1
|
Hansen CE, Hollaus D, Kamermans A, de Vries HE. Tension at the gate: sensing mechanical forces at the blood-brain barrier in health and disease. J Neuroinflammation 2024; 21:325. [PMID: 39696463 DOI: 10.1186/s12974-024-03321-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024] Open
Abstract
Microvascular brain endothelial cells tightly limit the entry of blood components and peripheral cells into the brain by forming the blood-brain barrier (BBB). The BBB is regulated by a cascade of mechanical and chemical signals including shear stress and elasticity of the adjacent endothelial basement membrane (BM). During physiological aging, but especially in neurological diseases including multiple sclerosis (MS), stroke, small vessel disease, and Alzheimer's disease (AD), the BBB is exposed to inflammation, rigidity changes of the BM, and disturbed cerebral blood flow (CBF). These altered forces lead to increased vascular permeability, reduced endothelial reactivity to vasoactive mediators, and promote leukocyte transmigration. Whereas the molecular players involved in leukocyte infiltration have been described in detail, the importance of mechanical signalling throughout this process has only recently been recognized. Here, we review relevant features of mechanical forces acting on the BBB under healthy and pathological conditions, as well as the endothelial mechanosensory elements detecting and responding to altered forces. We demonstrate the underlying complexity by focussing on the family of transient receptor potential (TRP) ion channels. A better understanding of these processes will provide insights into the pathogenesis of several neurological disorders and new potential leads for treatment.
Collapse
Affiliation(s)
- Cathrin E Hansen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - David Hollaus
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
2
|
Sukumar VK, Tai YK, Chan CW, Iversen JN, Wu KY, Fong CHH, Lim JSJ, Franco-Obregón A. Brief Magnetic Field Exposure Stimulates Doxorubicin Uptake into Breast Cancer Cells in Association with TRPC1 Expression: A Precision Oncology Methodology to Enhance Chemotherapeutic Outcome. Cancers (Basel) 2024; 16:3860. [PMID: 39594815 PMCID: PMC11592624 DOI: 10.3390/cancers16223860] [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: 10/21/2024] [Revised: 11/04/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Background/Objectives: Doxorubicin (DOX) is commonly used as a chemotherapeutic agent for the treatment of breast cancer. Nonetheless, its systemic delivery via intravenous injection and toxicity towards healthy tissues commonly result in a broad range of detrimental side effects. Breast cancer severity was previously shown to be correlated with TRPC1 channel expression that conferred upon it enhanced vulnerability to pulsed electromagnetic field (PEMF) therapy. PEMF therapy was also previously shown to enhance breast cancer cell vulnerability to DOX in vitro and in vivo that correlated with TRPC1 expression and mitochondrial respiratory rates. Methods: DOX uptake was assessed by measuring its innate autofluorescence within murine 4T1 or human MCF7 breast cancer cells following magnetic exposure. Cellular vulnerability to doxorubicin uptake was assessed by monitoring mitochondrial activity and cellular DNA content. Results: Here, we demonstrate that 10 min of PEMF exposure could augment DOX uptake into 4T1 and MCF7 breast cancer cells. DOX uptake could be increased by TRPC1 overexpression, whereas inhibiting the activity of TRPC1 channels with SKF-96356 or genetic knockdown, precluded DOX uptake. PEMF exposure enhances DOX-mediated killing of breast cancer cells, reducing the IC50 value of DOX by half, whereas muscle cells, representative of collateral tissues, were less sensitive to PEMF-enhanced DOX-mediated cytotoxicity. Vesicular loading of DOX correlated with TRPC1 expression. Conclusions: This study presents a novel TRPC1-mediated mechanism through which PEMF therapy may enhance DOX cytotoxicity in breast cancer cells, paving the way for the development of localized non-invasive PEMF platforms to improve cancer outcomes with lower systemic levels of DOX.
Collapse
Affiliation(s)
- Viresh Krishnan Sukumar
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; (V.K.S.); (J.S.J.L.)
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Yee Kit Tai
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; (V.K.S.); (J.S.J.L.)
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Ching Wan Chan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Jan Nikolas Iversen
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Kwan Yu Wu
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Charlene Hui Hua Fong
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - Joline Si Jing Lim
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; (V.K.S.); (J.S.J.L.)
- Experimental Therapeutics Programme, Cancer Science Institute, Singapore 117599, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University Singapore, Singapore 119228, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Hospital, Singapore 119074, Singapore
| | - Alfredo Franco-Obregón
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; (V.K.S.); (J.S.J.L.)
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore; (J.N.I.); (K.Y.W.); (C.H.H.F.)
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| |
Collapse
|
3
|
Garcia-Sanchez J, Lin D, Liu WW. Mechanosensitive ion channels in glaucoma pathophysiology. Vision Res 2024; 223:108473. [PMID: 39180975 PMCID: PMC11398070 DOI: 10.1016/j.visres.2024.108473] [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: 06/17/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/27/2024]
Abstract
Force sensing is a fundamental ability that allows cells and organisms to interact with their physical environment. The eye is constantly subjected to mechanical forces such as blinking and eye movements. Furthermore, elevated intraocular pressure (IOP) can cause mechanical strain at the optic nerve head, resulting in retinal ganglion cell death (RGC) in glaucoma. How mechanical stimuli are sensed and affect cellular physiology in the eye is unclear. Recent studies have shown that mechanosensitive ion channels are expressed in many ocular tissues relevant to glaucoma and may influence IOP regulation and RGC survival. Furthermore, variants in mechanosensitive ion channel genes may be associated with risk for primary open angle glaucoma. These findings suggest that mechanosensitive channels may be important mechanosensors mediating cellular responses to pressure signals in the eye. In this review, we focus on mechanosensitive ion channels from three major channel families-PIEZO, two-pore potassium and transient receptor potential channels. We review the key properties of these channels, their effects on cell function and physiology, and discuss their possible roles in glaucoma pathophysiology.
Collapse
Affiliation(s)
- Julian Garcia-Sanchez
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Danting Lin
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Wendy W Liu
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
4
|
Franco-Obregón A, Tai YK. Are Aminoglycoside Antibiotics TRPing Your Metabolic Switches? Cells 2024; 13:1273. [PMID: 39120305 PMCID: PMC11311832 DOI: 10.3390/cells13151273] [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: 07/03/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 08/10/2024] Open
Abstract
Transient receptor potential (TRP) channels are broadly implicated in the developmental programs of most tissues. Amongst these tissues, skeletal muscle and adipose are noteworthy for being essential in establishing systemic metabolic balance. TRP channels respond to environmental stimuli by supplying intracellular calcium that instigates enzymatic cascades of developmental consequence and often impinge on mitochondrial function and biogenesis. Critically, aminoglycoside antibiotics (AGAs) have been shown to block the capacity of TRP channels to conduct calcium entry into the cell in response to a wide range of developmental stimuli of a biophysical nature, including mechanical, electromagnetic, thermal, and chemical. Paradoxically, in vitro paradigms commonly used to understand organismal muscle and adipose development may have been led astray by the conventional use of streptomycin, an AGA, to help prevent bacterial contamination. Accordingly, streptomycin has been shown to disrupt both in vitro and in vivo myogenesis, as well as the phenotypic switch of white adipose into beige thermogenic status. In vivo, streptomycin has been shown to disrupt TRP-mediated calcium-dependent exercise adaptations of importance to systemic metabolism. Alternatively, streptomycin has also been used to curb detrimental levels of calcium leakage into dystrophic skeletal muscle through aberrantly gated TRPC1 channels that have been shown to be involved in the etiology of X-linked muscular dystrophies. TRP channels susceptible to AGA antagonism are critically involved in modulating the development of muscle and adipose tissues that, if administered to behaving animals, may translate to systemwide metabolic disruption. Regenerative medicine and clinical communities need to be made aware of this caveat of AGA usage and seek viable alternatives, to prevent contamination or infection in in vitro and in vivo paradigms, respectively.
Collapse
Affiliation(s)
- Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Competence Center for Applied Biotechnology and Molecular Medicine, University of Zürich, 8057 Zürich, Switzerland
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Yee Kit Tai
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| |
Collapse
|
5
|
Li C, Zhao M, Liu X, Li Y, Xu B, Zhou L, Sun X, Sun W, Kang N, Ji Z, Li T, An H, Wang F, Wu C, Ye JY, Zhang JR, Wang Q, Zhao X, Li Z, Liu W. Ion channel TRPV2 is critical in enhancing B cell activation and function. J Exp Med 2024; 221:e20221042. [PMID: 38353705 PMCID: PMC10866685 DOI: 10.1084/jem.20221042] [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: 06/16/2022] [Revised: 11/28/2023] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Abstract
The function of transient receptor potential vanilloid (TRPV) cation channels governing B cell activation remains to be explored. We present evidence that TRPV2 is highly expressed in B cells and plays a crucial role in the formation of the B cell immunological synapse and B cell activation. Physiologically, TRPV2 expression level is positively correlated to influenza-specific antibody production and is low in newborns and seniors. Pathologically, a positive correlation is established between TRPV2 expression and the clinical manifestations of systemic lupus erythematosus (SLE) in adult and child SLE patients. Correspondingly, mice with deficient TRPV2 in B cells display impaired antibody responses following immunization. Mechanistically, the pore and N-terminal domains of TRPV2 are crucial for gating cation permeation and executing mechanosensation in B cells upon antigen stimulation. These processes synergistically contribute to membrane potential depolarization and cytoskeleton remodeling within the B cell immunological synapse, fostering efficient B cell activation. Thus, TRPV2 is critical in augmenting B cell activation and function.
Collapse
Affiliation(s)
- Cuifeng Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Meng Zhao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Xiaohang Liu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Yuxin Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Bihua Xu
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Lina Zhou
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, PR China
| | - Xiaolin Sun
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Wenbo Sun
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Na Kang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Zhenglin Ji
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Tong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Haoran An
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| | - Fei Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jing-Ying Ye
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Jing-Ren Zhang
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| | - Qingwen Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Xiaodong Zhao
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, PR China
- Department of Rheumatology and Immunology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Zhanguo Li
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Wanli Liu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| |
Collapse
|
6
|
Liu WW, Kinzy TG, Cooke Bailey JN, Xu Z, Hysi P, Wiggs JL. Mechanosensitive ion channel gene survey suggests potential roles in primary open angle glaucoma. Sci Rep 2023; 13:15871. [PMID: 37741866 PMCID: PMC10517927 DOI: 10.1038/s41598-023-43072-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023] Open
Abstract
Although glaucoma is a disease modulated by eye pressure, the mechanisms of pressure sensing in the eye are not well understood. Here, we investigated associations between mechanosensitive ion channel gene variants and primary open-angle glaucoma (POAG). Common (minor allele frequency > 5%) single nucleotide polymorphisms located within the genomic regions of 20 mechanosensitive ion channel genes in the K2P, TMEM63, PIEZO and TRP channel families were assessed using genotype data from the NEIGHBORHOOD consortium of 3853 cases and 33,480 controls. Rare (minor allele frequency < 1%) coding variants were assessed using exome array genotyping data for 2606 cases and 2606 controls. Association with POAG was analyzed using logistic regression adjusting for age and sex. Two rare PIEZO1 coding variants with protective effects were identified in the NEIGHBOR dataset: R1527H, (OR 0.17, P = 0.0018) and a variant that alters a canonical splice donor site, g.16-88737727-C-G Hg38 (OR 0.38, P = 0.02). Both variants showed similar effects in the UK Biobank and the R1527H also in the FinnGen database. Several common variants also reached study-specific thresholds for association in the NEIGHBORHOOD dataset. These results identify novel variants in several mechanosensitive channel genes that show associations with POAG, suggesting that these channels may be potential therapeutic targets.
