1
|
Réthoré L, Guihot A, Grimaud L, Proux C, Barré B, Guillonneau F, Guette C, Boissard A, Henry C, Cayon J, Perrot R, Henrion D, Legros C, Legendre C. A Novel Function of Na V Channel β3 Subunit in Endothelial Cell Alignment Through Autophagy Modulation. FASEB J 2025; 39:e70663. [PMID: 40445729 PMCID: PMC12124425 DOI: 10.1096/fj.202401558rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 04/25/2025] [Accepted: 05/14/2025] [Indexed: 06/02/2025]
Abstract
Endothelial cells (EC) play a pivotal role in vascular homeostasis. By sensing shear stress generated by blood flow, EC endorse vasculoprotection through mechanotransduction signaling pathways. Various ion channels are involved in mechanosignaling, and here, we investigated the endothelial voltage-gated Na+ channels (NaV channels), since their mechanosensitivity has been previously demonstrated in cardiomyocytes. First, we showed that EC from aorta (TeloHAEC) behave as EC from umbilical vein (HUVEC) under laminar shear stress (LSS). For both EC models, cell alignment and elongation occurred with the activation of the KLF2/KLF4 atheroprotective signaling pathways. We found that LSS decreased the expression of SCN5A, encoding NaV1.5, while LSS increased that of SCN3B, encoding NaVβ3. We demonstrated that the KLF4 transcription factor is involved in SCN3B expression under both static and LSS conditions. Interestingly, SCN3B silencing impaired EC alignment induced by LSS. The characterization of NaVβ3 interactome by coimmunoprecipitation and proteomic analysis revealed that mTOR, implicated in autophagy, binds to NaVβ3. This result was evidenced by the colocalization between NaVβ3 and mTOR inside cells. Moreover, we showed that SCN3B silencing led to the decrease in LC3B expression and the number of LC3B positive autophagosomes. Furthermore, we showed that NaVβ3 is retained within the cell and colocalized with LAMP1 and LC3B. Finally, we found that resveratrol, a stimulating-autophagy and vasculoprotective molecule, induced KLF4 together with NaVβ3 expression. Altogether, our findings highlight a novel role of NaVβ3 in endothelial function and cell alignment as an actor in shear stress vasculoprotective intracellular pathway through autophagy modulation.
Collapse
Affiliation(s)
- Léa Réthoré
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, Univ AngersAngersFrance
| | - Anne‐Laure Guihot
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, Univ AngersAngersFrance
| | - Linda Grimaud
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, Univ AngersAngersFrance
| | - Coralyne Proux
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, Univ AngersAngersFrance
| | - Benjamin Barré
- Univ AngersAngersFrance
- Institut de Cancérologie de l'Ouest (ICO)AngersFrance
- Prot'ICO (ICO Proteomic Core Facility)AngersFrance
| | - François Guillonneau
- Institut de Cancérologie de l'Ouest (ICO)AngersFrance
- Prot'ICO (ICO Proteomic Core Facility)AngersFrance
- INSERM, CNRS, CRCI2NA, Nantes Université, Univ AngersAngersFrance
| | - Catherine Guette
- Institut de Cancérologie de l'Ouest (ICO)AngersFrance
- Prot'ICO (ICO Proteomic Core Facility)AngersFrance
- INSERM, CNRS, CRCI2NA, Nantes Université, Univ AngersAngersFrance
| | - Alice Boissard
- Institut de Cancérologie de l'Ouest (ICO)AngersFrance
- Prot'ICO (ICO Proteomic Core Facility)AngersFrance
| | - Cécile Henry
- Institut de Cancérologie de l'Ouest (ICO)AngersFrance
- Prot'ICO (ICO Proteomic Core Facility)AngersFrance
| | - Jérôme Cayon
- SFR ICAT, PACeM (Plateforme d'Analyse Cellulaire et Moléculaire), Univ AngersAngersFrance
| | - Rodolphe Perrot
- SFR ICAT, SCIAM (Service Commun d'Imageries et d'Analyses Microscopiques), Univ AngersAngersFrance
| | - Daniel Henrion
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, Univ AngersAngersFrance
| | - Christian Legros
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, Univ AngersAngersFrance
| | - Claire Legendre
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, Univ AngersAngersFrance
| |
Collapse
|
2
|
Chen Z, Sang L, Qixi Z, Li X, Liu Y, Bai Z. Ultrasound-responsive nanoparticles for imaging and therapy of brain tumors. Mater Today Bio 2025; 32:101661. [PMID: 40206140 PMCID: PMC11979416 DOI: 10.1016/j.mtbio.2025.101661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/26/2025] [Accepted: 03/13/2025] [Indexed: 04/11/2025] Open
Abstract
Central nervous system (CNS) cancers, particularly glioblastoma (GBM), are associated with high mortality and disability rates. Despite aggressive surgical resection, radiotherapy, and chemotherapy, patient survival remains poor. The blood-brain barrier (BBB) significantly impedes therapeutic efficacy, making BBB penetration a critical focus of research. Focused ultrasound (FUS) combined with microbubbles (MBs) can transiently open the BBB through mechanisms such as cavitation, modulation of tight junction protein expression, and enhanced vesicular transport in endothelial cells. This review highlights precision delivery and personalized treatment strategies under ultrasound visualization, including precise control of ultrasound parameters and modulation of the immune microenvironment. We discuss the applications of ultrasound-responsive nanoparticles in brain tumor therapy, including enhanced radiotherapy, gene delivery, immunotherapy, and sonodynamic therapy (SDT), with a particular emphasis on piezoelectric catalytic immunotherapy. Finally, we provide insights into the clinical translation potential of ultrasound-responsive nanoparticles for personalized and precision treatment of brain tumors.
Collapse
Affiliation(s)
- Zhiguang Chen
- Department of Ultrasound, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Liang Sang
- Department of Ultrasound, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Zhai Qixi
- Department of Ultrasound, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | | | | | | |
Collapse
|
3
|
Vasylyev DV, Liu CJ, Waxman SG. Sodium channels in non-excitable cells: powerful actions and therapeutic targets beyond Hodgkin and Huxley. Trends Cell Biol 2025; 35:381-398. [PMID: 39743470 DOI: 10.1016/j.tcb.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Voltage-gated sodium channels (VGSCs) are best known for their role in the generation and propagation of action potentials in neurons, muscle cells, and cardiac myocytes, which have traditionally been labeled as 'excitable'. However, emerging evidence challenges this traditional perspective. It is now clear that VGSCs are also expressed in a broad spectrum of cells outside the neuromuscular realm, where they regulate diverse cellular functions. In this review, we summarize current knowledge on the expression, regulation, and function of VGSCs in non-neuromuscular cells, highlighting their contributions to physiological processes and pathological conditions. Dynamic expression patterns of VGSCs in different cell types, involvement of VGSCs in cellular functions, such as phagocytosis, motility, and cytokine release, and their potential as therapeutic targets for diseases that include inflammatory disorders, osteoarthritis (OA), and cancer, are discussed. This new understanding of VGSCs and their effects on cells outside the neuromuscular realm opens new avenues for research and therapeutic interventions.
Collapse
Affiliation(s)
- Dmytro V Vasylyev
- Department of Neurology and Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Chuan-Ju Liu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA.
| |
Collapse
|
4
|
Zolfaghari H, Andiapen M, Baumbach A, Mathur A, Kerswell RR. Wall shear stress and pressure patterns in aortic stenosis patients with and without aortic dilation captured by high-performance image-based computational fluid dynamics. PLoS Comput Biol 2023; 19:e1011479. [PMID: 37851683 PMCID: PMC10635572 DOI: 10.1371/journal.pcbi.1011479] [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: 12/13/2022] [Revised: 11/09/2023] [Accepted: 08/24/2023] [Indexed: 10/20/2023] Open
Abstract
Spatial patterns of elevated wall shear stress and pressure due to blood flow past aortic stenosis (AS) are studied using GPU-accelerated patient-specific computational fluid dynamics. Three cases of moderate to severe AS, one with a dilated ascending aorta and two within the normal range (root diameter less than 4cm) are simulated for physiological waveforms obtained from echocardiography. The computational framework is built based on sharp-interface Immersed Boundary Method, where aortic geometries segmented from CT angiograms are integrated into a high-order incompressible Navier-Stokes solver. The key question addressed here is, given the presence of turbulence due to AS which increases wall shear stress (WSS) levels, why some AS patients undergo much less aortic dilation. Recent case studies of AS have linked the existence of an elevated WSS hotspot (due to impingement of AS on the aortic wall) to the dilation process. Herein we further investigate the WSS distribution for cases with and without dilation to understand the possible hemodynamics which may impact the dilation process. We show that the spatial distribution of elevated WSS is significantly more focused for the case with dilation than those without dilation. We further show that this focal area accommodates a persistent pocket of high pressure, which may have contributed to the dilation process through an increased wall-normal forcing. The cases without dilation, on the contrary, showed a rather oscillatory pressure behaviour, with no persistent pressure "buildup" effect. We further argue that a more proximal branching of the aortic arch could explain the lack of a focal area of elevated WSS and pressure, because it interferes with the impingement process due to fluid suction effects. These phenomena are further illustrated using an idealized aortic geometry. We finally show that a restored inflow eliminates the focal area of elevated WSS and pressure zone from the ascending aorta.
