular Chemistry, Jena, Germany
- Centre for Biomolecular Magnetic Resonance (BMRZ), Goethe University, Frankfurt am Main, Germany
| | - Christoph Wiedemann
- Friedrich Schiller University Jena, Faculty of Chemistry and Earth Sciences, Institute of Organic Chemistry and Macromolecular Chemistry, Jena, Germany
| | - Brett A McCray
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stefan L Schäfer
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Jasmin Jansen
- Department of Biology, University of Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Frederike Tebbe
- Friedrich Schiller University Jena, Faculty of Chemistry and Earth Sciences, Institute of Organic Chemistry and Macromolecular Chemistry, Jena, Germany
| | - Sarah-Ana Mitrovic
- Department of Chemistry, Section Biochemistry, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Julia Nöth
- Department of Chemistry, Section Biochemistry, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Ainara Claveras Cabezudo
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
- IMPRS on Cellular Biophysics, Frankfurt am Main, Germany
| | - Jack K Donohue
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cy M Jeffries
- European Molecular Biology Laboratory, EMBL Hamburg Unit, Deutsches Elektronen-Synchrotron, Hamburg, Germany
| | - Wieland Steinchen
- Center for Synthetic Microbiology (SYNMIKRO) & Department of Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Florian Stengel
- Department of Biology, University of Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Charlotte J Sumner
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gerhard Hummer
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
- Institute of Biophysics, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Ute A Hellmich
- Friedrich Schiller University Jena, Faculty of Chemistry and Earth Sciences, Institute of Organic Chemistry and Macromolecular Chemistry, Jena, Germany.
- Centre for Biomolecular Magnetic Resonance (BMRZ), Goethe University, Frankfurt am Main, Germany.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|