Collapse
Affiliation(s)
- Wendy W Liu
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, 2370 Watson Court, Palo Alto, CA, 94303, USA.
| | - Tyler G Kinzy
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Jessica N Cooke Bailey
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Zihe Xu
- Department of Ophthalmology, King's College London, St. Thomas' Hospital, London, UK
| | - Pirro Hysi
- Department of Ophthalmology, King's College London, St. Thomas' Hospital, London, UK
- Department of Twin Research and Genetic Epidemiology, King's College London, St. Thomas' Hospital, London, UK
| | - Janey L Wiggs
- Massachusetts Eye and Ear, Harvard Medical School Boston, Boston, MA, USA
| |
Collapse
|
7
|
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: 3.0] [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
|
8
|
Gong J, Chen J, Gu P, Shang Y, Ruppell KT, Yang Y, Wang F, Wen Q, Xiang Y. Shear stress activates nociceptors to drive Drosophila mechanical nociception. Neuron 2022; 110:3727-3742.e8. [PMID: 36087585 DOI: 10.1016/j.neuron.2022.08.015] [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: 09/23/2021] [Revised: 06/07/2022] [Accepted: 08/11/2022] [Indexed: 12/15/2022]
Abstract
Mechanical nociception is essential for animal survival. However, the forces involved in nociceptor activation and the underlying mechanotransduction mechanisms remain elusive. Here, we address these problems by investigating nocifensive behavior in Drosophila larvae. We show that strong poking stimulates nociceptors with a mixture of forces including shear stress and stretch. Unexpectedly, nociceptors are selectively activated by shear stress, but not stretch. Both the shear stress responses of nociceptors and nocifensive behavior require transient receptor potential A1 (TrpA1), which is specifically expressed in nociceptors. We further demonstrate that expression of mammalian or Drosophila TrpA1 in heterologous cells confers responses to shear stress but not stretch. Finally, shear stress activates TrpA1 in a membrane-delimited manner, through modulation of membrane fluidity. Together, our study reveals TrpA1 as an evolutionarily conserved mechanosensitive channel specifically activated by shear stress and suggests a critical role of shear stress in activating nociceptors to drive mechanical nociception.
Collapse
Affiliation(s)
- Jiaxin Gong
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jiazhang Chen
- Department of Physics, Worcester Polytechnic Institute, Worcester, MA 01605, USA
| | - Pengyu Gu
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Ye Shang
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Program in Neuroscience, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Kendra Takle Ruppell
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Program in Neuroscience, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Ying Yang
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Fei Wang
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Program in Neuroscience, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Qi Wen
- Department of Physics, Worcester Polytechnic Institute, Worcester, MA 01605, USA.
| | - Yang Xiang
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Program in Neuroscience, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
9
|
Liu C, Zhang W. Molecular basis of somatosensation in insects. Curr Opin Neurobiol 2022; 76:102592. [DOI: 10.1016/j.conb.2022.102592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 11/16/2022]
|
10
|
Serna JA, Rueda-Gensini L, Céspedes-Valenzuela DN, Cifuentes J, Cruz JC, Muñoz-Camargo C. Recent Advances on Stimuli-Responsive Hydrogels Based on Tissue-Derived ECMs and Their Components: Towards Improving Functionality for Tissue Engineering and Controlled Drug Delivery. Polymers (Basel) 2021; 13:3263. [PMID: 34641079 PMCID: PMC8512780 DOI: 10.3390/polym13193263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/14/2022] Open
Abstract
Due to their highly hydrophilic nature and compositional versatility, hydrogels have assumed a protagonic role in the development of physiologically relevant tissues for several biomedical applications, such as in vivo tissue replacement or regeneration and in vitro disease modeling. By forming interconnected polymeric networks, hydrogels can be loaded with therapeutic agents, small molecules, or cells to deliver them locally to specific tissues or act as scaffolds for hosting cellular development. Hydrogels derived from decellularized extracellular matrices (dECMs), in particular, have gained significant attention in the fields of tissue engineering and regenerative medicine due to their inherently high biomimetic capabilities and endowment of a wide variety of bioactive cues capable of directing cellular behavior. However, these hydrogels often exhibit poor mechanical stability, and their biological properties alone are not enough to direct the development of tissue constructs with functional phenotypes. This review highlights the different ways in which external stimuli (e.g., light, thermal, mechanical, electric, magnetic, and acoustic) have been employed to improve the performance of dECM-based hydrogels for tissue engineering and regenerative medicine applications. Specifically, we outline how these stimuli have been implemented to improve their mechanical stability, tune their microarchitectural characteristics, facilitate tissue morphogenesis and enable precise control of drug release profiles. The strategic coupling of the bioactive features of dECM-based hydrogels with these stimulation schemes grants considerable advances in the development of functional hydrogels for a wide variety of applications within these fields.
Collapse
Affiliation(s)
| | | | | | | | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (J.A.S.); (L.R.-G.); (D.N.C.-V.); (J.C.)
| | - Carolina Muñoz-Camargo
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (J.A.S.); (L.R.-G.); (D.N.C.-V.); (J.C.)
| |
Collapse
|
11
|
Abstract
Mechanosensing is a key feature through which organisms can receive inputs from the environment and convert them into specific functional and behavioral outputs. Mechanosensation occurs in many cells and tissues, regulating a plethora of molecular processes based on the distribution of forces and stresses both at the cell membrane and at the intracellular organelles levels, through complex interactions between cells’ microstructures, cytoskeleton, and extracellular matrix. Although several primary and secondary mechanisms have been shown to contribute to mechanosensation, a fundamental pathway in simple organisms and mammals involves the presence of specialized sensory neurons and the presence of different types of mechanosensitive ion channels on the neuronal cell membrane. In this contribution, we present a review of the main ion channels which have been proven to be significantly involved in mechanotransduction in neurons. Further, we discuss recent studies focused on the biological mechanisms and modeling of mechanosensitive ion channels’ gating, and on mechanotransduction modeling at different scales and levels of details.
Collapse
|
12
|
Kurth F, Tai YK, Parate D, van Oostrum M, Schmid YRF, Toh SJ, Yap JLY, Wollscheid B, Othman A, Dittrich PS, Franco-Obregón A. Cell-Derived Vesicles as TRPC1 Channel Delivery Systems for the Recovery of Cellular Respiratory and Proliferative Capacities. ACTA ACUST UNITED AC 2020; 4:e2000146. [PMID: 32875708 DOI: 10.1002/adbi.202000146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/28/2020] [Indexed: 11/07/2022]
Abstract
Pulsed electromagnetic fields (PEMFs) are capable of specifically activating a TRPC1-mitochondrial axis underlying cell expansion and mitohormetic survival adaptations. This study characterizes cell-derived vesicles (CDVs) generated from C2C12 murine myoblasts and shows that they are equipped with the sufficient molecular machinery to confer mitochondrial respiratory capacity and associated proliferative responses upon their fusion with recipient cells. CDVs derived from wild type C2C12 myoblasts include the cation-permeable transient receptor potential (TRP) channels, TRPC1 and TRPA1, and directly respond to PEMF exposure with TRPC1-mediated calcium entry. By contrast, CDVs derived from C2C12 muscle cells in which TRPC1 has been genetically knocked-down using CRISPR/Cas9 genome editing, do not. Wild type C2C12-derived CDVs are also capable of restoring PEMF-induced proliferative and mitochondrial activation in two C2C12-derived TRPC1 knockdown clonal cell lines in accordance to their endogenous degree of TRPC1 suppression. C2C12 wild type CDVs respond to menthol with calcium entry and accumulation, likewise verifying TRPA1 functional gating and further corroborating compartmental integrity. Proteomic and lipidomic analyses confirm the surface membrane origin of the CDVs providing an initial indication of the minimal cellular machinery required to recover mitochondrial function. CDVs hence possess the potential of restoring respiratory and proliferative capacities to senescent cells and tissues.
Collapse
Affiliation(s)
- Felix Kurth
- Department of Biosystems Science and Engineering, Bioanalytics Group, ETH Zurich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Yee Kit Tai
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore
| | - Dinesh Parate
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore
| | - Marc van Oostrum
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Otto-Stern-Weg 3, Zurich, 8093, Switzerland
| | - Yannick R F Schmid
- Department of Biosystems Science and Engineering, Bioanalytics Group, ETH Zurich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Shi Jie Toh
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore
| | - Jasmine Lye Yee Yap
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore
| | - Bernd Wollscheid
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Otto-Stern-Weg 3, Zurich, 8093, Switzerland
| | - Alaa Othman
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Otto-Stern-Weg 3, Zurich, 8093, Switzerland.,Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, 8093, Switzerland.,Institute for Clinical Chemistry, University Hospital Zurich, Zurich, 8091, Switzerland
| | - Petra S Dittrich
- Department of Biosystems Science and Engineering, Bioanalytics Group, ETH Zurich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore.,Institute for Health Innovation & Technology, iHealthtech, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore
| |
Collapse
|
13
|
Potekhina Y, Filatova A, Tregubova E, Mokhov D. Mechanosensitivity of Cells and Its Role in the Regulation of Physiological Functions and the Implementation of Physiotherapeutic Effects (Review). Sovrem Tekhnologii Med 2020; 12:77-89. [PMID: 34795996 PMCID: PMC8596276 DOI: 10.17691/stm2020.12.4.10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Indexed: 01/11/2023] Open
Abstract
Regulatory signals in the body are not limited to chemical and electrical ones. There is another type of important signals for cells: those are mechanical signals (coming from the environment or arising from within the body), which have been less known in the literature. The review summarizes new information on the mechanosensitivity of various cells of connective tissue and nervous system. Participation of mechanical stimuli in the regulation of growth, development, differentiation, and functioning of tissues is described. The data focus on bone remodeling, wound healing, neurite growth, and the formation of neural networks. Mechanotransduction, cellular organelles, and mechanosensitive molecules involved in these processes are discussed as well as the role of the extracellular matrix. The importance of mechanical characteristics of cells in the pathogenesis of diseases is highlighted. Finally, the possible role of mechanosensitivity in mediating the physiotherapeutic effects is addressed.