Collapse
Affiliation(s)
- Hadi Zolfaghari
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, United Kingdom
| | - Mervyn Andiapen
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
| | - Andreas Baumbach
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Anthony Mathur
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom
| | - Rich R. Kerswell
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
5
|
Prideaux M, Smargiassi A, Peng G, Brotto M, Robling AG, Bonewald LF. L-BAIBA Synergizes with Sub-Optimal Mechanical Loading to Promote New Bone Formation. JBMR Plus 2023; 7:e10746. [PMID: 37283651 PMCID: PMC10241089 DOI: 10.1002/jbm4.10746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/21/2023] [Accepted: 03/28/2023] [Indexed: 06/08/2023] Open
Abstract
The L-enantiomer of β-aminoisobutyric acid (BAIBA) is secreted by contracted muscle in mice, and exercise increases serum levels in humans. In mice, L-BAIBA reduces bone loss with unloading, but whether it can have a positive effect with loading is unknown. Since synergism can be more easily observed with sub-optimal amounts of factors/stimulation, we sought to determine whether L-BAIBA could potentiate the effects of sub-optimal loading to enhance bone formation. L-BAIBA was provided in drinking water to C57Bl/6 male mice subjected to either 7 N or 8.25 N of sub-optimal unilateral tibial loading for 2 weeks. The combination of 8.25 N and L-BAIBA significantly increased the periosteal mineral apposition rate and bone formation rate compared to loading alone or BAIBA alone. Though L-BAIBA alone had no effect on bone formation, grip strength was increased, suggesting a positive effect on muscle function. Gene expression analysis of the osteocyte-enriched bone showed that the combination of L-BAIBA and 8.25 N induced the expression of loading-responsive genes such as Wnt1, Wnt10b, and the TGFb and BMP signaling pathways. One dramatic change was the downregulation of histone genes in response to sub-optimal loading and/or L-BAIBA. To determine early gene expression, the osteocyte fraction was harvested within 24 hours of loading. A dramatic effect was observed with L-BAIBA and 8.25 N loading as genes were enriched for pathways regulating the extracellular matrix (Chad, Acan, Col9a2), ion channel activity (Scn4b, Scn7a, Cacna1i), and lipid metabolism (Plin1, Plin4, Cidec). Few changes in gene expression were observed with sub-optimal loading or L-BAIBA alone after 24 hours. These results suggest that these signaling pathways are responsible for the synergistic effects between L-BAIBA and sub-optimal loading. Showing that a small muscle factor can enhance the effects of sub-optimal loading of bone may be of relevance for individuals unable to benefit from optimal exercise. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Matt Prideaux
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of MedicineIndiana UniversityIndianapolisINUSA
| | - Alberto Smargiassi
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of MedicineIndiana UniversityIndianapolisINUSA
| | - Gang Peng
- Indiana Center for Musculoskeletal Health, Department of Medicine and Molecular Genetics, School of MedicineIndiana UniversityIndianapolisINUSA
| | - Marco Brotto
- Bone‐Muscle Research Center, College of Nursing and Health InnovationUniversity of Texas‐ArlingtonArlingtonTXUSA
| | - Alexander G Robling
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of MedicineIndiana UniversityIndianapolisINUSA
| | - Lynda F Bonewald
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of MedicineIndiana UniversityIndianapolisINUSA
| |
Collapse
|
6
|
Park J, Proux C, Ehanno W, Réthoré L, Vessières E, Bourreau J, Favre J, Kauffenstein G, Mattei C, Tricoire-Leignel H, Henrion D, Legendre C, Legros C. Tetrodotoxin Decreases the Contractility of Mesenteric Arteries, Revealing the Contribution of Voltage-Gated Na + Channels in Vascular Tone Regulation. Mar Drugs 2023; 21:md21030196. [PMID: 36976245 PMCID: PMC10059581 DOI: 10.3390/md21030196] [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: 02/21/2023] [Revised: 03/11/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
Tetrodotoxin (TTX) poisoning through the consumption of contaminated fish leads to lethal symptoms, including severe hypotension. This TTX-induced hypotension is likely due to the downfall of peripheral arterial resistance through direct or indirect effects on adrenergic signaling. TTX is a high-affinity blocker of voltage-gated Na+ (NaV) channels. In arteries, NaV channels are expressed in sympathetic nerve endings, both in the intima and media. In this present work, we aimed to decipher the role of NaV channels in vascular tone using TTX. We first characterized the expression of NaV channels in the aorta, a model of conduction arteries, and in mesenteric arteries (MA), a model of resistance arteries, in C57Bl/6J mice, by Western blot, immunochemistry, and absolute RT-qPCR. Our data showed that these channels are expressed in both endothelium and media of aorta and MA, in which scn2a and scn1b were the most abundant transcripts, suggesting that murine vascular NaV channels consist of NaV1.2 channel subtype with NaVβ1 auxiliary subunit. Using myography, we showed that TTX (1 µM) induced complete vasorelaxation in MA in the presence of veratridine and cocktails of antagonists (prazosin and atropine with or without suramin) that suppressed the effects of neurotransmitter release. In addition, TTX (1 µM) strongly potentiated the flow-mediated dilation response of isolated MA. Altogether, our data showed that TTX blocks NaV channels in resistance arteries and consecutively decreases vascular tone. This could explain the drop in total peripheral resistance observed during mammal tetrodotoxications.
Collapse
Affiliation(s)
- Joohee Park
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, University Angers, 49000 Angers, France
| | - Coralyne Proux
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, University Angers, 49000 Angers, France
| | - William Ehanno
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, University Angers, 49000 Angers, France
| | - Léa Réthoré
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, University Angers, 49000 Angers, France
| | - Emilie Vessières
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, University Angers, 49000 Angers, France
| | - Jennifer Bourreau
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, University Angers, 49000 Angers, France
| | - Julie Favre
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, University Angers, 49000 Angers, France
- UMR INSERM 1121, CRBS, Strasbourg University, 67000 Strasbourg, France
| | - Gilles Kauffenstein
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, University Angers, 49000 Angers, France
- UMR INSERM 1260, CRBS, Strasbourg University, 67084 Strasbourg, France
| | - César Mattei
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, University Angers, 49000 Angers, France
| | | | - Daniel Henrion
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, University Angers, 49000 Angers, France
| | - Claire Legendre
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, University Angers, 49000 Angers, France
| | - Christian Legros
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, University Angers, 49000 Angers, France
| |
Collapse
|
7
|
Feher A, Pócsi M, Papp F, Szanto TG, Csoti A, Fejes Z, Nagy B, Nemes B, Varga Z. Functional Voltage-Gated Sodium Channels Are Present in the Human B Cell Membrane. Cells 2022; 11:1225. [PMID: 35406789 PMCID: PMC8998058 DOI: 10.3390/cells11071225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/31/2022] [Accepted: 04/03/2022] [Indexed: 02/04/2023] Open
Abstract
B cells express various ion channels, but the presence of voltage-gated sodium (NaV) channels has not been confirmed in the plasma membrane yet. In this study, we have identified several NaV channels, which are expressed in the human B cell membrane, by electrophysiological and molecular biology methods. The sensitivity of the detected sodium current to tetrodotoxin was between the values published for TTX-sensitive and TTX-insensitive channels, which suggests the co-existence of multiple NaV1 subtypes in the B cell membrane. This was confirmed by RT-qPCR results, which showed high expression of TTX-sensitive channels along with the lower expression of TTX-insensitive NaV1 channels. The biophysical characteristics of the currents also supported the expression of multiple NaV channels. In addition, we investigated the potential functional role of NaV channels by membrane potential measurements. Removal of Na+ from the extracellular solution caused a reversible hyperpolarization, supporting the role of NaV channels in shaping and maintaining the resting membrane potential. As this study was mainly limited to electrophysiological properties, we cannot exclude the possible non-canonical functions of these channels. This work concludes that the presence of voltage-gated sodium channels in the plasma membrane of human B cells should be recognized and accounted for in the future.
Collapse
Affiliation(s)
- Adam Feher
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (A.F.); (F.P.); (T.G.S.); (A.C.)
| | - Marianna Pócsi
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.P.); (Z.F.); (B.N.J.)
| | - Ferenc Papp
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (A.F.); (F.P.); (T.G.S.); (A.C.)
| | - Tibor G. Szanto
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (A.F.); (F.P.); (T.G.S.); (A.C.)
| | - Agota Csoti
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (A.F.); (F.P.); (T.G.S.); (A.C.)
| | - Zsolt Fejes
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.P.); (Z.F.); (B.N.J.)
| | - Béla Nagy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.P.); (Z.F.); (B.N.J.)
| | - Balázs Nemes
- Department of Organ Transplantation, Faculty of Medicine, Institute of Surgery, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Zoltan Varga
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (A.F.); (F.P.); (T.G.S.); (A.C.)
| |
Collapse
|
8
|
Lillo MA, Gaete PS, Puebla M, Burboa PC, Poblete I, Figueroa XF. Novel Pannexin-1-Coupled Signaling Cascade Involved in the Control of Endothelial Cell Function and NO-Dependent Relaxation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2678134. [PMID: 33688389 PMCID: PMC7914086 DOI: 10.1155/2021/2678134] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 01/03/2021] [Accepted: 01/13/2021] [Indexed: 12/11/2022]
Abstract
Deletion of pannexin-1 (Panx-1) leads not only to a reduction in endothelium-derived hyperpolarization but also to an increase in NO-mediated vasodilation. Therefore, we evaluated the participation of Panx-1-formed channels in the control of membrane potential and [Ca2+]i of endothelial cells. Changes in NO-mediated vasodilation, membrane potential, superoxide anion (O2 ·-) formation, and endothelial cell [Ca2+]i were analyzed in rat isolated mesenteric arterial beds and primary cultures of mesenteric endothelial cells. Inhibition of Panx-1 channels with probenecid (1 mM) or the Panx-1 blocking peptide 10Panx (60 μM) evoked an increase in the ACh (100 nM)-induced vasodilation of KCl-contracted mesenteries and in the phosphorylation level of endothelial NO synthase (eNOS) at serine 1177 (P-eNOSS1177) and Akt at serine 473 (P-AktS473). In addition, probenecid or 10Panx application activated a rapid, tetrodotoxin (TTX, 300 nM)-sensitive, membrane potential depolarization and [Ca2+]i increase in endothelial cells. Interestingly, the endothelial cell depolarization was converted into a transient spike after removing Ca2+ ions from the buffer solution and in the presence of 100 μM mibefradil or 10 μM Ni2+. As expected, Ni2+ also abolished the increment in [Ca2+]i. Expression of Nav1.2, Nav1.6, and Cav3.2 isoforms of voltage-dependent Na+ and Ca2+ channels was confirmed by immunocytochemistry. Furthermore, the Panx-1 channel blockade was associated with an increase in O2 ·- production. Treatment with 10 μM TEMPOL or 100 μM apocynin prevented the increase in O2 ·- formation, ACh-induced vasodilation, P-eNOSS1177, and P-AktS473 observed in response to Panx-1 inhibition. These findings indicate that the Panx-1 channel blockade triggers a novel complex signaling pathway initiated by the sequential activation of TTX-sensitive Nav channels and Cav3.2 channels, leading to an increase in NO-mediated vasodilation through a NADPH oxidase-dependent P-eNOSS1177, which suggests that Panx-1 may be involved in the endothelium-dependent control of arterial blood pressure.
Collapse
Affiliation(s)
- Mauricio A. Lillo
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330025, Chile
| | - Pablo S. Gaete
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330025, Chile
| | - Mariela Puebla
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina-Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Pía C. Burboa
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330025, Chile
| | - Inés Poblete
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330025, Chile
| | - Xavier F. Figueroa
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330025, Chile
| |
Collapse
|
9
|
Lambert M, Capuano V, Olschewski A, Sabourin J, Nagaraj C, Girerd B, Weatherald J, Humbert M, Antigny F. Ion Channels in Pulmonary Hypertension: A Therapeutic Interest? Int J Mol Sci 2018; 19:ijms19103162. [PMID: 30322215 PMCID: PMC6214085 DOI: 10.3390/ijms19103162] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 12/25/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a multifactorial and severe disease without curative therapies. PAH pathobiology involves altered pulmonary arterial tone, endothelial dysfunction, distal pulmonary vessel remodeling, and inflammation, which could all depend on ion channel activities (K⁺, Ca2+, Na⁺ and Cl-). This review focuses on ion channels in the pulmonary vasculature and discusses their pathophysiological contribution to PAH as well as their therapeutic potential in PAH.