Collapse
Affiliation(s)
- Yu.P. Potekhina
- Professor, Department of Normal Physiology named after N.Y. Belenkov; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - A.I. Filatova
- Student, Faculty of Pediatrics; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - E.S. Tregubova
- Professor, Department of Osteopathy; North-Western State Medical University named after I.I. Mechnikov, 41 Kirochnaya St., Saint Petersburg, 191015, Russia; Associate Professor, Institute of Osteopathy; Saint Petersburg State University, 7/9 Universitetskaya naberezhnaya, Saint Petersburg, 199034, Russia
| | - D.E. Mokhov
- Head of the Department of Osteopathy; North-Western State Medical University named after I.I. Mechnikov, 41 Kirochnaya St., Saint Petersburg, 191015, Russia; Director of the Institute of Osteopathy Saint Petersburg State University, 7/9 Universitetskaya naberezhnaya, Saint Petersburg, 199034, Russia
| |
Collapse
|
14
|
Camacho Londoño JE, Kuryshev V, Zorn M, Saar K, Tian Q, Hübner N, Nawroth P, Dietrich A, Birnbaumer L, Lipp P, Dieterich C, Freichel M. Transcriptional signatures regulated by TRPC1/C4-mediated Background Ca 2+ entry after pressure-overload induced cardiac remodelling. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2020; 159:86-104. [PMID: 32738354 DOI: 10.1016/j.pbiomolbio.2020.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/03/2020] [Accepted: 07/21/2020] [Indexed: 01/17/2023]
Abstract
AIMS After summarizing current concepts for the role of TRPC cation channels in cardiac cells and in processes triggered by mechanical stimuli arising e.g. during pressure overload, we analysed the role of TRPC1 and TRPC4 for background Ca2+ entry (BGCE) and for cardiac pressure overload induced transcriptional remodelling. METHODS AND RESULTS Mn2+-quench analysis in cardiomyocytes from several Trpc-deficient mice revealed that both TRPC1 and TRPC4 are required for BGCE. Electrically-evoked cell shortening of cardiomyocytes from TRPC1/C4-DKO mice was reduced, whereas parameters of cardiac contractility and relaxation assessed in vivo were unaltered. As pathological cardiac remodelling in mice depends on their genetic background, and the development of cardiac remodelling was found to be reduced in TRPC1/C4-DKO mice on a mixed genetic background, we studied TRPC1/C4-DKO mice on a C57BL6/N genetic background. Cardiac hypertrophy was reduced in those mice after chronic isoproterenol infusion (-51.4%) or after one week of transverse aortic constriction (TAC; -73.0%). This last manoeuvre was preceded by changes in the pressure overload induced transcriptional program as analysed by RNA sequencing. Genes encoding specific collagens, the Mef2 target myomaxin and the gene encoding the mechanosensitive channel Piezo2 were up-regulated after TAC in wild type but not in TRPC1/C4-DKO hearts. CONCLUSIONS Deletion of the TRPC1 and TRPC4 channel proteins protects against development of pathological cardiac hypertrophy independently of the genetic background. To determine if the TRPC1/C4-dependent changes in the pressure overload induced alterations in the transcriptional program causally contribute to cardio-protection needs to be elaborated in future studies.
Collapse
Affiliation(s)
- Juan E Camacho Londoño
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, 69120, Germany.
| | - Vladimir Kuryshev
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120, Heidelberg, Germany; Innere Medizin III, Bioinformatik und Systemkardiologie, Klaus Tschira Institute for Computational Cardiology, Ruprecht-Karls-Universität Heidelberg, 69120, Heidelberg, Germany
| | - Markus Zorn
- Department of Medicine I and Clinical Chemistry, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Kathrin Saar
- Max-Delbrück-Centrum für Molekulare Medizin (MDC), 13125, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13347, Berlin, Germany
| | - Qinghai Tian
- Medical Faculty, Centre for Molecular Signalling (PZMS), Institute for Molecular Cell Biology and Research Center for Molecular Imaging and Screening, Saarland University, 66421 Homburg/Saar, Germany
| | - Norbert Hübner
- Max-Delbrück-Centrum für Molekulare Medizin (MDC), 13125, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13347, Berlin, Germany; Berlin Institute of Health (BIH), 10178, Berlin, Germany; Charité -Universitätsmedizin, 10117, Berlin, Germany
| | - Peter Nawroth
- Department of Medicine I and Clinical Chemistry, University Hospital Heidelberg, 69120, Heidelberg, Germany; German Center for Diabetes Research (DZD), Germany; Institute for Diabetes and Cancer IDC Helmholtz Center Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Dept. of Medicine I, Heidelberg University Hospital, Heidelberg, Germany
| | - Alexander Dietrich
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Member of the German Center for Lung Research (DZL), Ludwig-Maximilians-Universität, 80336, München, Germany
| | - Lutz Birnbaumer
- Laboratory of Neurobiology, NIEHS, North Carolina, USA and Institute of Biomedical Research (BIOMED), Catholic University of Argentina, C1107AFF Buenos Aires, Argentina
| | - Peter Lipp
- Medical Faculty, Centre for Molecular Signalling (PZMS), Institute for Molecular Cell Biology and Research Center for Molecular Imaging and Screening, Saarland University, 66421 Homburg/Saar, Germany
| | - Christoph Dieterich
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, 69120, Germany; Innere Medizin III, Bioinformatik und Systemkardiologie, Klaus Tschira Institute for Computational Cardiology, Ruprecht-Karls-Universität Heidelberg, 69120, Heidelberg, Germany
| | - Marc Freichel
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, 69120, Germany.
| |
Collapse
|
15
|
De Felice D, Alaimo A. Mechanosensitive Piezo Channels in Cancer: Focus on altered Calcium Signaling in Cancer Cells and in Tumor Progression. Cancers (Basel) 2020; 12:cancers12071780. [PMID: 32635333 PMCID: PMC7407875 DOI: 10.3390/cancers12071780] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/16/2022] Open
Abstract
Mechanotransduction, the translation of mechanical stimuli into biological signals, is a crucial mechanism involved in the function of fundamentally all cell types. In many solid tumors, the malignant transformation is often associated with drastic changes in cell mechanical features. Extracellular matrix stiffness, invasive growth, and cell mobility are just a few hallmarks present in cancer cells that, by inducing mechanical stimuli, create positive feedbacks promoting cancer development. Among the molecular players involved in these pathophysiological processes, the mechanosensitive Ca2+-permeable Piezo channels have emerged as major transducers of mechanical stress into Ca2+ dependent signals. Piezo channels are overexpressed in several cancers, such as in breast, gastric, and bladder, whereas their downregulation has been described in other cancers. Still, the roles of mechanosensitive Piezos in cancer are somewhat puzzling. In this review, we summarize the current knowledge on the pathophysiological roles of these Ca2+-permeable channels, with special emphasis on their functional involvement in different cancer types progression.
Collapse
|
16
|
Jin P, Jan LY, Jan YN. Mechanosensitive Ion Channels: Structural Features Relevant to Mechanotransduction Mechanisms. Annu Rev Neurosci 2020; 43:207-229. [PMID: 32084327 DOI: 10.1146/annurev-neuro-070918-050509] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Activation of mechanosensitive ion channels underlies a variety of fundamental physiological processes that require sensation of mechanical force. Different mechanosensitive channels adapt distinctive structures and mechanotransduction mechanisms to fit their biological roles. How mechanosensitive channels work, especially in animals, has been extensively studied in the past decade. Here we review key findings in the functional and structural characterizations of these channels and highlight the structural features relevant to the mechanotransduction mechanism of each specific channel.
Collapse
Affiliation(s)
- Peng Jin
- Department of Physiology, University of California, San Francisco, California 94158, USA;
| | - Lily Yeh Jan
- Department of Physiology, University of California, San Francisco, California 94158, USA; .,Department of Biochemistry and Biophysics and Howard Hughes Medical Institute, University of California, San Francisco, California 94158, USA
| | - Yuh-Nung Jan
- Department of Physiology, University of California, San Francisco, California 94158, USA; .,Department of Biochemistry and Biophysics and Howard Hughes Medical Institute, University of California, San Francisco, California 94158, USA
| |
Collapse
|
17
|
Paraschiv A, Hegde S, Ganti R, Pilizota T, Šarić A. Dynamic Clustering Regulates Activity of Mechanosensitive Membrane Channels. PHYSICAL REVIEW LETTERS 2020; 124:048102. [PMID: 32058787 DOI: 10.1103/physrevlett.124.048102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Indexed: 06/10/2023]
Abstract
Experiments have suggested that bacterial mechanosensitive channels separate into 2D clusters, the role of which is unclear. By developing a coarse-grained computer model we find that clustering promotes the channel closure, which is highly dependent on the channel concentration and membrane stress. This behaviour yields a tightly regulated gating system, whereby at high tensions channels gate individually, and at lower tensions the channels spontaneously aggregate and inactivate. We implement this positive feedback into the model for cell volume regulation, and find that the channel clustering protects the cell against excessive loss of cytoplasmic content.
Collapse
Affiliation(s)
- Alexandru Paraschiv
- Department of Physics and Astronomy, Institute for the Physics of Living Systems University College London, London WC1E 6BT, United Kingdom
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
| | - Smitha Hegde
- Centre for Synthetic and Systems Biology University of Edinburgh, Edinburgh EH9 3FF, United Kingdom
| | - Raman Ganti
- Institute for Medical Engineering and Science Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Teuta Pilizota
- Centre for Synthetic and Systems Biology University of Edinburgh, Edinburgh EH9 3FF, United Kingdom
| | - Anđela Šarić
- Department of Physics and Astronomy, Institute for the Physics of Living Systems University College London, London WC1E 6BT, United Kingdom
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
18
|
Martinac B, Nikolaev YA, Silvani G, Bavi N, Romanov V, Nakayama Y, Martinac AD, Rohde P, Bavi O, Cox CD. Cell membrane mechanics and mechanosensory transduction. CURRENT TOPICS IN MEMBRANES 2020; 86:83-141. [DOI: 10.1016/bs.ctm.2020.08.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
19
|
Nikolaev YA, Cox CD, Ridone P, Rohde PR, Cordero-Morales JF, Vásquez V, Laver DR, Martinac B. Mammalian TRP ion channels are insensitive to membrane stretch. J Cell Sci 2019; 132:jcs238360. [PMID: 31722978 PMCID: PMC6918743 DOI: 10.1242/jcs.238360] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 10/29/2019] [Indexed: 12/27/2022] Open
Abstract
TRP channels of the transient receptor potential ion channel superfamily are involved in a wide variety of mechanosensory processes, including touch sensation, pain, blood pressure regulation, bone loading and detection of cerebrospinal fluid flow. However, in many instances it is unclear whether TRP channels are the primary transducers of mechanical force in these processes. In this study, we tested stretch activation of eleven TRP channels from six mammalian subfamilies. We found that these TRP channels were insensitive to short membrane stretches in cellular systems. Furthermore, we purified TRPC6 and demonstrated its insensitivity to stretch in liposomes, an artificial bilayer system free from cellular components. Additionally, we demonstrated that, when expressed in C. elegans neurons, mouse TRPC6 restores the mechanoresponse of a touch insensitive mutant but requires diacylglycerol for activation. These results strongly suggest that the mammalian members of the TRP ion channel family are insensitive to tension induced by cell membrane stretching and, thus, are more likely to be activated by cytoplasmic tethers or downstream components and to act as amplifiers of cellular mechanosensory signaling cascades.
Collapse
Affiliation(s)
- Yury A Nikolaev
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney 2010, Australia
- Human Physiology, School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle 2308, Australia
| | - Charles D Cox
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney 2010, Australia
| | - Pietro Ridone
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney 2010, Australia
| | - Paul R Rohde
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney 2010, Australia
| | - Julio F Cordero-Morales
- Department of Physiology, College of Medicine, The University of Tennessee Health Science Center, Memphis 38163, USA
| | - Valeria Vásquez
- Department of Physiology, College of Medicine, The University of Tennessee Health Science Center, Memphis 38163, USA
| | - Derek R Laver
- Human Physiology, School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle 2308, Australia
| | - Boris Martinac
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney 2010, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney 2052, Australia
| |
Collapse
|
20
|
Yap JLY, Tai YK, Fröhlich J, Fong CHH, Yin JN, Foo ZL, Ramanan S, Beyer C, Toh SJ, Casarosa M, Bharathy N, Kala MP, Egli M, Taneja R, Lee CN, Franco-Obregón A. Ambient and supplemental magnetic fields promote myogenesis via a TRPC1-mitochondrial axis: evidence of a magnetic mitohormetic mechanism. FASEB J 2019; 33:12853-12872. [PMID: 31518158 DOI: 10.1096/fj.201900057r] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We show that both supplemental and ambient magnetic fields modulate myogenesis. A lone 10 min exposure of myoblasts to 1.5 mT amplitude supplemental pulsed magnetic fields (PEMFs) accentuated in vitro myogenesis by stimulating transient receptor potential (TRP)-C1-mediated calcium entry and downstream nuclear factor of activated T cells (NFAT)-transcriptional and P300/CBP-associated factor (PCAF)-epigenetic cascades, whereas depriving myoblasts of ambient magnetic fields slowed myogenesis, reduced TRPC1 expression, and silenced NFAT-transcriptional and PCAF-epigenetic cascades. The expression levels of peroxisome proliferator-activated receptor γ coactivator 1α, the master regulator of mitochondriogenesis, was also enhanced by brief PEMF exposure. Accordingly, mitochondriogenesis and respiratory capacity were both enhanced with PEMF exposure, paralleling TRPC1 expression and pharmacological sensitivity. Clustered regularly interspaced short palindromic repeats-Cas9 knockdown of TRPC1 precluded proliferative and mitochondrial responses to supplemental PEMFs, whereas small interfering RNA gene silencing of TRPM7 did not, coinciding with data that magnetoreception did not coincide with the expression or function of other TRP channels. The aminoglycoside antibiotics antagonized and down-regulated TRPC1 expression and, when applied concomitantly with PEMF exposure, attenuated PEMF-stimulated calcium entry, mitochondrial respiration, proliferation, differentiation, and epigenetic directive in myoblasts, elucidating why the developmental potential of magnetic fields may have previously escaped detection. Mitochondrial-based survival adaptations were also activated upon PEMF stimulation. Magnetism thus deploys an authentic myogenic directive that relies on an interplay between mitochondria and TRPC1 to reach fruition.-Yap, J. L. Y., Tai, Y. K., Fröhlich, J., Fong, C. H. H., Yin, J. N., Foo, Z. L., Ramanan, S., Beyer, C., Toh, S. J., Casarosa, M., Bharathy, N., Kala, M. P., Egli, M., Taneja, R., Lee, C. N., Franco-Obregón, A. Ambient and supplemental magnetic fields promote myogenesis via a TRPC1-mitochondrial axis: evidence of a magnetic mitohormetic mechanism.
Collapse
Affiliation(s)
- Jasmine Lye Yee Yap
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems (BICEPS) Laboratory, National University of Singapore, Singapore
| | - Yee Kit Tai
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems (BICEPS) Laboratory, National University of Singapore, Singapore
| | - Jürg Fröhlich
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Institute for Electromagnetic Fields, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | - Charlene Hui Hua Fong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems (BICEPS) Laboratory, National University of Singapore, Singapore
| | - Jocelyn Naixin Yin
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems (BICEPS) Laboratory, National University of Singapore, Singapore
| | - Zi Ling Foo
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems (BICEPS) Laboratory, National University of Singapore, Singapore
| | - Sharanya Ramanan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems (BICEPS) Laboratory, National University of Singapore, Singapore
| | - Christian Beyer
- Institute for Electromagnetic Fields, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland.,Centre Suisse d'Électronique et de Microtechnique (CSEM SA), Neuchâtel, Switzerland
| | - Shi Jie Toh
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems (BICEPS) Laboratory, National University of Singapore, Singapore
| | - Marco Casarosa
- Department of Experimental and Clinical Biomedical Sciences Mario Serio, University of Florence, Florence, Italy
| | - Narendra Bharathy
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Children's Cancer Therapy Development Institute, Beaverton, Oregon, USA
| | - Monica Palanichamy Kala
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Marcel Egli
- Institute of Medical Engineering, Lucerne University of Applied Sciences and Arts, Hergiswil, Switzerland; and
| | - Reshma Taneja
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chuen Neng Lee
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Institute for Health Innovation and Technology, iHealthtech, National University of Singapore, Singapore
| | - Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems (BICEPS) Laboratory, National University of Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Institute for Health Innovation and Technology, iHealthtech, National University of Singapore, Singapore
| |
Collapse
|
21
|
Canales J, Morales D, Blanco C, Rivas J, Díaz N, Angelopoulos I, Cerda O. A TR(i)P to Cell Migration: New Roles of TRP Channels in Mechanotransduction and Cancer. Front Physiol 2019; 10:757. [PMID: 31275168 PMCID: PMC6591513 DOI: 10.3389/fphys.2019.00757] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/31/2019] [Indexed: 12/20/2022] Open
Abstract
Cell migration is a key process in cancer metastasis, allowing malignant cells to spread from the primary tumor to distant organs. At the molecular level, migration is the result of several coordinated events involving mechanical forces and cellular signaling, where the second messenger Ca2+ plays a pivotal role. Therefore, elucidating the regulation of intracellular Ca2+ levels is key for a complete understanding of the mechanisms controlling cellular migration. In this regard, understanding the function of Transient Receptor Potential (TRP) channels, which are fundamental determinants of Ca2+ signaling, is critical to uncovering mechanisms of mechanotransduction during cell migration and, consequently, in pathologies closely linked to it, such as cancer. Here, we review recent studies on the association between TRP channels and migration-related mechanotransduction events, as well as in the involvement of TRP channels in the migration-dependent pathophysiological process of metastasis.
Collapse
Affiliation(s)
- Jimena Canales
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases, Santiago, Chile
| | - Diego Morales
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases, Santiago, Chile
| | - Constanza Blanco
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases, Santiago, Chile
| | - José Rivas
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases, Santiago, Chile
| | - Nicolás Díaz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases, Santiago, Chile
| | - Ioannis Angelopoulos
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases, Santiago, Chile
| | - Oscar Cerda
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases, Santiago, Chile.,The Wound Repair, Treatment and Health (WoRTH) Initiative, Santiago, Chile
| |
Collapse
|
22
|
Growth Factor Signaling Regulates Mechanical Nociception in Flies and Vertebrates. J Neurosci 2019; 39:6012-6030. [PMID: 31138657 DOI: 10.1523/jneurosci.2950-18.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 05/15/2019] [Accepted: 05/21/2019] [Indexed: 02/06/2023] Open
Abstract
Mechanical sensitization is one of the most difficult clinical pain problems to treat. However, the molecular and genetic bases of mechanical nociception are unclear. Here we develop a Drosophila model of mechanical nociception to investigate the ion channels and signaling pathways that regulate mechanical nociception. We fabricated von Frey filaments that span the subthreshold to high noxious range for Drosophila larvae. Using these, we discovered that pressure (force/area), rather than force per se, is the main determinant of aversive rolling responses to noxious mechanical stimuli. We demonstrated that the RTK PDGF/VEGF receptor (Pvr) and its ligands (Pvfs 2 and 3) are required for mechanical nociception and normal dendritic branching. Pvr is expressed and functions in class IV sensory neurons, whereas Pvf2 and Pvf3 are produced by multiple tissues. Constitutive overexpression of Pvr and its ligands or inducible overexpression of Pvr led to mechanical hypersensitivity that could be partially separated from morphological effects. Genetic analyses revealed that the Piezo and Pain ion channels are required for mechanical hypersensitivity observed upon ectopic activation of Pvr signaling. PDGF, but not VEGF, peptides caused mechanical hypersensitivity in rats. Pharmacological inhibition of VEGF receptor Type 2 (VEGFR-2) signaling attenuated mechanical nociception in rats, suggesting a conserved role for PDGF and VEGFR-2 signaling in regulating mechanical nociception. VEGFR-2 inhibition also attenuated morphine analgesic tolerance in rats. Our results reveal that a conserved RTK signaling pathway regulates baseline mechanical nociception in flies and rats.SIGNIFICANCE STATEMENT Hypersensitivity to touch is poorly understood and extremely difficult to treat. Using a refined Drosophila model of mechanical nociception, we discovered a conserved VEGF-related receptor tyrosine kinase signaling pathway that regulates mechanical nociception in flies. Importantly, pharmacological inhibition of VEGF receptor Type 2 signaling in rats causes analgesia and blocks opioid tolerance. We have thus established a robust, genetically tractable system for the rapid identification and functional analysis of conserved genes underlying mechanical pain sensitivity.
Collapse
|
23
|
Schlosser G. A Short History of Nearly Every Sense-The Evolutionary History of Vertebrate Sensory Cell Types. Integr Comp Biol 2019; 58:301-316. [PMID: 29741623 DOI: 10.1093/icb/icy024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Evolving from filter feeding chordate ancestors, vertebrates adopted a more active life style. These ecological and behavioral changes went along with an elaboration of the vertebrate head including novel complex paired sense organs such as the eyes, inner ears, and olfactory epithelia. However, the photoreceptors, mechanoreceptors, and chemoreceptors used in these sense organs have a long evolutionary history and homologous cell types can be recognized in many other bilaterians or even cnidarians. After briefly introducing some of the major sensory cell types found in vertebrates, this review summarizes the phylogenetic distribution of sensory cell types in metazoans and presents a scenario for the evolutionary history of various sensory cell types involving several cell type diversification and fusion events. It is proposed that the evolution of novel cranial sense organs in vertebrates involved the redeployment of evolutionarily ancient sensory cell types for building larger and more complex sense organs.