Collapse
Affiliation(s)
- Mélanie Lambert
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Véronique Capuano
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Stiftingtalstrasse 24, Graz 8010, Austria.
- Department of Physiology, Medical University Graz, Neue Stiftingtalstraße 6, Graz 8010, Austria.
| | - Jessica Sabourin
- Signalisation et Physiopathologie Cardiovasculaire, UMRS 1180, Univ. Paris-Sud, INSERM, Université Paris-Saclay, 92296 Châtenay-Malabry, France.
| | - Chandran Nagaraj
- Ludwig Boltzmann Institute for Lung Vascular Research, Stiftingtalstrasse 24, Graz 8010, Austria.
| | - Barbara Girerd
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Jason Weatherald
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
- Division of Respirology, Department of Medicine, University of Calgary, Calgary, AB T1Y 6J4, Canada.
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB T1Y 6J4, Canada.
| | - Marc Humbert
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Fabrice Antigny
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| |
Collapse
|
10
|
The invasiveness of human cervical cancer associated to the function of Na V1.6 channels is mediated by MMP-2 activity. Sci Rep 2018; 8:12995. [PMID: 30158710 PMCID: PMC6115389 DOI: 10.1038/s41598-018-31364-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 08/16/2018] [Indexed: 12/17/2022] Open
Abstract
Voltage-gated sodium (NaV) channels have been related with cell migration and invasiveness in human cancers. We previously reported the contribution of NaV1.6 channels activity with the invasion capacity of cervical cancer (CeCa) positive to Human Papilloma Virus type 16 (HPV16), which accounts for 50% of all CeCa cases. Here, we show that NaV1.6 gene (SCN8A) overexpression is a general characteristic of CeCa, regardless of the HPV type. In contrast, no differences were observed in NaV1.6 channel expression between samples of non-cancerous and cervical intraepithelial neoplasia. Additionally, we found that CeCa cell lines, C33A, SiHa, CaSki and HeLa, express mainly the splice variant of SCN8A that lacks exon 18, shown to encode for an intracellularly localized NaV1.6 channel, whereas the full-length adult form was present in CeCa biopsies. Correlatively, patch-clamp experiments showed no evidence of whole-cell sodium currents (INa) in CeCa cell lines. Heterologous expression of full-length NaV1.6 isoform in C33A cells produced INa, which were sufficient to significantly increase invasion capacity and matrix metalloproteinase type 2 (MMP-2) activity. These data suggest that upregulation of NaV1.6 channel expression occurs when cervical epithelium have been transformed into cancer cells, and that NaV1.6-mediated invasiveness of CeCa cells involves MMP-2 activity. Thus, our findings support the notion about using NaV channels as therapeutic targets against cancer metastasis.
Collapse
|
11
|
Lillo MA, Gaete PS, Puebla M, Ardiles NM, Poblete I, Becerra A, Simon F, Figueroa XF. Critical contribution of Na +-Ca 2+ exchanger to the Ca 2+-mediated vasodilation activated in endothelial cells of resistance arteries. FASEB J 2018; 32:2137-2147. [PMID: 29217667 DOI: 10.1096/fj.201700365rr] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Na+-Ca2+ exchanger (NCX) contributes to control the intracellular free Ca2+ concentration ([Ca2+]i), but the functional activation of NCX reverse mode (NCXrm) in endothelial cells is controversial. We evaluated the participation of NCXrm-mediated Ca2+ uptake in the endothelium-dependent vasodilation of rat isolated mesenteric arterial beds. In phenylephrine-contracted mesenteries, the acetylcholine (ACh)-induced vasodilation was abolished by treatment with the NCXrm blockers SEA0400, KB-R7943, or SN-6. Consistent with that, the ACh-induced hyperpolarization observed in primary cultures of mesenteric endothelial cells and in smooth muscle of isolated mesenteric resistance arteries was attenuated by KB-R7943 and SEA0400, respectively. In addition, both blockers abolished the NO production activated by ACh in intact mesenteric arteries. In contrast, the inhibition of NCXrm did not affect the vasodilator responses induced by the Ca2+ ionophore, ionomycin, and the NO donor, S-nitroso- N-acetylpenicillamine. Furthermore, SEA0400, KB-R7943, and a small interference RNA directed against NCX1 blunted the increase in [Ca2+]i induced by ACh or ATP in cultured endothelial cells. The analysis by proximity ligation assay showed that the NO-synthesizing enzyme, eNOS, and NCX1 were associated in endothelial cell caveolae of intact mesenteric resistance arteries. These results indicate that the activation of NCXrm has a central role in Ca2+-mediated vasodilation initiated by ACh in endothelial cells of resistance arteries.-Lillo, M. A., Gaete, P. S., Puebla, M., Ardiles, N. M., Poblete, I., Becerra, A., Simon, F., Figueroa, X. F. Critical contribution of Na+-Ca2+ exchanger to the Ca2+-mediated vasodilation activated in endothelial cells of resistance arteries.
Collapse
Affiliation(s)
- Mauricio A Lillo
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo S Gaete
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mariela Puebla
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás M Ardiles
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Inés Poblete
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alvaro Becerra
- Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.,Departamento de Ciencias Químicas y Biológicas, Facultad de Salud, Deporte y Recreación, Universidad Bernardo O'Higgins, Santiago, Chile
| | - Felipe Simon
- Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Xavier F Figueroa
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
12
|
García-Cardeña G, Slegtenhorst BR. Hemodynamic Control of Endothelial Cell Fates in Development. Annu Rev Cell Dev Biol 2017; 32:633-648. [PMID: 27712101 DOI: 10.1146/annurev-cellbio-100814-125610] [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] [Indexed: 11/09/2022]
Abstract
Biomechanical forces are emerging as critical regulators of embryogenesis, particularly in the developing cardiovascular system. From the onset of blood flow, the embryonic vasculature is continuously exposed to a variety of hemodynamic forces. These biomechanical stimuli are key determinants of vascular cell specification and remodeling and the establishment of vascular homeostasis. In recent years, major advances have been made in our understanding of mechano-activated signaling networks that control both spatiotemporal and structural aspects of vascular development. It has become apparent that a major site for mechanotransduction is situated at the interface of blood and the vessel wall and that this process is controlled by the vascular endothelium. In this review, we discuss the hemodynamic control of endothelial cell fates, focusing on arterial-venous specification, lymphatic development, and the endothelial-to-hematopoietic transition, and present some recent insights into the mechano-activated pathways driving these cell fate decisions in the developing embryo.
Collapse
Affiliation(s)
- Guillermo García-Cardeña
- Program in Developmental and Regenerative Biology, Harvard Medical School, Boston, Massachusetts 02115; .,Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115
| | - Bendix R Slegtenhorst
- Program in Developmental and Regenerative Biology, Harvard Medical School, Boston, Massachusetts 02115; .,Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115.,Department of Surgery, Erasmus MC-University Medical Center, 3015 CE, Rotterdam, The Netherlands
| |
Collapse
|
13
|
Abstract
Flowing blood exerts a frictional force, fluid shear stress (FSS), on the endothelial cells that line the blood and lymphatic vessels. The magnitude, pulsatility, and directional characteristics of FSS are constantly sensed by the endothelium. Sustained increases or decreases in FSS induce vessel remodeling to maintain proper perfusion of tissue. In this review, we discuss these mechanisms and their relevance to physiology and disease, and propose a model for how information from different mechanosensors might be integrated to govern remodeling.
Collapse
Affiliation(s)
- Nicolas Baeyens
- Yale Cardiovascular Research Center, Department of Internal Medicine
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Department of Internal Medicine Department of Cell Biology, Yale University, New Haven, CT 06511 Department of Biomedical Engineering, Yale University, New Haven, CT 06511
| |
Collapse
|
14
|
Abstract
Fluid shear stress is an important environmental cue that governs vascular physiology and pathology, but the molecular mechanisms that mediate endothelial responses to flow are only partially understood. Gating of ion channels by flow is one mechanism that may underlie many of the known responses. Here, we review the literature on endothelial ion channels whose activity is modulated by flow with an eye toward identifying important questions for future research.
Collapse
Affiliation(s)
- Kristin A Gerhold
- Department of Internal Medicine (Cardiology), Yale Cardiovascular Research Center, Yale University, New Haven, Connecticut; and
| | - Martin A Schwartz
- Department of Internal Medicine (Cardiology), Yale Cardiovascular Research Center, Yale University, New Haven, Connecticut; and Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, Connecticut
| |
Collapse
|
15
|
Nuclear envelope proteins modulate proliferation of vascular smooth muscle cells during cyclic stretch application. Proc Natl Acad Sci U S A 2016; 113:5293-8. [PMID: 27114541 DOI: 10.1073/pnas.1604569113] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Cyclic stretch is an important inducer of vascular smooth muscle cell (VSMC) proliferation, which is crucial in vascular remodeling during hypertension. However, the molecular mechanism remains unclear. We studied the effects of emerin and lamin A/C, two important nuclear envelope proteins, on VSMC proliferation in hypertension and the underlying mechano-mechanisms. In common carotid artery of hypertensive rats in vivo and in cultured cells subjected to high (15%) cyclic stretch in vitro, VSMC proliferation was increased significantly, and the expression of emerin and lamin A/C was repressed compared with normotensive or normal (5%) cyclic stretch controls. Using targeted siRNA to mimic the repressed expression of emerin or lamin A/C induced by 15% stretch, we found that VSMC proliferation was enhanced under static and 5%-stretch conditions. Overexpression of emerin or lamin A/C reversed VSMC proliferation induced by 15% stretch. Hence, emerin and lamin A/C play critical roles in suppressing VSMC hyperproliferation induced by hyperstretch. ChIP-on-chip and MOTIF analyses showed that the DNAs binding with emerin contain three transcription factor motifs: CCNGGA, CCMGCC, and ABTTCCG; DNAs binding with lamin A/C contain the motifs CVGGAA, GCCGCYGC, and DAAGAAA. Protein/DNA array proved that altered emerin or lamin A/C expression modulated the activation of various transcription factors. Furthermore, accelerating local expression of emerin or lamin A/C reversed cell proliferation in the carotid artery of hypertensive rats in vivo. Our findings establish the pathogenetic role of emerin and lamin A/C repression in stretch-induced VSMC proliferation and suggest mechanobiological mechanism underlying this process that involves the sequence-specific binding of emerin and lamin A/C to specific transcription factor motifs.
Collapse
|
16
|
Burgess A, Shah K, Hough O, Hynynen K. Focused ultrasound-mediated drug delivery through the blood-brain barrier. Expert Rev Neurother 2016; 15:477-91. [PMID: 25936845 DOI: 10.1586/14737175.2015.1028369] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Despite recent advances in blood-brain barrier (BBB) research, it remains a significant hurdle for the pharmaceutical treatment of brain diseases. Focused ultrasound (FUS) is one method to transiently increase permeability of the BBB to promote drug delivery to specific brain regions. An introduction to the BBB and a brief overview of the methods, which can be used to circumvent the BBB to promote drug delivery, is provided. In particular, we discuss the advantages and limitations of FUS technology and the efficacy of FUS-mediated drug delivery in models of disease. MRI for targeting and evaluating FUS treatments, combined with administration of microbubbles, allows for transient, reproducible BBB opening. The integration of a real-time acoustic feedback controller has improved treatment safety. Successful clinical translation of FUS has the potential to transform the treatment of brain disease worldwide without requiring the development of new pharmaceutical agents.
Collapse
Affiliation(s)
- Alison Burgess
- Physical Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, S665, Toronto, ON M4N 3M5, Canada
| | | | | | | |
Collapse
|
17
|
Burgess A, Hynynen K. Microbubble-Assisted Ultrasound for Drug Delivery in the Brain and Central Nervous System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 880:293-308. [PMID: 26486344 DOI: 10.1007/978-3-319-22536-4_16] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The blood-brain barrier is a serious impediment to the delivery of pharmaceutical treatments for brain diseases, including cancer, neurodegenerative and neuropsychatric diseases. Focused ultrasound, when combined with microbubbles, has emerged as an effective method to transiently and locally open the blood-brain barrier to promote drug delivery to the brain. Focused ultrasound has been used to successfully deliver a wide variety of therapeutic agents to pre-clinical disease models. The requirement for clinical translation of focused ultrasound technology is considered.
Collapse
Affiliation(s)
- Alison Burgess
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Kullervo Hynynen
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada. .,Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
18
|
Siegel G, Meyer-Rath G, Ermilov E, Rodríguez M, Malmsten M, Claesson P, Saunders R, Hetzer R, Lindman B. Flow sensing in the cardiovascular system. Colloids Surf A Physicochem Eng Asp 2015. [DOI: 10.1016/j.colsurfa.2014.10.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
19
|
Roger S, Gillet L, Le Guennec JY, Besson P. Voltage-gated sodium channels and cancer: is excitability their primary role? Front Pharmacol 2015; 6:152. [PMID: 26283962 PMCID: PMC4518325 DOI: 10.3389/fphar.2015.00152] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 07/09/2015] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels (NaV) are molecular characteristics of excitable cells. Their activation, triggered by membrane depolarization, generates transient sodium currents that initiate action potentials in neurons and muscle cells. Sodium currents were discovered by Hodgkin and Huxley using the voltage clamp technique and reported in their landmark series of papers in 1952. It was only in the 1980's that sodium channel proteins from excitable membranes were molecularly characterized by Catterall and his collaborators. Non-excitable cells can also express NaV channels in physiological conditions as well as in pathological conditions. These NaV channels can sustain biological roles that are not related to the generation of action potentials. Interestingly, it is likely that the abnormal expression of NaV in pathological tissues can reflect the re-expression of a fetal phenotype. This is especially true in epithelial cancer cells for which these channels have been identified and sodium currents recorded, while it was not the case for cells from the cognate normal tissues. In cancers, the functional activity of NaV appeared to be involved in regulating the proliferative, migrative, and invasive properties of cells. This review is aimed at addressing the non-excitable roles of NaV channels with a specific emphasis in the regulation of cancer cell biology.
Collapse
Affiliation(s)
- Sébastien Roger
- Inserm UMR1069, Nutrition, Croissance et Cancer, Université François-Rabelais de Tours Tours, France ; Département de Physiologie Animale, UFR Sciences and Techniques, Université François-Rabelais de Tours Tours, France
| | - Ludovic Gillet
- Department of Clinical Research, University of Bern Bern, Switzerland
| | | | - Pierre Besson
- Inserm UMR1069, Nutrition, Croissance et Cancer, Université François-Rabelais de Tours Tours, France
| |
Collapse
|
20
|
Han Y, Wang L, Yao QP, Zhang P, Liu B, Wang GL, Shen BR, Cheng B, Wang Y, Jiang ZL, Qi YX. Nuclear envelope proteins Nesprin2 and LaminA regulate proliferation and apoptosis of vascular endothelial cells in response to shear stress. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1165-73. [PMID: 25721888 DOI: 10.1016/j.bbamcr.2015.02.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 01/19/2015] [Accepted: 02/15/2015] [Indexed: 11/27/2022]
Abstract
The dysfunction of vascular endothelial cells (ECs) influenced by flow shear stress is crucial for vascular remodeling. However, the roles of nuclear envelope (NE) proteins in shear stress-induced EC dysfunction are still unknown. Our results indicated that, compared with normal shear stress (NSS), low shear stress (LowSS) suppressed the expression of two types of NE proteins, Nesprin2 and LaminA, and increased the proliferation and apoptosis of ECs. Targeted small interfering RNA (siRNA) and gene overexpression plasmid transfection revealed that Nesprin2 and LaminA participate in the regulation of EC proliferation and apoptosis. A protein/DNA array was further used to detect the activation of transcription factors in ECs following transfection with target siRNAs and overexpression plasmids. The regulation of AP-2 and TFIID mediated by Nesprin2 and the activation of Stat-1, Stat-3, Stat-5 and Stat-6 by LaminA were verified under shear stress. Furthermore, using Ingenuity Pathway Analysis software and real-time RT-PCR, the effects of Nesprin2 or LaminA on the downstream target genes of AP-2, TFIID, and Stat-1, Stat-3, Stat-5 and Stat-6, respectively, were investigated under LowSS. Our study has revealed that NE proteins are novel mechano-sensitive molecules in ECs. LowSS suppresses the expression of Nesprin2 and LaminA, which may subsequently modulate the activation of important transcription factors and eventually lead to EC dysfunction.
Collapse
Affiliation(s)
- Yue Han
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Lu Wang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Qing-Ping Yao
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Zhang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Liu
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Liang Wang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Bao-Rong Shen
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Binbin Cheng
- Department of Bioengineering, University of CA, San Diego, USA
| | - Yingxiao Wang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China; Department of Bioengineering, University of CA, San Diego, USA
| | - Zong-Lai Jiang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ying-Xin Qi
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
21
|
Wild J, Soehnlein O, Dietel B, Urschel K, Garlichs CD, Cicha I. Rubbing salt into wounded endothelium: sodium potentiates proatherogenic effects of TNF-α under non-uniform shear stress. Thromb Haemost 2014; 112:183-95. [PMID: 24573382 DOI: 10.1160/th13-11-0908] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 02/10/2014] [Indexed: 02/07/2023]
Abstract
Increased consumption of sodium is a risk factor for hypertension and cardiovascular diseases. In vivo studies indicated that high dietary sodium may have a direct negative influence on endothelium. We investigated the effects of high sodium on the endothelial activation during early steps of atherogenesis. Endothelial cells (HUVECs) grown in a model of arterial bifurcations were exposed to shear stress in the presence of normal or high (+ 30 mmol/l) sodium. Adherent THP-1 cells, and the adhesion molecule expression were quantified. Sodium channel blockers, pathways' inhibitors, and siRNA against tonicity-responsive enhancer binding protein (TonEBP) were used to identify the mechanisms of sodium effects on endothelium. ApoE-deficient mice on low-fat diet received water containing normal or high salt (8% w/v) for four weeks, and the influence of dietary salt on inflammatory cell adhesion in the common carotid artery and carotid bifurcation was measured by intravital microscopy. In vitro, high sodium dramatically increased the endothelial responsiveness to tumour necrosis factor-α under non-uniform shear stress. Sodium-induced increase in monocytic cell adhesion was mediated by reactive oxygen species and the endothelial nitric oxygen synthase, and was sensitive to the knockdown of TonEBP. The results were subsequently confirmed in the ApoE-deficient mice. As compared with normal-salt group, high-salt intake significantly enhanced the adhesion of circulating CD11b+ cells to carotid bifurcations, but not to the straight segment of common carotid artery. In conclusion, elevated sodium has a direct effect on endothelial activation under atherogenic shear stress in vitro and in vivo, and promotes the endothelial-leukocyte interactions even in the absence of increased lipid concentrations.
Collapse
Affiliation(s)
| | | | | | | | | | - I Cicha
- Iwona Cicha, PhD, Cardiovascular Nanomedicine Unit, Section of Experimental Oncology and Nanomedicine, ENT Department, University of Erlangen-Nuremberg, Glückstr. 10, 91054 Erlangen, Germany, Tel.: +49 9131 8543953, Fax: +49 9131 8534282, E-mail:
| |
Collapse
|
22
|
Black J, Waxman S. Noncanonical Roles of Voltage-Gated Sodium Channels. Neuron 2013; 80:280-91. [DOI: 10.1016/j.neuron.2013.09.012] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2013] [Indexed: 12/19/2022]
|
23
|
Lee HJ, Li N, Evans SM, Diaz MF, Wenzel PL. Biomechanical force in blood development: extrinsic physical cues drive pro-hematopoietic signaling. Differentiation 2013; 86:92-103. [PMID: 23850217 DOI: 10.1016/j.diff.2013.06.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/17/2013] [Accepted: 06/19/2013] [Indexed: 02/07/2023]
Abstract
The hematopoietic system is dynamic during development and in adulthood, undergoing countless spatial and temporal transitions during the course of one's life. Microenvironmental cues in the many unique hematopoietic niches differ, characterized by distinct soluble molecules, membrane-bound factors, and biophysical features that meet the changing needs of the blood system. Research from the last decade has revealed the importance of substrate elasticity and biomechanical force in determination of stem cell fate. Our understanding of the role of these factors in hematopoiesis is still relatively poor; however, the developmental origin of blood cells from the endothelium provides a model for comparison. Many endothelial mechanical sensors and second messenger systems may also determine hematopoietic stem cell fate, self renewal, and homing behaviors. Further, the intimate contact of hematopoietic cells with mechanosensitive cell types, including osteoblasts, endothelial cells, mesenchymal stem cells, and pericytes, places them in close proximity to paracrine signaling downstream of mechanical signals. The objective of this review is to present an overview of the sensors and intracellular signaling pathways activated by mechanical cues and highlight the role of mechanotransductive pathways in hematopoiesis.