Collapse
Affiliation(s)
- Gerhard Schlosser
- School of Natural Sciences and Regenerative Medicine Institute (REMEDI), National University of Ireland, Biomedical Sciences Building, Newcastle Road, Galway H91 TK33, Ireland
| |
Collapse
|
24
|
[Correlation between expressions of VEGF and TRPC6 and their roles in podocyte injury in rats with diabetic nephropathy]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38. [PMID: 29643035 PMCID: PMC6744170 DOI: 10.3969/j.issn.1673-4254.2018.03.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVE To analyze the correlation between the expressions of vascular endothelial growth factor (VEGF) and transient receptor potential canonical 6 (TRPC6) and their role in podocyte injury in rats with diabetic nephropathy. METHODS Forty SD rats with diabetic nephropathy induced by intraperitoneal injection of 65 mg/kg streptozotocin were randomized equally into 5 groups, including a diabetic nephropathy model group and 4 treatment groups, with 8 normal SD rats as the normal control group. In the 4 treatment groups, the rats received intraperitoneal injections with SU5416 at 5 mg/kg or 10 mg/kg twice a week or with LY294002 at 1 mg/kg or 2 mg/kg once daily for 8 weeks. Blood glucose, serum creatinine, blood urea nitrogen, and 24-h urinary protein levels of the rats were detected at different time points, and the pathologies in the renal tissue were observed using HE staining, PAS staining and immunohistochemistry. The expressions of VEGF, nephrin, and TRPC6 at mRNA and protein levels were detected using RT-PCR and Western blotting. RESULTS Compared with normal control rats, the diabetic rats showed significantly increased fasting blood glucose, serum creatinine, blood urea nitrogen and 24-h urinary protein levels with decreased expressions of nephrin mRNA and protein (P<0.05) and increased expressions of VEGF and TRPC6 (P<0.05). Compared with the untreated diabetic rats, the rats with SU5416 treatment showed increased 24-h urinary protein, urea nitrogen, and nephrin expression and decreased TRPC6 expression without significant changes in fasting blood glucose, serum creatinine, or VEGF expression. The rats treated with LY294002 showed decreased 24-h urinary protein and TRPC6 expression without significant changes in fasting blood glucose, serum creatinine, urea nitrogen, or expressions of nephrin and VEGF. CONCLUSION The regulatory effect of VEGF on TRPC6 can be blocked by inhibiting VEGFR-2 or blocking PI3K/Akt signaling pathway.
Collapse
|
25
|
Luo X, Li M, Zhan K, Yang W, Zhang L, Wang K, Yu P, Zhang L. Selective inhibition of TRPM2 channel by two novel synthesized ADPR analogues. Chem Biol Drug Des 2018; 91:552-566. [PMID: 29034580 PMCID: PMC5813235 DOI: 10.1111/cbdd.13119] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 07/04/2017] [Accepted: 08/21/2017] [Indexed: 01/02/2023]
Abstract
Transient receptor potential melastatin-2 (TRPM2) channel critical for monitoring internal body temperature is implicated in the pathological processes such as neurodegeneration. However, lacking selective and potent TRPM2 inhibitors impedes investigation and validation of the channel as a drug target. To discover novel and selective TRPM2 inhibitors, a series of adenosine 5'-diphosphoribose analogues were synthesized, and their activities and selectivity were evaluated. Whole-cell patch-clamp recordings were employed for screen and evaluation of synthesized compounds. Two compounds, 7i and 8a, were identified as TRPM2 inhibitors with IC50 of 5.7 and 5.4 μm, respectively. Both 7i and 8a inhibited TRPM2 current without affecting TRPM7, TRPM8, TRPV1 and TRPV3. These two TRPM2 inhibitors can serve as new pharmacological tools for further investigation and validation of TRPM2 channel as a drug target, and the summarized structure-activity relationship (SAR) may also provide insights into further improving existing inhibitors as potential lead compounds.
Collapse
Affiliation(s)
- Xiao Luo
- State Key Laboratory of Natural and Biomimetic DrugsPeking UniversityBeijingChina
| | - Meng Li
- Department of NeurobiologyNeuroscience Research InstitutePeking University Health Science CenterPeking University School of Pharmaceutical SciencesBeijingChina
| | - Kaiyu Zhan
- Department of NeurobiologyZhejiang University School of MedicineHangzhouZhejiangChina
| | - Wei Yang
- Department of NeurobiologyZhejiang University School of MedicineHangzhouZhejiangChina
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic DrugsPeking UniversityBeijingChina
| | - KeWei Wang
- Department of NeurobiologyNeuroscience Research InstitutePeking University Health Science CenterPeking University School of Pharmaceutical SciencesBeijingChina
- Department of PharmacologySchool of PharmacyQingdao UniversityQingdaoChina
| | - Peilin Yu
- Department of ToxicologySchool of Public HealthZhejiang UniversityHangzhouZhejiangChina
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic DrugsPeking UniversityBeijingChina
| |
Collapse
|
26
|
Zheng W, Hu R, Cai R, Hofmann L, Hu Q, Fatehi M, Long W, Kong T, Tang J, Light P, Flockerzi V, Cao Y, Chen X. Identification and characterization of hydrophobic gate residues in TRP channels. FASEB J 2018; 32:639-653. [DOI: 10.1096/fj.201700599rr] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Wang Zheng
- Institute of Biomedical and Pharmaceutical SciencesKey Laboratory of Fermentation Engineering of Ministry of EducationCollege of BioengineeringHubei University of TechnologyWuhanChina
- Membrane Protein Disease Research GroupDepartment of PhysiologyTongji UniversityShanghaiChina
| | - Ruikun Hu
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Ruiqi Cai
- Membrane Protein Disease Research GroupDepartment of PhysiologyTongji UniversityShanghaiChina
| | - Laura Hofmann
- Experimentelle und Klinische Pharmakologie und ToxikologieUniversität des SaarlandesHomburgGermany
| | - Qiaolin Hu
- Membrane Protein Disease Research GroupDepartment of PhysiologyTongji UniversityShanghaiChina
| | - Mohammad Fatehi
- Department of PharmacologyFaculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - Wentong Long
- Department of PharmacologyFaculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - Tim Kong
- Membrane Protein Disease Research GroupDepartment of PhysiologyTongji UniversityShanghaiChina
| | - Jingfeng Tang
- Institute of Biomedical and Pharmaceutical SciencesKey Laboratory of Fermentation Engineering of Ministry of EducationCollege of BioengineeringHubei University of TechnologyWuhanChina
| | - Peter Light
- Department of PharmacologyFaculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - Veit Flockerzi
- Experimentelle und Klinische Pharmakologie und ToxikologieUniversität des SaarlandesHomburgGermany
| | - Ying Cao
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Xing‐Zhen Chen
- Membrane Protein Disease Research GroupDepartment of PhysiologyTongji UniversityShanghaiChina
| |
Collapse
|
27
|
Parate D, Franco-Obregón A, Fröhlich J, Beyer C, Abbas AA, Kamarul T, Hui JHP, Yang Z. Enhancement of mesenchymal stem cell chondrogenesis with short-term low intensity pulsed electromagnetic fields. Sci Rep 2017; 7:9421. [PMID: 28842627 PMCID: PMC5572790 DOI: 10.1038/s41598-017-09892-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/28/2017] [Indexed: 12/22/2022] Open
Abstract
Pulse electromagnetic fields (PEMFs) have been shown to recruit calcium-signaling cascades common to chondrogenesis. Here we document the effects of specified PEMF parameters over mesenchymal stem cells (MSC) chondrogenic differentiation. MSCs undergoing chondrogenesis are preferentially responsive to an electromagnetic efficacy window defined by field amplitude, duration and frequency of exposure. Contrary to conventional practice of administering prolonged and repetitive exposures to PEMFs, optimal chondrogenic outcome is achieved in response to brief (10 minutes), low intensity (2 mT) exposure to 6 ms bursts of magnetic pulses, at 15 Hz, administered only once at the onset of chondrogenic induction. By contrast, repeated exposures diminished chondrogenic outcome and could be attributed to calcium entry after the initial induction. Transient receptor potential (TRP) channels appear to mediate these aspects of PEMF stimulation, serving as a conduit for extracellular calcium. Preventing calcium entry during the repeated PEMF exposure with the co-administration of EGTA or TRP channel antagonists precluded the inhibition of differentiation. This study highlights the intricacies of calcium homeostasis during early chondrogenesis and the constraints that are placed on PEMF-based therapeutic strategies aimed at promoting MSC chondrogenesis. The demonstrated efficacy of our optimized PEMF regimens has clear clinical implications for future regenerative strategies for cartilage.
Collapse
Affiliation(s)
- Dinesh Parate
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 11, 1E Kent Ridge Road, Singapore, 119288, Singapore
| | - Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 8, IE Kent Ridge Road, Singapore, 119228, Singapore. .,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, MD6, 14 medical Drive, #14-01, Singapore, 117599, Singapore.
| | - Jürg Fröhlich
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 8, IE Kent Ridge Road, Singapore, 119228, Singapore.,Institute for Electromagnetic Fields, Swiss Federal Institute of Technology (ETH), Rämistrasse 101, 8092, Zurich, Switzerland
| | - Christian Beyer
- Institute for Electromagnetic Fields, Swiss Federal Institute of Technology (ETH), Rämistrasse 101, 8092, Zurich, Switzerland
| | - Azlina A Abbas
- Tissue Engineering Group (TEG), National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Pantai Valley, Kuala Lumpur, 50603, Malaysia
| | - Tunku Kamarul
- Tissue Engineering Group (TEG), National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Pantai Valley, Kuala Lumpur, 50603, Malaysia
| | - James H P Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 11, 1E Kent Ridge Road, Singapore, 119288, Singapore. .,Tissue Engineering Program, Life Sciences Institute, National University of Singapore, DSO (Kent Ridge) Building, #04-01, 27 Medical Drive, Singapore, 117510, Singapore.
| | - Zheng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 11, 1E Kent Ridge Road, Singapore, 119288, Singapore. .,Tissue Engineering Program, Life Sciences Institute, National University of Singapore, DSO (Kent Ridge) Building, #04-01, 27 Medical Drive, Singapore, 117510, Singapore.
| |
Collapse
|
28
|
Thorpe SD, Lee DA. Dynamic regulation of nuclear architecture and mechanics-a rheostatic role for the nucleus in tailoring cellular mechanosensitivity. Nucleus 2017; 8:287-300. [PMID: 28152338 PMCID: PMC5499908 DOI: 10.1080/19491034.2017.1285988] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Nuclear architecture, a function of both chromatin and nucleoskeleton structure, is known to change with stem cell differentiation and differs between various somatic cell types. These changes in nuclear architecture are associated with the regulation of gene expression and genome function in a cell-type specific manner. Biophysical stimuli are known effectors of differentiation and also elicit stimuli-specific changes in nuclear architecture. This occurs via the process of mechanotransduction whereby extracellular mechanical forces activate several well characterized signaling cascades of cytoplasmic origin, and potentially some recently elucidated signaling cascades originating in the nucleus. Recent work has demonstrated changes in nuclear mechanics both with pluripotency state in embryonic stem cells, and with differentiation progression in adult mesenchymal stem cells. This review explores the interplay between cytoplasmic and nuclear mechanosensitivity, highlighting a role for the nucleus as a rheostat in tuning the cellular mechano-response.