Collapse
Affiliation(s)
- Hyun Jung Lee
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, TX 77030, USA; Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
24
|
Kim HN, Jiao A, Hwang NS, Kim MS, Kang DH, Kim DH, Suh KY. Nanotopography-guided tissue engineering and regenerative medicine. Adv Drug Deliv Rev 2013; 65:536-58. [PMID: 22921841 PMCID: PMC5444877 DOI: 10.1016/j.addr.2012.07.014] [Citation(s) in RCA: 267] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Revised: 07/19/2012] [Accepted: 07/23/2012] [Indexed: 12/14/2022]
Abstract
Human tissues are intricate ensembles of multiple cell types embedded in complex and well-defined structures of the extracellular matrix (ECM). The organization of ECM is frequently hierarchical from nano to macro, with many proteins forming large scale structures with feature sizes up to several hundred microns. Inspired from these natural designs of ECM, nanotopography-guided approaches have been increasingly investigated for the last several decades. Results demonstrate that the nanotopography itself can activate tissue-specific function in vitro as well as promote tissue regeneration in vivo upon transplantation. In this review, we provide an extensive analysis of recent efforts to mimic functional nanostructures in vitro for improved tissue engineering and regeneration of injured and damaged tissues. We first characterize the role of various nanostructures in human tissues with respect to each tissue-specific function. Then, we describe various fabrication methods in terms of patterning principles and material characteristics. Finally, we summarize the applications of nanotopography to various tissues, which are classified into four types depending on their functions: protective, mechano-sensitive, electro-active, and shear stress-sensitive tissues. Some limitations and future challenges are briefly discussed at the end.
Collapse
Affiliation(s)
- Hong Nam Kim
- Division of WCU Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul 151-742, Republic of Korea
| | - Alex Jiao
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Nathaniel S. Hwang
- School of Chemical and Biological Engineering, Institute for Chemical Processing, Seoul National University, Seoul 151-742, Republic of Korea
| | - Min Sung Kim
- Division of WCU Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul 151-742, Republic of Korea
| | - Do Hyun Kang
- Division of WCU Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul 151-742, Republic of Korea
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Kahp-Yang Suh
- Division of WCU Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul 151-742, Republic of Korea
- Institute of Biological Engineering, Seoul National University, Seoul 151-742, Republic of Korea
| |
Collapse
|
25
|
Johnson BD, Mather KJ, Wallace JP. Mechanotransduction of shear in the endothelium: basic studies and clinical implications. Vasc Med 2012; 16:365-77. [PMID: 22003002 DOI: 10.1177/1358863x11422109] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The endothelium plays an integral role in the development and progression of atherosclerosis. Hemodynamic forces, particularly shear stress, have a powerful influence on endothelial phenotype and function; however, there is no clear consensus on how endothelial cells sense shear. Nevertheless, multiple endothelial cell signal transduction pathways are activated when exposed to shear stress in vitro. The type of shear, laminar or oscillatory, impacts which signal transduction pathways are initiated as well as which subsequent genes are up- or down-regulated, thereby influencing endothelial phenotype and function. Recently, human studies have examined the impact of shear stress and different shear patterns at rest and during exercise on endothelial function. Current evidence supports the theory that augmented exercise-induced shear stress contributes to improved endothelial function following acute exercise and exercise training, whereas retrograde shear initiates vascular dysfunction. The purpose of this review is to examine the current theories on how endothelial cells sense shear stress, to provide an overview on shear stress-induced signal transduction pathways and subsequent gene expression, and to review the current literature pertaining to shear stress and shear patterns at rest as well as during exercise in humans and the related effects on endothelial function.
Collapse
|
26
|
Black JA, Waxman SG. Sodium channels and microglial function. Exp Neurol 2011; 234:302-15. [PMID: 21985863 DOI: 10.1016/j.expneurol.2011.09.030] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 09/09/2011] [Accepted: 09/26/2011] [Indexed: 12/19/2022]
Abstract
Microglia are resident immune cells that provide continuous surveillance within the central nervous system (CNS) and respond to perturbations of brain and spinal cord parenchyma with an array of effector functions, including proliferation, migration, phagocytosis, secretions of multiple cytokines/chemokines and promotion of repair. To sense alterations within their environment, microglia express a large number of cell surface receptors, ion channels and adhesion molecules, which activate complex and dynamic signaling pathways. In the present chapter, we review studies that demonstrate that microglia in vivo and in vitro express specific voltage-gated sodium channel isoforms, and that blockade of sodium channel activity can attenuate several effector functions of microglia. These studies also provide strong evidence that Nav1.6 is the predominant sodium channel isoform expressed in microglia and that its activity contributes to the response of microglia to multiple activating signals.
Collapse
Affiliation(s)
- Joel A Black
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06511, USA.
| | | |
Collapse
|
27
|
Andrikopoulos P, Fraser SP, Patterson L, Ahmad Z, Burcu H, Ottaviani D, Diss JKJ, Box C, Eccles SA, Djamgoz MBA. Angiogenic functions of voltage-gated Na+ Channels in human endothelial cells: modulation of vascular endothelial growth factor (VEGF) signaling. J Biol Chem 2011; 286:16846-60. [PMID: 21385874 PMCID: PMC3089528 DOI: 10.1074/jbc.m110.187559] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 02/15/2011] [Indexed: 12/29/2022] Open
Abstract
Voltage-gated sodium channel (VGSC) activity has previously been reported in endothelial cells (ECs). However, the exact isoforms of VGSCs present, their mode(s) of action, and potential role(s) in angiogenesis have not been investigated. The main aims of this study were to determine the role of VGSC activity in angiogenic functions and to elucidate the potentially associated signaling mechanisms using human umbilical vein endothelial cells (HUVECs) as a model system. Real-time PCR showed that the primary functional VGSC α- and β-subunit isoforms in HUVECs were Nav1.5, Nav1.7, VGSCβ1, and VGSCβ3. Western blots verified that VGSCα proteins were expressed in HUVECs, and immunohistochemistry revealed VGSCα expression in mouse aortic ECs in vivo. Electrophysiological recordings showed that the channels were functional and suppressed by tetrodotoxin (TTX). VGSC activity modulated the following angiogenic properties of HUVECs: VEGF-induced proliferation or chemotaxis, tubular differentiation, and substrate adhesion. Interestingly, different aspects of angiogenesis were controlled by the different VGSC isoforms based on TTX sensitivity and effects of siRNA-mediated gene silencing. Additionally, we show for the first time that TTX-resistant (TTX-R) VGSCs (Nav1.5) potentiate VEGF-induced ERK1/2 activation through the PKCα-B-RAF signaling axis. We postulate that this potentiation occurs through modulation of VEGF-induced HUVEC depolarization and [Ca(2+)](i). We conclude that VGSCs regulate multiple angiogenic functions and VEGF signaling in HUVECs. Our results imply that targeting VGSC expression/activity could be a novel strategy for controlling angiogenesis.
Collapse
Affiliation(s)
- Petros Andrikopoulos
- From the Division of Cell and Molecular Biology, Neuroscience Solutions to Cancer Research Group, Sir Alexander Fleming Building, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom and
- the Cancer Research UK Cancer Therapeutics Unit, Institute of Cancer Research, McElwain Laboratories, Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, United Kingdom
| | - Scott P. Fraser
- From the Division of Cell and Molecular Biology, Neuroscience Solutions to Cancer Research Group, Sir Alexander Fleming Building, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom and
| | - Lisa Patterson
- the Cancer Research UK Cancer Therapeutics Unit, Institute of Cancer Research, McElwain Laboratories, Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, United Kingdom
| | - Zahida Ahmad
- the Cancer Research UK Cancer Therapeutics Unit, Institute of Cancer Research, McElwain Laboratories, Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, United Kingdom
| | - Hakan Burcu
- From the Division of Cell and Molecular Biology, Neuroscience Solutions to Cancer Research Group, Sir Alexander Fleming Building, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom and
- the Cancer Research UK Cancer Therapeutics Unit, Institute of Cancer Research, McElwain Laboratories, Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, United Kingdom
| | - Diego Ottaviani
- From the Division of Cell and Molecular Biology, Neuroscience Solutions to Cancer Research Group, Sir Alexander Fleming Building, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom and
- the Cancer Research UK Cancer Therapeutics Unit, Institute of Cancer Research, McElwain Laboratories, Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, United Kingdom
| | - James K. J. Diss
- From the Division of Cell and Molecular Biology, Neuroscience Solutions to Cancer Research Group, Sir Alexander Fleming Building, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom and
| | - Carol Box
- the Cancer Research UK Cancer Therapeutics Unit, Institute of Cancer Research, McElwain Laboratories, Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, United Kingdom
| | - Suzanne A. Eccles
- the Cancer Research UK Cancer Therapeutics Unit, Institute of Cancer Research, McElwain Laboratories, Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, United Kingdom
| | - Mustafa B. A. Djamgoz
- From the Division of Cell and Molecular Biology, Neuroscience Solutions to Cancer Research Group, Sir Alexander Fleming Building, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom and
| |
Collapse
|
28
|
Abstract
Endothelial cells are stimulated by shear stress throughout the vasculature and respond with changes in gene expression and by morphological reorganization. Mechanical sensors of the cell are varied and include cell surface sensors that activate intracellular chemical signaling pathways. Here, possible mechanical sensors of the cell including reorganization of the cytoskeleton and the nucleus are discussed in relation to shear flow. A mutation in the nuclear structural protein lamin A, related to Hutchinson-Gilford progeria syndrome, is reviewed specifically as the mutation results in altered nuclear structure and stiffer nuclei; animal models also suggest significantly altered vascular structure. Nuclear and cellular deformation of endothelial cells in response to shear stress provides partial understanding of possible mechanical regulation in the microcirculation. Increasing sophistication of fluid flow simulations inside the vessel is also an emerging area relevant to the microcirculation as visualization in situ is difficult. This integrated approach to study--including medicine, molecular and cell biology, biophysics and engineering--provides a unique understanding of multi-scale interactions in the microcirculation.
Collapse
Affiliation(s)
- Kris Noel Dahl
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA.
| | | | | |
Collapse
|
29
|
|
30
|
Shin HY, Schwartz EA, Bizios R, Gerritsen ME. Receptor-Mediated Basic Fibroblast Growth Factor Signaling Regulates Cyclic Pressure–Induced Human Endothelial Cell Proliferation. ACTA ACUST UNITED AC 2009; 11:285-91. [PMID: 15763948 DOI: 10.1080/10623320490904205] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Vascular endothelial cells sense and respond to pressure by molecular mechanism(s) which, to date, remain poorly understood. The present study investigated basic fibroblast growth factor (bFGF) signaling as a putative mechanotransduction pathway involved in the proliferative responses of human umbilical vein endothelia cells (HUVECs) to 60/20 mm Hg cyclic pressure at 1 Hz for 24 h. Under these conditions, the enhanced proliferative response of these HUVECs was not associated with an increased synthesis/release of bFGF, but involved rapid (within 30 min from the onset of exposure to pressure) tyrosine phosphorylation of the bFGF receptor, FGFR-2. Furthermore, monoclonal antibodies to either bFGF or FGFR-2 attenuated the increased proliferation of HUVECs exposed to 60/20 mm Hg cyclic pressure. HUVECs proliferation under 60/20 mm Hg at 1 Hz cyclic pressure is, therefore, dependent upon bFGF and involves FGFR-2 activation.