Collapse
Affiliation(s)
- Stephen D Thorpe
- a Institute of Bioengineering, School of Engineering and Materials Science , Queen Mary University of London , London , UK
| | - David A Lee
- a Institute of Bioengineering, School of Engineering and Materials Science , Queen Mary University of London , London , UK
| |
Collapse
|
29
|
|
30
|
Iyer SR, Shah SB, Valencia AP, Schneider MF, Hernández-Ochoa EO, Stains JP, Blemker SS, Lovering RM. Altered nuclear dynamics in MDX myofibers. J Appl Physiol (1985) 2016; 122:470-481. [PMID: 27979987 DOI: 10.1152/japplphysiol.00857.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/02/2016] [Accepted: 12/06/2016] [Indexed: 01/17/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disorder in which the absence of dystrophin leads to progressive muscle degeneration and weakness. Although the genetic basis is known, the pathophysiology of dystrophic skeletal muscle remains unclear. We examined nuclear movement in wild-type (WT) and muscular dystrophy mouse model for DMD (MDX) (dystrophin-null) mouse myofibers. We also examined expression of proteins in the linkers of nucleoskeleton and cytoskeleton (LINC) complex, as well as nuclear transcriptional activity via histone H3 acetylation and polyadenylate-binding nuclear protein-1. Because movement of nuclei is not only LINC dependent but also microtubule dependent, we analyzed microtubule density and organization in WT and MDX myofibers, including the application of a unique 3D tool to assess microtubule core structure. Nuclei in MDX myofibers were more mobile than in WT myofibers for both distance traveled and velocity. MDX muscle shows reduced expression and labeling intensity of nesprin-1, a LINC protein that attaches the nucleus to the microtubule and actin cytoskeleton. MDX nuclei also showed altered transcriptional activity. Previous studies established that microtubule structure at the cortex is disrupted in MDX myofibers; our analyses extend these findings by showing that microtubule structure in the core is also disrupted. In addition, we studied malformed MDX myofibers to better understand the role of altered myofiber morphology vs. microtubule architecture in the underlying susceptibility to injury seen in dystrophic muscles. We incorporated morphological and microtubule architectural concepts into a simplified finite element mathematical model of myofiber mechanics, which suggests a greater contribution of myofiber morphology than microtubule structure to muscle biomechanical performance.NEW & NOTEWORTHY Microtubules provide the means for nuclear movement but show altered organization in the muscular dystrophy mouse model (MDX) (dystrophin-null) muscle. Here, MDX myofibers show increased nuclear movement, altered transcriptional activity, and altered linkers of nucleoskeleton and cytoskeleton complex expression compared with healthy myofibers. Microtubule architecture was incorporated in finite element modeling of passive stretch, revealing a role of fiber malformation, commonly found in MDX muscle. The results suggest that alterations in microtubule architecture in MDX muscle affect nuclear movement, which is essential for muscle function.
Collapse
Affiliation(s)
- Shama R Iyer
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland
| | - Sameer B Shah
- Departments of Orthopaedic Surgery and Bioengineering, University of California San Diego, La Jolla, California
| | - Ana P Valencia
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland
| | - Martin F Schneider
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Erick O Hernández-Ochoa
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Joseph P Stains
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland
| | - Silvia S Blemker
- Department of Biomedical Engineering and Mechanical and Aerospace Engineering, University of Virginia, Charlottesville, Virginia; and
| | - Richard M Lovering
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland; .,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
31
|
Abstract
Cell types are the basic building blocks of multicellular organisms and are extensively diversified in animals. Despite recent advances in characterizing cell types, classification schemes remain ambiguous. We propose an evolutionary definition of a cell type that allows cell types to be delineated and compared within and between species. Key to cell type identity are evolutionary changes in the 'core regulatory complex' (CoRC) of transcription factors, that make emergent sister cell types distinct, enable their independent evolution and regulate cell type-specific traits termed apomeres. We discuss the distinction between developmental and evolutionary lineages, and present a roadmap for future research.
Collapse
|
32
|
Anguita E, Villalobo A. Src-family tyrosine kinases and the Ca 2+ signal. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:915-932. [PMID: 27818271 DOI: 10.1016/j.bbamcr.2016.10.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/25/2016] [Accepted: 10/30/2016] [Indexed: 01/08/2023]
Abstract
In this review, we shall describe the rich crosstalk between non-receptor Src-family kinases (SFKs) and the Ca2+ transient generated in activated cells by a variety of extracellular and intracellular stimuli, resulting in diverse signaling events. The exchange of information between SFKs and Ca2+ is reciprocal, as it flows in both directions. These kinases are main actors in pathways leading to the generation of the Ca2+ signal, and reciprocally, the Ca2+ signal modulates SFKs activity and functions. We will cover how SFKs participate in the generation of the cytosolic Ca2+ rise upon activation of a series of receptors and the mechanism of clearance of this Ca2+ signal. The role of SFKs modulating Ca2+-translocating channels participating in these events will be amply discussed. Finally, the role of the Ca2+ sensor protein calmodulin on the activity of c-Src, and potentially on other SFKs, will be outlined as well. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
Affiliation(s)
- Estefanía Anguita
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029 Madrid, Spain
| | - Antonio Villalobo
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029 Madrid, Spain.
| |
Collapse
|
33
|
Mittal R, Aranke M, Debs LH, Nguyen D, Patel AP, Grati M, Mittal J, Yan D, Chapagain P, Eshraghi AA, Liu XZ. Indispensable Role of Ion Channels and Transporters in the Auditory System. J Cell Physiol 2016; 232:743-758. [DOI: 10.1002/jcp.25631] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 09/30/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Mayank Aranke
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Luca H. Debs
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Desiree Nguyen
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Amit P. Patel
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - M'hamed Grati
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Jeenu Mittal
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Denise Yan
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Prem Chapagain
- Department of Physics; Florida International University; Miami Florida
- Biomolecular Science Institute; Florida International University; Miami Florida
| | - Adrien A. Eshraghi
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Xue Zhong Liu
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| |
Collapse
|
34
|
Wu X, Indzhykulian AA, Niksch PD, Webber RM, Garcia-Gonzalez M, Watnick T, Zhou J, Vollrath MA, Corey DP. Hair-Cell Mechanotransduction Persists in TRP Channel Knockout Mice. PLoS One 2016; 11:e0155577. [PMID: 27196058 PMCID: PMC4873267 DOI: 10.1371/journal.pone.0155577] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 05/01/2016] [Indexed: 12/17/2022] Open
Abstract
Members of the TRP superfamily of ion channels mediate mechanosensation in some organisms, and have been suggested as candidates for the mechanotransduction channel in vertebrate hair cells. Some TRP channels can be ruled out based on lack of an inner ear phenotype in knockout animals or pore properties not similar to the hair-cell channel. Such studies have excluded Trpv4, Trpa1, Trpml3, Trpm1, Trpm3, Trpc1, Trpc3, Trpc5, and Trpc6. However, others remain reasonable candidates. We used data from an RNA-seq analysis of gene expression in hair cells as well as data on TRP channel conductance to narrow the candidate group. We then characterized mice lacking functional Trpm2, Pkd2, Pkd2l1, Pkd2l2 and Pkd1l3, using scanning electron microscopy, auditory brainstem response, permeant dye accumulation, and single-cell electrophysiology. In all of these TRP-deficient mice, and in double and triple knockouts, mechanotransduction persisted. Together with published studies, these results argue against the participation of any of the 33 mouse TRP channels in hair cell transduction.
Collapse
MESH Headings
- Animals
- Calcium Channels/genetics
- Cochlea/physiology
- Ear, Inner/physiology
- Evoked Potentials, Auditory, Brain Stem/genetics
- Gene Expression Profiling
- Gene Expression Regulation
- Hair Cells, Auditory/physiology
- Hearing
- Mechanotransduction, Cellular
- Membrane Glycoproteins/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Electron, Scanning
- Patch-Clamp Techniques
- Receptors, Cell Surface/genetics
- TRPM Cation Channels/genetics
- TRPP Cation Channels/genetics
- Transient Receptor Potential Channels/genetics
Collapse
Affiliation(s)
- Xudong Wu
- Department of Neurobiology, Harvard Medical School and Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
| | - Artur A. Indzhykulian
- Department of Neurobiology, Harvard Medical School and Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
| | - Paul D. Niksch
- Department of Neurobiology, Harvard Medical School and Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
| | - Roxanna M. Webber
- Department of Neurobiology, Harvard Medical School and Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
| | - Miguel Garcia-Gonzalez
- Department of Medicine, Division of Nephrology, University of Maryland, Baltimore, Maryland, United States of America
| | - Terry Watnick
- Department of Medicine, Division of Nephrology, University of Maryland, Baltimore, Maryland, United States of America
| | - Jing Zhou
- Renal Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Melissa A. Vollrath
- Department of Neurobiology, Harvard Medical School and Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
- Department of Physiology, McGill University Montréal, Québec, Canada
| | - David P. Corey
- Department of Neurobiology, Harvard Medical School and Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
35
|
Evidence for the involvement of ASIC3 in sensory mechanotransduction in proprioceptors. Nat Commun 2016; 7:11460. [PMID: 27161260 PMCID: PMC4866049 DOI: 10.1038/ncomms11460] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 03/29/2016] [Indexed: 01/12/2023] Open
Abstract
Acid-sensing ion channel 3 (ASIC3) is involved in acid nociception, but its possible role in neurosensory mechanotransduction is disputed. We report here the generation of Asic3-knockout/eGFPf-knockin mice and subsequent characterization of heterogeneous expression of ASIC3 in the dorsal root ganglion (DRG). ASIC3 is expressed in parvalbumin (Pv+) proprioceptor axons innervating muscle spindles. We further generate a floxed allele of Asic3 (Asic3f/f) and probe the role of ASIC3 in mechanotransduction in neurite-bearing Pv+ DRG neurons through localized elastic matrix movements and electrophysiology. Targeted knockout of Asic3 disrupts spindle afferent sensitivity to dynamic stimuli and impairs mechanotransduction in Pv+ DRG neurons because of substrate deformation-induced neurite stretching, but not to direct neurite indentation. In behavioural tasks, global knockout (Asic3−/−) and Pv-Cre::Asic3f/f mice produce similar deficits in grid and balance beam walking tasks. We conclude that, at least in mouse, ASIC3 is a molecular determinant contributing to dynamic mechanosensitivity in proprioceptors. Acid-sensing ion channel 3 (ASIC3) is known to play a role in nociception, but its role in low threshold neurosensory mechanotransduction is unclear. Here, the authors target ASIC3 expression in dorsal root ganglion parvalbumin positive neurons and find ASIC3 contributes to dynamic proprioception responses.