Collapse
Affiliation(s)
- Hainsworth Y Shin
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180-3590, USA
| | | | | | | |
Collapse
|
31
|
Chatterjee S, Chapman KE, Fisher AB. Lung ischemia: a model for endothelial mechanotransduction. Cell Biochem Biophys 2008; 52:125-38. [PMID: 18982455 PMCID: PMC2667227 DOI: 10.1007/s12013-008-9030-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2008] [Indexed: 10/21/2022]
Abstract
Endothelial cells in vivo are constantly exposed to shear associated with blood flow and altered shear stress elicits cellular responses (mechanotransduction). This review describes the role of shear sensors and signal transducers in these events. The major focus is the response to removal of shear as occurs when blood flow is compromised (i.e., ischemia). Pulmonary ischemia studied with the isolated murine lung or flow adapted pulmonary microvascular endothelial cells in vitro results in endothelial generation of reactive oxygen species (ROS) and NO. The response requires caveolae and is initiated by endothelial cell depolarization via K(ATP) channel closure followed by activation of NADPH oxidase (NOX2) and NO synthase (eNOS), signaling through MAP kinases, and endothelial cell proliferation. These physiological mediators can promote vasodilation and angiogenesis as compensation for decreased tissue perfusion.
Collapse
Affiliation(s)
- Shampa Chatterjee
- Institute for Environmental Medicine, University of Pennsylvania Medical Center, 1 John Morgan Building, 3620 Hamilton Walk, Philadelphia, PA, 19104-6068, USA
| | | | | |
Collapse
|
32
|
Black JA, Liu S, Carrithers M, Carrithers LM, Waxman SG. Exacerbation of experimental autoimmune encephalomyelitis after withdrawal of phenytoin and carbamazepine. Ann Neurol 2007; 62:21-33. [PMID: 17654737 DOI: 10.1002/ana.21172] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE In vitro observations and studies in murine experimental autoimmune encephalomyelitis (EAE) have shown protective effects of sodium channel blockers on central nervous system axons and improved clinical status when treatment is continued throughout the period of observation. Several clinical studies of sodium channel blockers are under way in patients with multiple sclerosis. Here we asked whether a protective effect would persist after withdrawal of a sodium channel blocker. METHODS We studied a mouse model of myelin oligodendrocyte glycoprotein-induced EAE treated with phenytoin or carbamazepine. RESULTS Both phenytoin and carbamazepine significantly improved the clinical course of the disease. Withdrawal of phenytoin resulted in acute exacerbation, accompanied by a significantly increased inflammatory infiltrate within the central nervous system and the death of nearly 60% of EAE mice. There were no clinical worsening or deaths in control mice after withdrawal of phenytoin. Withdrawal of carbamazepine led to acute worsening of EAE symptoms, increased inflammatory infiltrate, and was associated with the death of 8% of mice. INTERPRETATION These results, together with results showing effects of sodium channel blockers in immune cells, raise questions about the long-term effects of sodium channel blockers in neuroinflammatory disorders, and suggest that clinical studies of sodium channel blockers in these disorders should be planned carefully.
Collapse
Affiliation(s)
- Joel A Black
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | |
Collapse
|
33
|
Figueroa XF, Chen CC, Campbell KP, Damon DN, Day KH, Ramos S, Duling BR. Are voltage-dependent ion channels involved in the endothelial cell control of vasomotor tone? Am J Physiol Heart Circ Physiol 2007; 293:H1371-83. [PMID: 17513486 DOI: 10.1152/ajpheart.01368.2006] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the microcirculation, longitudinal conduction of vasomotor responses provides an essential means of coordinating flow distribution among vessels in a complex network. Spread of current along the vessel axis can display a regenerative component, which leads to propagation of vasomotor signals over many millimeters; the ionic basis for the regenerative response is unknown. We examined the responses to 10 s of focal electrical stimulation (30 Hz, 2 ms, 30 V) of mouse cremaster arterioles to test the hypothesis that voltage-dependent Na(+) (Na(v)) and Ca(2+) channels might be activated in long-distance signaling in microvessels. Electrical stimulation evoked a vasoconstriction at the site of stimulation and a spreading, nondecremental conducted dilation. Endothelial damage (air bubble) blocked conduction of the vasodilation, indicating an involvement of the endothelium. The Na(v) channel blocker bupivacaine also blocked conduction, and TTX attenuated it. The Na(v) channel activator veratridine induced an endothelium-dependent dilation. The Na(v) channel isoforms Na(v)1.2, Na(v)1.6, and Na(v)1.9 were detected in the endothelial cells of cremaster arterioles by immunocytochemistry. These findings are consistent with the involvement of Na(v) channels in the conducted response. BAPTA buffering of endothelial cell Ca(2+) delayed and reduced the conducted dilation, which was almost eliminated by Ni(2+), amiloride, or deletion of alpha(1H) T-type Ca(2+) (Ca(v)3.2) channels. Blockade of endothelial nitric oxide synthase or Ca(2+)-activated K(+) channels also inhibited the conducted vasodilation. Our findings indicate that an electrically induced signal can propagate along the vessel axis via the endothelium and can induce sequential activation of Na(v) and Ca(v)3.2 channels. The resultant Ca(2+) influx activates endothelial nitric oxide synthase and Ca(2+)-activated K(+) channels, triggering vasodilation.
Collapse
Affiliation(s)
- Xavier F Figueroa
- Departamento de Ciencias Fisiológicas, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | | | | | | | |
Collapse
|
34
|
Morris CE, Juranka PF. Lipid stress at play: mechanosensitivity of voltage-gated channels. CURRENT TOPICS IN MEMBRANES 2007; 59:297-338. [PMID: 25168141 DOI: 10.1016/s1063-5823(06)59011-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Membrane stretch modulates the activity of voltage-gated channels (VGCs). These channels are nearly ubiquitous among eukaryotes and they are present, too, in prokaryotes, so the potential ramifications of VGC mechanosensitivity are diverse. In situ traumatic stretch can irreversibly alter VGC activity with lethal results but that is pathology. This chapter discusses the reversible responses of VGCs to stretch, with the general relation of stretch stimuli to other forms of lipid stress, and briefly, with some irreversible stretch effects (=stretch trauma). A working assumption throughout is that mechanosensitive (MS) VGC motions-that is, motions that respond reversibly to bilayer stretch-are susceptible to other forms of lipid stress, such as the stresses produced when amphiphilic molecules (anesthetics, lipids, alcohols, and lipophilic drugs) are inserted into the bilayer. Insofar as these molecules change the bilayer's lateral pressure profile, they can be termed bilayer mechanical reagents (BMRs). The chapter also discusses the MS VGC behavior against the backdrop of eukaryotic channels more widely accepted as "MS channels"--namely, the transient receptor potential (TRP)-based MS cation channels.
Collapse
Affiliation(s)
- Catherine E Morris
- Neuroscience, Ottawa Health Research Institute, Ottawa Hospital, Ottawa, Ontario K1Y 4E9, Canada
| | - Peter F Juranka
- Neuroscience, Ottawa Health Research Institute, Ottawa Hospital, Ottawa, Ontario K1Y 4E9, Canada
| |
Collapse
|
35
|
Abstract
Vascular endothelial cells (ECs) play significant roles in regulating circulatory functions. Mechanical stimuli, including the stretch and shear stress resulting from circulatory pressure and flow, modulate EC functions by activating mechanosensors, signaling pathways, and gene and protein expressions. Mechanical forces with a clear direction (e.g., the pulsatile shear stress and the uniaxial circumferential stretch existing in the straight part of the arterial tree) cause only transient molecular signaling of pro-inflammatory and proliferative pathways, which become downregulated when such directed mechanical forces are sustained. In contrast, mechanical forces without a definitive direction (e.g., disturbed flow and relatively undirected stretch seen at branch points and other regions of complex geometry) cause sustained molecular signaling of pro-inflammatory and proliferative pathways. The EC responses to directed mechanical stimuli involve the remodeling of EC structure to minimize alterations in intracellular stress/strain and elicit adaptive changes in EC signaling in the face of sustained stimuli; these cellular events constitute a feedback control mechanism to maintain vascular homeostasis and are atheroprotective. Such a feedback mechanism does not operate effectively in regions of complex geometry, where the mechanical stimuli do not have clear directions, thus placing these areas at risk for atherogenesis. The mechanotransduction-induced EC adaptive processes in the straight part of the aorta represent a case of the “Wisdom of the Cell,” as a part of the more general concept of the “Wisdom of the Body” promulgated by Cannon, to maintain cellular homeostasis in the face of external perturbations.
Collapse
Affiliation(s)
- Shu Chien
- Dept of Bioengineering, PFBH, Rm 134, Univ of California, San Diego, La Jolla, CA 92093-0412, USA.
| |
Collapse
|
36
|
Lee-Kwon W, Goo JH, Zhang Z, Silldorff EP, Pallone TL. Vasa recta voltage-gated Na+ channel Nav1.3 is regulated by calmodulin. Am J Physiol Renal Physiol 2006; 292:F404-14. [PMID: 16912065 DOI: 10.1152/ajprenal.00070.2006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Rat descending vasa recta (DVR) express a tetrodotoxin (TTX)-sensitive voltage-operated Na(+) (Na(V)) conductance. We examined expression of Na(V) isoforms in DVR and tested for regulation of Na(V) currents by calmodulin (CaM). RT-PCR in isolated permeabilized DVR using degenerate primers targeted to TTX-sensitive isoforms amplified a product whose sequence identified only Na(V)1.3. Immunoblot of outer medullary homogenate verified Na(V)1.3 expression, and fluorescent immunochemistry showed Na(V)1.3 expression in isolated vessels. Immunochemistry in outer medullary serial sections confirmed that Na(V)1.3 is confined to alpha-smooth muscle actin-positive vascular bundles. Na(V)1.3 possesses a COOH-terminal CaM binding motifs. Using pull-down assays and immunoprecipitation experiments, we verified that CaM binds to either full-length Na(V)1.3 or a GST-Na(V)1.3 COOH-terminal fusion protein. In patch-clamp experiments, Na(V) currents were suppressed by calmodulin inhibitory peptide (CIP; 100 nM) or the CaM inhibitor N-(6-aminohexyl)-5-chloro-1-naphthalene-sulphonamide hydrochloride (W7). Neither CIP nor W7 altered the voltage dependence of pericyte Na(V) currents; however, raising electrode free Ca(2+) from 20 to approximately 2,000 nM produced a depolarizing shift of activation. In vitro binding of CaM to GST-Na(V)1.3C was not affected by Ca(2+) concentration. We conclude that Na(V)1.3 is expressed by DVR, binds to CaM, and is regulated by CaM and Ca(2+). Inhibition of CaM binding suppresses pericyte Na(V) currents.