Collapse
|
36
|
Svechtarova MI, Buzzacchera I, Toebes BJ, Lauko J, Anton N, Wilson CJ. Sensor Devices Inspired by the Five Senses: A Review. ELECTROANAL 2016. [DOI: 10.1002/elan.201600047] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
| | | | - B. Jelle Toebes
- NovioSense BV; Transistorweg 5 6534 AT Nijmegen The Netherlands
| | - Jan Lauko
- NovioSense BV; Transistorweg 5 6534 AT Nijmegen The Netherlands
| | - Nicoleta Anton
- Universitatea de Medicina si Farmacie Grigore T.; Popa, Str. Universitatii nr. 16 700115 Iasi Romania
| | | |
Collapse
|
37
|
Abstract
Nonallergic rhinitis is a common disease that affects many Americans. It is characterized by nasal symptoms of congestion and rhinorrhea without evidence of allergic sensitization. The pathophysiology of the disease has not been studied extensively. In the following article, the author concentrates on summarizing the available information related to cellular inflammation and neurogenic mechanisms in patients with nonallergic rhinitis. The author also explores nasal reactivity to various stimuli in these patients.
Collapse
|
38
|
Navarro AP, Collins MA, Folker ES. The nucleus is a conserved mechanosensation and mechanoresponse organelle. Cytoskeleton (Hoboken) 2016; 73:59-67. [PMID: 26849407 DOI: 10.1002/cm.21277] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 01/22/2016] [Accepted: 01/22/2016] [Indexed: 11/05/2022]
Abstract
Cells in vivo exist in a dynamic environment where they experience variable mechanical influences. The precise mechanical environment influences cell-cell interactions, cell-extracellular matrix interactions, and in-turn, cell morphology and cell function. Therefore, the ability of each cell to constantly and rapidly alter their behavior in response to variations in their mechanical environment is essential for cell viability, development, and function. Mechanotransduction, the process by which mechanical force is translated into a biochemical signal to activate downstream cellular responses, is thus crucial to cell function during development and homeostasis. Although much research has focused on how protein complexes at the cell cortex respond to mechanical stress to initiate mechanotransduction, the nucleus has emerged as crucial to the ability of the cell to perceive and respond to changes in its mechanical environment. This additional method for mechanosensing allows for direct transmission of force through the cytoskeleton to the nucleus, which can increase the speed at which a cell changes its transcriptional profile. This review discusses recent work demonstrating the importance of the nucleus in mediating the cellular response to internal and external force, establishing the nucleus as an important mechanosensing organelle.
Collapse
Affiliation(s)
- Alexandra P Navarro
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02142
| | - Mary Ann Collins
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, 02467
| | - Eric S Folker
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, 02467
| |
Collapse
|
39
|
Ferreira LGB, Faria RX. TRPing on the pore phenomenon: what do we know about transient receptor potential ion channel-related pore dilation up to now? J Bioenerg Biomembr 2016; 48:1-12. [PMID: 26728159 DOI: 10.1007/s10863-015-9634-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 12/02/2015] [Indexed: 01/06/2023]
Abstract
Ion channels allow for rapid ion diffusion through the plasma membrane. In some conditions, ion channels induce changes in the critical plasma membrane permeability that permit 900-Da solutes to enter cells. This process is known as the pore phenomenon. Some transient receptor potential (TRP) channel subtypes have been highlighted such as the P2X7 receptor, plasma membrane VDAC-1 channel, and pannexin hemichannels. The TRP ion channels are considered multimodal transducers that respond to several kinds of stimuli. In addition, many TRP channel subtypes are involved in physiological and pathophysiological processes such as inflammation, pain, and cancer. The TRPA1, TRPM8, and TRPV1-4 subtypes have been shown to promote large-molecular-weight solute uptake, including impermeable fluorescent dyes, QX-314 hydrophilic lidocaine derivative, gabapentin, and antineoplastic drugs. This review discusses the current knowledge of TRP-associated pores and encourages scientists to study their features and explore them as novel therapeutic tools.
Collapse
Affiliation(s)
- L G B Ferreira
- Laboratory of Inflammation, Oswaldo Cruz Institute, FIOCRUZ, Av. Brasil, n° 4365, Manguinhos, CEP 21045-900, Rio de Janeiro, Brazil.
| | - R X Faria
- Laboratory of Cellular Communication, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil, n° 4365, Manguinhos, CEP 21045-900, Rio de Janeiro, Brazil
| |
Collapse
|
40
|
The contribution of TRPC1, TRPC3, TRPC5 and TRPC6 to touch and hearing. Neurosci Lett 2015; 610:36-42. [PMID: 26520460 PMCID: PMC4683096 DOI: 10.1016/j.neulet.2015.10.052] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 11/21/2022]
Abstract
The contribution of multiple TRPC channels on mechanosensory function is demonstrated. We observe a critical role for TRPC channels in touch sensation. TRPC channels contribute to cutaneous and auditory mechanosensation in a combinatorial manner, but have no direct role in cochlear mechanotransduction.
Transient receptor potential channels have diverse roles in mechanosensation. Evidence is accumulating that members of the canonical subfamily of TRP channels (TRPC) are involved in touch and hearing. Characteristic features of TRP channels include their high structural homology and their propensity to form heteromeric complexes which suggests potential functional redundancy. We previously showed that TRPC3 and TRPC6 double knockout animals have deficits in light touch and hearing whilst single knockouts were apparently normal. We have extended these studies to analyse deficits in global quadruple TRPC1, 3, 5 and 6 null mutant mice. We examined both touch and hearing in behavioural and electrophysiological assays, and provide evidence that the quadruple knockout mice have larger deficits than the TRPC3 TRPC6 double knockouts. Mechano-electrical transducer currents of cochlear outer hair cells were however normal. This suggests that TRPC1, TRPC3, TRPC5 and TRPC6 channels contribute to cutaneous and auditory mechanosensation in a combinatorial manner, but have no direct role in cochlear mechanotransduction.
Collapse
|
41
|
Vitzthum C, Clauss WG, Fronius M. Mechanosensitive activation of CFTR by increased cell volume and hydrostatic pressure but not shear stress. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:2942-51. [PMID: 26357939 DOI: 10.1016/j.bbamem.2015.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 09/03/2015] [Accepted: 09/05/2015] [Indexed: 12/20/2022]
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) is a Cl(-) channel that is essential for electrolyte and fluid homeostasis. Preliminary evidence indicates that CFTR is a mechanosensitive channel. In lung epithelia, CFTR is exposed to different mechanical forces such as shear stress (Ss) and membrane distention. The present study questioned whether Ss and/or stretch influence CFTR activity (wild type, ∆F508, G551D). Human CFTR (hCFTR) was heterologously expressed in Xenopus oocytes and the response to the mechanical stimulus and forskolin/IBMX (FI) was measured by two-electrode voltage-clamp experiments. Ss had no influence on hCFTR activity. Injection of an intracellular analogous solution to increase cell volume alone did not affect hCFTR activity. However, hCFTR activity was augmented by injection after pre-stimulation with FI. The response to injection was similar in channels carrying the common mutations ∆F508 and G551D compared to wild type hCFTR. Stretch-induced CFTR activation was further assessed in Ussing chamber measurements using Xenopus lung preparations. Under control conditions increased hydrostatic pressure (HP) decreased the measured ion current including activation of a Cl(-) secretion that was unmasked by the CFTR inhibitor GlyH-101. These data demonstrate activation of CFTR in vitro and in a native pulmonary epithelium in response to mechanical stress. Mechanosensitive regulation of CFTR is highly relevant for pulmonary physiology that relies on ion transport processes facilitated by pulmonary epithelial cells.
Collapse
Affiliation(s)
- Constanze Vitzthum
- Institute of Animal Physiology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Wolfgang G Clauss
- Institute of Animal Physiology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Martin Fronius
- Department of Physiology, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
42
|
Forcing open TRP channels: Mechanical gating as a unifying activation mechanism. Biochem Biophys Res Commun 2015; 460:22-5. [PMID: 25998730 DOI: 10.1016/j.bbrc.2015.02.067] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 02/15/2015] [Indexed: 11/21/2022]
Abstract
Transient receptor potential (TRP) proteins are cation channels that comprise a superfamily of molecular sensors that enable animals to detect a wide variety of environmental stimuli. This versatility enables vertebrate and invertebrate TRP channels to function in a diversity of senses, ranging from vision to taste, smell, touch, hearing, proprioception and thermosensation. Moreover, many individual TRP channels are activated through a surprising range of sensory stimuli. The multitasking nature of TRP channels raises the question as to whether seemingly disparate activators gate TRPs through common strategies. In this regard, a recent major advance is the discovery that a phospholipase C (PLC)-dependent signaling cascade activates the TRP channels in Drosophila photoreceptor cells through generation of force in the lipid-bilayer. The premise of this review is that mechanical force is a unifying, common strategy for gating TRP channels. In addition to several TRP channels that function in mechanosensation and are gated by force applied to the cells, changes in temperature or alterations in the concentration of lipophilic second messengers through stimulation of signaling cascades, cause architectural modifications of the cell membrane, which in turn activate TRP channels through mechanical force. Consequently, TRPs are capable of functioning as stretch-activated channels, even in cases in which the stimuli that initiate the signaling cascades are not mechanical. We propose that most TRPs are actually mechanosensitive channels (MSCs), which undergo conformational changes in response to tension imposed on the lipid bilayer, resulting in channel gating.
Collapse
|
43
|
A novel PKD2L1 C-terminal domain critical for trimerization and channel function. Sci Rep 2015; 5:9460. [PMID: 25820328 PMCID: PMC4377555 DOI: 10.1038/srep09460] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 03/06/2015] [Indexed: 11/08/2022] Open
Abstract
As a transient receptor potential (TRP) superfamily member, polycystic kidney disease 2-like-1 (PKD2L1) is also called TRPP3 and has similar membrane topology as voltage-gated cation channels. PKD2L1 is involved in hedgehog signaling, intestinal development, and sour tasting. PKD2L1 and PKD1L3 form heterotetramers with 3:1 stoichiometry. C-terminal coiled-coil-2 (CC2) domain (G699-W743) of PKD2L1 was reported to be important for its trimerization but independent studies showed that CC2 does not affect PKD2L1 channel function. It thus remains unclear how PKD2L1 proteins oligomerize into a functional channel. By SDS-PAGE, blue native PAGE and mutagenesis we here identified a novel C-terminal domain called C1 (K575-T622) involved in stronger homotrimerization than the non-overlapping CC2, and found that the PKD2L1 N-terminus is critical for dimerization. By electrophysiology and Xenopus oocyte expression, we found that C1, but not CC2, is critical for PKD2L1 channel function. Our co-immunoprecipitation and dynamic light scattering experiments further supported involvement of C1 in trimerization. Further, C1 acted as a blocking peptide that inhibits PKD2L1 trimerization as well as PKD2L1 and PKD2L1/PKD1L3 channel function. Thus, our study identified C1 as the first PKD2L1 domain essential for both PKD2L1 trimerization and channel function, and suggest that PKD2L1 and PKD2L1/PKD1L3 channels share the PKD2L1 trimerization process.