Collapse
Affiliation(s)
- Whaseon Lee-Kwon
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore 21201, USA
| | | | | | | | | |
Collapse
|
37
|
Fuster V, Moreno PR, Fayad ZA, Corti R, Badimon JJ. Atherothrombosis and high-risk plaque: part I: evolving concepts. J Am Coll Cardiol 2005; 46:937-54. [PMID: 16168274 DOI: 10.1016/j.jacc.2005.03.074] [Citation(s) in RCA: 502] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2004] [Revised: 01/04/2005] [Accepted: 03/04/2005] [Indexed: 12/29/2022]
Abstract
Atherothrombosis is a complex disease in which cholesterol deposition, inflammation, and thrombus formation play a major role. Rupture of high-risk, vulnerable plaques is responsible for coronary thrombosis, the main cause of unstable angina, acute myocardial infarction, and sudden cardiac death. In addition to rupture, plaque erosion may also lead to occlusive thrombosis and acute coronary events. Atherothrombosis can be evaluated according to histologic criteria, most commonly categorized by the American Heart Association (AHA) classification. However, this classification does not include the thin cap fibroatheroma, the most common form of high-risk, vulnerable plaque. Furthermore, the AHA classification does not include plaque erosion. As a result, new classifications have emerged and are reviewed in this article. The disease is asymptomatic during a long period and dramatically changes its course when complicated by thrombosis. This is summarized in five phases, from early lesions to plaque rupture, followed by plaque healing and fibrocalcification. For the early phases, the role of endothelial dysfunction, cholesterol transport, high-density lipoprotein, and proteoglycans are discussed. Furthermore, the innate and adaptive immune response to autoantigens, the Toll-like receptors, and the mechanisms of calcification are carefully analyzed. For the advanced phases, the role of eccentric remodeling, vasa vasorum neovascularization, and mechanisms of plaque rupture are systematically evaluated. In the final thrombosis section, focal and circulating tissue factor associated with apoptotic macrophages and circulatory monocytes is examined, closing the link between inflammation, plaque rupture, and blood thrombogenicity.
Collapse
Affiliation(s)
- Valentin Fuster
- Zena and Michael A. Wiener Cardiovascular Institute and the Marie-Josee and Henry R. Kravis Cardiovascular Health Center, The Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | |
Collapse
|
38
|
Abstract
Many cells are constantly exposed to fluid mechanical forces generated by flowing blood, and wall shear stresses modulate aspects of their structure and function. However, the mechanisms for mechanotransduction of flow are not well understood. Here we report that TRPM7, which is both an ion channel and a functional kinase, is translocated within cells in response to laminar flow. After exposure of cells to physiological values of laminar fluid flow, the number of TRPM7 molecules localized at or near the plasma membrane increased up to 2-fold, in less than 100 seconds. This increase in membrane-localized GFP-TRPM7, as seen by total internal reflection fluorescence microscopy, closely correlated with increases in TRPM7 current. Both endogenous and heterologously expressed TRPM7 was found in tubulovesicular structures that were translocated to the region of the plasma membrane on induction of shear stress. In vascular smooth muscle cells, but not in several types of endothelial cells, fluid flow increased endogenous native TRPM7 current amplitude. We hypothesize that TRPM7 plays a role in pathological response to vessel wall injury.
Collapse
Affiliation(s)
- Elena Oancea
- Howard Hughes Medical Institute, Department of Cardiology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
39
|
Fraser SP, Diss JKJ, Lloyd LJ, Pani F, Chioni AM, George AJT, Djamgoz MBA. T-lymphocyte invasiveness: control by voltage-gated Na+channel activity. FEBS Lett 2004; 569:191-4. [PMID: 15225632 DOI: 10.1016/j.febslet.2004.05.063] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2004] [Accepted: 05/04/2004] [Indexed: 01/12/2023]
Abstract
Whole-cell patch-clamp recordings showed that a sub-population (10%) of Jurkat cells, a model of human T-cells, expressed a functional voltage-gated sodium channel, which was tetrodotoxin (TTX)-resistant. Expression of voltage-gated sodium channel protein was confirmed by western blots. Semi-quantitative PCR analysis revealed that mRNAs for the alpha-subunits of multiple voltage-gated sodium channel subtypes were present but indicated that Na(v)1.5 was the predominant subtype, consistent with the TTX-resistant nature of the recorded currents. Importantly, 10 microM TTX reduced the number of Jurkat cells invading a Matrigel basement membrane by 93.0+/-5.5%. Since similar sodium channels have also been detected in normal human T-lymphocytes, it is concluded that the activity of voltage-gated sodium channels could represent a novel mechanism potentiating the invasive capacity of these cells.
Collapse
Affiliation(s)
- Scott P Fraser
- Department of Biological Sciences, Sir Alexander Fleming Building, Imperial College London, South Kensington Campus, London SW7 2AZ, UK.
| | | | | | | | | | | | | |
Collapse
|
40
|
Chien S. Molecular and mechanical bases of focal lipid accumulation in arterial wall. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2003; 83:131-51. [PMID: 12865076 DOI: 10.1016/s0079-6107(03)00053-1] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mechanical forces such as shear stress can modulate gene and protein expressions and hence cellular functions by activating membrane sensors and intracellular signaling. Using cultured endothelial cells, we have shown that laminar shear stress causes a transient increase in monocyte chemotactic protein-1 (MCP-1) expression, which involves the Ras-MAP kinase signaling pathway. We have demonstrated that integrins and the vascular endothelial growth factor receptor Flk-1 can sense shear stress, with integrins being upstream to Flk-1. Other possible membrane components involved in the sensing of shear stress include G-protein coupled receptors, intercellular junction proteins, membrane glycocalyx, and the lipid bilayer. Mechano-transduction involves the participation of a multitude of sensors, signaling molecules, and genes. Microarray analysis has demonstrated that shear stress can upregulate and downregulate different genes. Sustained shear stress downregulates atherogenic genes (e.g., MCP-1 and the genes that facilitate lipid accumulation) and upregulates growth-arrest genes. In contrast, disturbed flow observed at branch points and simulated in step-flow channels causes sustained activation of MCP-1 and the genes facilitating cell turnover and lipid accumulation. These findings provide a molecular basis for the explanation of the preferential localization of atherosclerotic lesions at regions of disturbed flow, such as the arterial branch points. The combination of mechanics and biology (from molecules-cells to organs-systems) can help to elucidate the physiological processes of mechano-chemical transduction and improving the methods of the management of important clinical conditions such as coronary artery disease.
Collapse
Affiliation(s)
- Shu Chien
- Department of Bioengineering and Medicine, The Whitaker Institute for Biomedical Engineering, University of California, San Diego, 9500 Gilman Drive, Mail Code 0427, SERF Room 221, La Jolla, CA 92093-0427, USA.
| |
Collapse
|
41
|
Düwel P, Jüngling E, Westhofen M, Lückhoff A. Potassium currents in vestibular type II hair cells activated by hydrostatic pressure. Neuroscience 2003; 116:963-72. [PMID: 12617937 DOI: 10.1016/s0306-4522(02)00776-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
An elevated hydrostatic pressure in the endolymphatic space of the inner ear is discussed as pathophysiological factor in hydrops-related diseases of the inner ear. An increase in pressure by fractions of 1 cm H(2)O is sufficient to induce vertigo-like symptoms in animal models. To establish a link between hydrostatic pressure and the function of vestibular hair cells, we studied potassium currents in isolated vestibular type II hair cells from guinea-pig utricles when the hydrostatic pressure was increased by raising the height of the bath from 0.2-0.5, 0.7 or 1.0 cm. Elevated pressure enhanced K(+) currents significantly; a rise in pressure from 0.2-0.5 cm H(2)O increased the total K(+) current at +40 mV by 22+/-14% (+/-S.D.). The pressure-sensitive current I(K,p) was non-inactivating during depolarizing pulses. It was maintained when the pressure was kept elevated for several minutes and receded promptly after return to a pressure of 0.2 cm H(2)O. Voltage-gated Ca(2+) currents, in contrast, were not altered by hydrostatic pressure. A pharmacological characterization of I(K,p) revealed that tetraetylammonium (100 mM) abolished all outward currents including I(K,p). I(K,p) was partly and reversibly inhibited by 4-aminopyridine. Dihydrostreptomycin, a blocker of the transduction channel, left I(K,p) unaffected. Charybdotoxin (100 nM), a blocker of Ca(2+)-dependent K(+) channels, completely yet reversibly abolished I(K,p). We conclude that small elevations in hydrostatic pressure evoke a charybdotoxin-sensitive, probably Ca(2+)-dependent K(+) current in vestibular hair cells. This is likely to alter their frequency response and may be a relevant mechanism how hydrostatic pressure disturbs transduction.
Collapse
Affiliation(s)
- P Düwel
- Institute of Physiology, University Hospital RWTH, Aachen, Germany.
| | | | | | | |
Collapse
|
42
|
Abstract
Blood vessels are permanently subjected to mechanical forces in the form of stretch, encompassing cyclic mechanical strain due to the pulsatile nature of blood flow, and shear stress. Alterations in stretch or shear stress invariably produce transformations in the vessel wall that will aim to accommodate the new conditions and to ultimately restore basal levels of tensile stress and shear stress. Vascular cells are equipped with numerous receptors that allow them to detect and respond to the mechanical forces generated by pressure and shear stress. The cytoskeleton and other structural components have an established role in mechanotransduction, being able to transmit and modulate tension within the cell via focal adhesion sites, integrins, cellular junctions and the extracellular matrix. Beyond the structural modifications incurred, mechanical forces can also initiate complex signal transduction cascades leading to functional changes within the cell. Many intracellular pathways, including the MAP kinase cascade, are activated by flow or stretch and initiate, via sequential phosphorylations, the activation of transcription factors and subsequent gene expression.
Collapse
|
43
|
Resnick N, Yahav H, Shay-Salit A, Shushy M, Schubert S, Zilberman LCM, Wofovitz E. Fluid shear stress and the vascular endothelium: for better and for worse. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2003; 81:177-99. [PMID: 12732261 DOI: 10.1016/s0079-6107(02)00052-4] [Citation(s) in RCA: 382] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
As blood flows, the vascular wall is constantly subjected to physical forces, which regulate important physiological blood vessel responses, as well as being implicated in the development of arterial wall pathologies. Changes in blood flow, thus generating altered hemodynamic forces are responsible for acute vessel tone regulation, the development of blood vessel structure during embryogenesis and early growth, as well as chronic remodeling and generation of adult blood vessels. The complex interaction of biomechanical forces, and more specifically shear stress, derived by the flow of blood and the vascular endothelium raise many yet to be answered questions:How are mechanical forces transduced by endothelial cells into a biological response, and is there a "shear stress receptor"?Are "mechanical receptors" and the final signaling pathways they evoke similar to other stimulus-response transduction systems?How do vascular endothelial cells differ in their response to physiological or pathological shear stresses?Can shear stress receptors or shear stress responsive genes serve as novel targets for the design of diagnostic and therapeutic modalities for cardiovascular pathologies?The current review attempts to bring together recent findings on the in vivo and in vitro responses of the vascular endothelium to shear stress and to address some of the questions raised above.
Collapse
Affiliation(s)
- Nitzan Resnick
- Department of Anatomy and Cell Biology, Bruce Rappaport Research Institute, Bruce Rappaport Faculty of Medicine, P.O. Box. 9697 Technion Bat-Galim, Haifa, 31096 Israel.
| | | | | | | | | | | | | |
Collapse
|
44
|
Paszkowiak JJ, Dardik A. Arterial wall shear stress: observations from the bench to the bedside. Vasc Endovascular Surg 2003; 37:47-57. [PMID: 12577139 DOI: 10.1177/153857440303700107] [Citation(s) in RCA: 222] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Shear stress is the tangential force of the flowing blood on the endothelial surface of the blood vessel. Shear is described mathematically or ideal fluids, and in vitro models have enabled researchers to describe the effects of shear on endothelial cells. High shear stress, as found in laminar flow, promotes endothelial cell survival and quiescence, alignment in the direction of flow, and secretion of substances that promote vasodilation and anticoagulation. Low shear stress, or changing shear stress direction as found in turbulent flow, promotes endothelial proliferation and apoptosis, shape change, and secretion of substances that promote vasoconstriction, coagulation, and platelet aggregation. The precise pathways by which endothelial cells sense shear stress to promote their quiescent or activated pathways are currently unknown. Clinical applications include increasing shear stress via creation of an arteriovenous fistula or vein cuff to promote bypass graft flow and patency. Since an abnormal level of shear stress is implicated in the pathogenesis of atherosclerosis, neointimal hyperplasia, and aneurysmal disease, additional research to understand the effects of shear stress on the blood vessel may provide insight to prevent vascular disease.
Collapse
|
45
|
Abstract
OBJECTIVE The endothelium is normally subjected to mechanical deformation resulting from shear stress and from strain associated with stretch of the vessel wall. These stimuli are detected by a mechanosensor that initiates a variety of signaling systems responsible for triggering the functional responses. The identity of the mechanosensor has not been established. This article discusses the different mechanisms of mechanosensing that have been proposed and reviews the literature with respect to signaling systems that are activated in response to stress and strain in endothelium. DATA SOURCES Published literature related to mechanotransduction, signal transduction pathways initiated by strain in endothelium, and pathophysiologic effects of abnormal shear forces in diseases. DATA EXTRACTION AND SYNTHESIS Proposed mechanisms of mechanosensing include stretch-sensitive ion channels, protein kinases associated with the cytoskeleton, integrin-cytoskeletal interactions, cytoskeletal-nuclear interactions, and oxidase systems capable of generating reactive oxygen species. However, the molecular identity of the mechanosensor is not known, nor is it clear whether multiple sensing mechanisms exist. CONCLUSIONS Many responses are initiated in cells subjected to mechanical deformation, including alterations in ion channel conductance, activation of signal transduction pathways, and altered expression of specific genes. Future progress in this field will require a critical distinction between cell systems that become activated during mechanical strain and the identity of the cellular mechanosensor that triggers subsequent responses.
Collapse
Affiliation(s)
- Mir H Ali
- University of Chicago, Pulmonary and Critical Care Medicine, 5841 South Maryland Avenue, Chicago, IL, USA
| | | |
Collapse
|
46
|
Yarbrough TL, Lu T, Lee HC, Shibata EF. Localization of cardiac sodium channels in caveolin-rich membrane domains: regulation of sodium current amplitude. Circ Res 2002; 90:443-9. [PMID: 11884374 DOI: 10.1161/hh0402.105177] [Citation(s) in RCA: 151] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study demonstrates that caveolae, omega-shaped membrane invaginations, are involved in cardiac sodium channel regulation by a mechanism involving the alpha subunit of the stimulatory heterotrimeric G-protein, Galpha(s), via stimulation of the cell surface beta-adrenergic receptor. Stimulation of beta-adrenergic receptors with 10 micromol/L isoproterenol in the presence of a protein kinase A inhibitor increased the whole-cell sodium current by a "direct" cAMP-independent G-protein mechanism. The addition of antibodies against caveolin-3 to the cell's cytoplasm via the pipette solution abrogated this direct G protein-induced increase in sodium current, whereas antibodies to caveolin-1 or caveolin-2 did not. Voltage-gated sodium channel proteins were found to associate with caveolin-rich membranes obtained by detergent-free buoyant density separation. The purity of the caveolar membrane fraction was verified by Western blot analyses, which indicated that endoplasmic/sarcoplasmic reticulum, endosomal compartments, Golgi apparatus, clathrin-coated vesicles, and sarcolemmal membranes were excluded from the caveolin-rich membrane fraction. Additionally, the sodium channel was found to colocalize with caveolar membranes by immunoprecipitation, indirect immunofluorescence, and immunogold transmission electron microscopy. These results suggest that stimulation of beta-adrenergic receptors, and thereby Galpha(s), promotes the presentation of cardiac sodium channels associated with caveolar membranes to the sarcolemma.
Collapse
Affiliation(s)
- Tracy L Yarbrough
- Department of Physiology, University of Iowa College of Medicine, Iowa City, Iowa 52242-1109, USA
| | | | | | | |
Collapse
|
47
|
Abstract
Endothelial cells (EC) form a unique signal-transducing surface in the vascular system. The abundance of ion channels in the plasma membrane of these nonexcitable cells has raised questions about their functional role. This review presents evidence for the involvement of ion channels in endothelial cell functions controlled by intracellular Ca(2+) signals, such as the production and release of many vasoactive factors, e.g., nitric oxide and PGI(2). In addition, ion channels may be involved in the regulation of the traffic of macromolecules by endocytosis, transcytosis, the biosynthetic-secretory pathway, and exocytosis, e.g., tissue factor pathway inhibitor, von Willebrand factor, and tissue plasminogen activator. Ion channels are also involved in controlling intercellular permeability, EC proliferation, and angiogenesis. These functions are supported or triggered via ion channels, which either provide Ca(2+)-entry pathways or stabilize the driving force for Ca(2+) influx through these pathways. These Ca(2+)-entry pathways comprise agonist-activated nonselective Ca(2+)-permeable cation channels, cyclic nucleotide-activated nonselective cation channels, and store-operated Ca(2+) channels or capacitative Ca(2+) entry. At least some of these channels appear to be expressed by genes of the trp family. The driving force for Ca(2+) entry is mainly controlled by large-conductance Ca(2+)-dependent BK(Ca) channels (slo), inwardly rectifying K(+) channels (Kir2.1), and at least two types of Cl( -) channels, i.e., the Ca(2+)-activated Cl(-) channel and the housekeeping, volume-regulated anion channel (VRAC). In addition to their essential function in Ca(2+) signaling, VRAC channels are multifunctional, operate as a transport pathway for amino acids and organic osmolytes, and are possibly involved in endothelial cell proliferation and angiogenesis. Finally, we have also highlighted the role of ion channels as mechanosensors in EC. Plasmalemmal ion channels may signal rapid changes in hemodynamic forces, such as shear stress and biaxial tensile stress, but also changes in cell shape and cell volume to the cytoskeleton and the intracellular machinery for metabolite traffic and gene expression.
Collapse
Affiliation(s)
- B Nilius
- Department of Physiology, KU Leuven, Campus Gasthuisberg, Leuven, Belgium.
| | | |
Collapse
|
48
|
Satlin LM, Sheng S, Woda CB, Kleyman TR. Epithelial Na(+) channels are regulated by flow. Am J Physiol Renal Physiol 2001; 280:F1010-8. [PMID: 11352841 DOI: 10.1152/ajprenal.2001.280.6.f1010] [Citation(s) in RCA: 166] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Na(+) absorption in the renal cortical collecting duct (CCD) is mediated by apical epithelial Na(+) channels (ENaCs). The CCD is subject to continuous variations in intraluminal flow rate that we speculate alters hydrostatic pressure, membrane stretch, and shear stress. Although ENaCs share limited sequence homology with putative mechanosensitive ion channels in Caenorhabditis elegans, controversy exists as to whether ENaCs are regulated by biomechanical forces. We examined the effect of varying the rate of fluid flow on whole cell Na(+) currents (I(Na)) in oocytes expressing mouse alpha,beta,gamma-ENaC (mENaC) and on net Na(+) absorption in microperfused rabbit CCDs. Oocytes injected with mENaC but not water responded to the initiation of superfusate flow (to 4-6 ml/min) with a reversible threefold stimulation of I(Na) without a change in reversal potential. The increase in I(Na) was variable among oocytes. CCDs responded to a threefold increase in rate of luminal flow with a twofold increase in the rate of net Na(+) absorption. An increase in luminal viscosity achieved by addition of 5% dextran to the luminal perfusate did not alter the rate of net Na(+) absorption, suggesting that shear stress does not influence Na(+) transport in the CCD. In sum, our data suggest that flow stimulation of ENaC activity and Na(+) absorption is mediated by an increase in hydrostatic pressure and/or membrane stretch. We propose that intraluminal flow rate may be an important regulator of channel activity in the CCD.
Collapse
Affiliation(s)
- L M Satlin
- Department of Pediatrics, Mount Sinai School of Medicine, New York 10029-6574, USA.
| | | | | | | |
Collapse
|
49
|
Resnick N, Yahav H, Schubert S, Wolfovitz E, Shay A. Signalling pathways in vascular endothelium activated by shear stress: relevance to atherosclerosis. Curr Opin Lipidol 2000; 11:167-77. [PMID: 10787179 DOI: 10.1097/00041433-200004000-00010] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Major advances in our understanding of how endothelial cells sense and respond to haemodynamic forces and, more specifically, to fluid shear stress have been achieved during the past 3 years. These include definition of potential shear stress receptors and multiple signalling pathways that mediate shear stress regulation of gene expression. A few studies have also pointed to the unique effects of complex shear stress on endothelial activation, thus leading to better understanding of the mechanisms that lead to the development of atherosclerosis.
Collapse
Affiliation(s)
- N Resnick
- The Department of Anatomy and Cell Biology, Bruce Rappaport Research Institute, Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel.
| | | | | | | | | |
Collapse
|