Collapse
|
44
|
Enyedi B, Niethammer P. Mechanisms of epithelial wound detection. Trends Cell Biol 2015; 25:398-407. [PMID: 25813429 DOI: 10.1016/j.tcb.2015.02.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 12/22/2022]
Abstract
Efficient wound healing requires the coordinated responses of various cell types within an injured tissue. To react to the presence of a wound, cells have to first detect it. Judging from their initial biochemical and morphological responses, many cells including leukocytes, epithelial cells, and endothelial cells detect wounds from over hundreds of micrometers within seconds-to-minutes. Wound detection involves the conversion of an injury-induced homeostatic perturbation, such as cell lysis, an unconstrained epithelial edge, or permeability barrier breakdown, into a chemical or physical signal. The signal is spatially propagated through the tissue to synchronize protective responses of cells near the wound site and at a distance. This review summarizes the triggers and mechanisms of wound detection in animals.
Collapse
Affiliation(s)
- Balázs Enyedi
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Philipp Niethammer
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
45
|
Are Aquaporins the Missing Transmembrane Osmosensors? J Membr Biol 2015; 248:753-65. [PMID: 25791748 DOI: 10.1007/s00232-015-9790-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 03/12/2015] [Indexed: 01/08/2023]
Abstract
Regulation of cell volume is central to homeostasis. It is assumed to begin with the detection of a change in water potential across the bounding membrane, but it is not clear how this is accomplished. While examples of general osmoreceptors (which sense osmotic pressure in one phase) and stretch-activated ion channels (which require swelling of a cell or organelle) are known, effective volume regulation requires true transmembrane osmosensors (TMOs) which directly detect a water potential difference spanning a membrane. At present, no TMO molecule has been unambiguously identified, and clear evidence for mammalian TMOs is notably lacking. In this paper, we set out a theory of TMOs which requires a water channel spanning the membrane that excludes the major osmotic solutes, responds directly without the need for any other process such as swelling, and signals to other molecules associated with the magnitude of changing osmotic differences. The most likely molecules that are fit for this purpose and which are also ubiquitous in eukaryotic cells are aquaporins (AQPs). We review experimental evidence from several systems which indicates that AQPs are essential elements in regulation and may be functioning as TMOs; i.e. the first step in an osmosensing sequence that signals osmotic imbalance in a cell or organelle. We extend this concept to several systems of current interest in which the cellular involvement of AQPs as simple water channels is puzzling or counter-intuitive. We suggest that, apart from regulatory volume changes in cells, AQPs may also be acting as TMOs in red cells, secretory granules and microorganisms.
Collapse
|
46
|
Schlosser G. Vertebrate cranial placodes as evolutionary innovations--the ancestor's tale. Curr Top Dev Biol 2015; 111:235-300. [PMID: 25662263 DOI: 10.1016/bs.ctdb.2014.11.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Evolutionary innovations often arise by tinkering with preexisting components building new regulatory networks by the rewiring of old parts. The cranial placodes of vertebrates, ectodermal thickenings that give rise to many of the cranial sense organs (ear, nose, lateral line) and ganglia, originated as such novel structures, when vertebrate ancestors elaborated their head in support of a more active and exploratory life style. This review addresses the question of how cranial placodes evolved by tinkering with ectodermal patterning mechanisms and sensory and neurosecretory cell types that have their own evolutionary history. With phylogenetic relationships among the major branches of metazoans now relatively well established, a comparative approach is used to infer, which structures evolved in which lineages and allows us to trace the origin of placodes and their components back from ancestor to ancestor. Some of the core networks of ectodermal patterning and sensory and neurosecretory differentiation were already established in the common ancestor of cnidarians and bilaterians and were greatly elaborated in the bilaterian ancestor (with BMP- and Wnt-dependent patterning of dorsoventral and anteroposterior ectoderm and multiple neurosecretory and sensory cell types). Rostral and caudal protoplacodal domains, giving rise to some neurosecretory and sensory cells, were then established in the ectoderm of the chordate and tunicate-vertebrate ancestor, respectively. However, proper cranial placodes as clusters of proliferating progenitors producing high-density arrays of neurosecretory and sensory cells only evolved and diversified in the ancestors of vertebrates.
Collapse
Affiliation(s)
- Gerhard Schlosser
- School of Natural Sciences & Regenerative Medicine Institute (REMEDI), National University of Ireland, Galway, Ireland.
| |
Collapse
|
47
|
G protein-coupled odorant receptors underlie mechanosensitivity in mammalian olfactory sensory neurons. Proc Natl Acad Sci U S A 2014; 112:590-5. [PMID: 25550517 DOI: 10.1073/pnas.1418515112] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mechanosensitive cells are essential for organisms to sense the external and internal environments, and a variety of molecules have been implicated as mechanical sensors. Here we report that odorant receptors (ORs), a large family of G protein-coupled receptors, underlie the responses to both chemical and mechanical stimuli in mouse olfactory sensory neurons (OSNs). Genetic ablation of key signaling proteins in odor transduction or disruption of OR-G protein coupling eliminates mechanical responses. Curiously, OSNs expressing different OR types display significantly different responses to mechanical stimuli. Genetic swap of putatively mechanosensitive ORs abolishes or reduces mechanical responses of OSNs. Furthermore, ectopic expression of an OR restores mechanosensitivity in loss-of-function OSNs. Lastly, heterologous expression of an OR confers mechanosensitivity to its host cells. These results indicate that certain ORs are both necessary and sufficient to cause mechanical responses, revealing a previously unidentified mechanism for mechanotransduction.
Collapse
|
48
|
Hamilton ES, Schlegel AM, Haswell ES. United in diversity: mechanosensitive ion channels in plants. ANNUAL REVIEW OF PLANT BIOLOGY 2014; 66:113-37. [PMID: 25494462 PMCID: PMC4470482 DOI: 10.1146/annurev-arplant-043014-114700] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Mechanosensitive (MS) ion channels are a common mechanism for perceiving and responding to mechanical force. This class of mechanoreceptors is capable of transducing membrane tension directly into ion flux. In plant systems, MS ion channels have been proposed to play a wide array of roles, from the perception of touch and gravity to the osmotic homeostasis of intracellular organelles. Three families of plant MS ion channels have been identified: the MscS-like (MSL), Mid1-complementing activity (MCA), and two-pore potassium (TPK) families. Channels from these families vary widely in structure and function, localize to multiple cellular compartments, and conduct chloride, calcium, and/or potassium ions. However, they are still likely to represent only a fraction of the MS ion channel diversity in plant systems.
Collapse
Affiliation(s)
- Eric S. Hamilton
- Department of Biology, Washington University in Saint Louis, Saint Louis, Missouri 63130
| | - Angela M. Schlegel
- Department of Biology, Washington University in Saint Louis, Saint Louis, Missouri 63130
| | - Elizabeth S. Haswell
- Department of Biology, Washington University in Saint Louis, Saint Louis, Missouri 63130
| |
Collapse
|
49
|
Hao J, Bonnet C, Amsalem M, Ruel J, Delmas P. Transduction and encoding sensory information by skin mechanoreceptors. Pflugers Arch 2014; 467:109-19. [PMID: 25416542 DOI: 10.1007/s00424-014-1651-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 11/06/2014] [Accepted: 11/07/2014] [Indexed: 12/25/2022]
Abstract
Physical contact with the external world occurs through specialized neural structures called mechanoreceptors. Cutaneous mechanoreceptors provide information to the central nervous system (CNS) about touch, pressure, vibration, and skin stretch. The physiological function of these mechanoreceptors is to convert physical forces into neuronal signals. Key questions concern the molecular identity of the mechanoelectric transducer channels and the mechanisms by which the physical parameters of the mechanical stimulus are encoded into patterns of action potentials (APs). Compelling data indicate that the biophysical traits of mechanosensitive channels combined with the collection of voltage-gated channels are essential to describe the nature of the stimulus. Recent research also points to a critical role of the auxiliary cell-nerve ending communication in encoding stimulus properties. This review describes the characteristics of ion channels responsible for translating mechanical stimuli into the neural codes that underlie touch perception and pain.
Collapse
Affiliation(s)
- Jizhe Hao
- Aix-Marseille-Université, CNRS, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, UMR 7286, CS80011, Bd Pierre Dramard, 13344, Marseille Cedex 15, France,
| | | | | | | | | |
Collapse
|
50
|
Pain related channels are differentially expressed in neuronal and non-neuronal cells of glabrous skin of fabry knockout male mice. PLoS One 2014; 9:e108641. [PMID: 25337704 PMCID: PMC4206276 DOI: 10.1371/journal.pone.0108641] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 09/03/2014] [Indexed: 11/20/2022] Open
Abstract
Fabry disease (FD) is one of the X-linked lysosomal storage disorders caused by deficient functioning of the alpha-galactosidase A (α-GalA) enzyme. The α-GalA deficiency leads to multi-systemic clinical manifestations caused by the preferential accumulation of globotriaosylceramide in the endothelium and vascular smooth muscles. A hallmark symptom of FD patients is peripheral pain that appears in the early stage of the disease. Pain in FD patients is a peripheral small-fiber idiopathic neuropathy, with intra-epidermal fiber density and integrity being used for diagnosing FD in humans. However, the molecular correlates underlying pain sensation in FD remain elusive. Here, we have employed the α-GalA gene KO mouse as a model of FD in rodents to investigate molecular changes in their peripheral nervous system that may account for their algesic symptoms. The α-GalA null mice display neuropathic pain as evidenced by thermal hyperalgesia and mechanical allodynia, with histological analyses showing alterations in cutaneous innervation. Additionally, KO mice showed a decreased and scattered pattern of neuronal terminations consistent with the reduction in neuronal terminations in skin biopsies of patients with small fiber neuropathies. At the molecular level KO animals showed an increase in the expression of TRPV1 and Nav1.8, and a decrease in the expression of TRPM8. Notably, these alterations are observed in young animals. Taken together, our findings imply that the α-GalA KO mouse is a good model in which to study the peripheral small fiber neuropathy exhibited by FD patients, and provides molecular evidence for a hyperexcitability of small nociceptors in FD.
Collapse
|