1
|
McDuffie EL, Panettieri RA, Scott CP. G 12/13 signaling in asthma. Respir Res 2024; 25:295. [PMID: 39095798 PMCID: PMC11297630 DOI: 10.1186/s12931-024-02920-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024] Open
Abstract
Shortening of airway smooth muscle and bronchoconstriction are pathognomonic for asthma. Airway shortening occurs through calcium-dependent activation of myosin light chain kinase, and RhoA-dependent calcium sensitization, which inhibits myosin light chain phosphatase. The mechanism through which pro-contractile stimuli activate calcium sensitization is poorly understood. Our review of the literature suggests that pro-contractile G protein coupled receptors likely signal through G12/13 to activate RhoA and mediate calcium sensitization. This hypothesis is consistent with the effects of pro-contractile agonists on RhoA and Rho kinase activation, actin polymerization and myosin light chain phosphorylation. Recognizing the likely role of G12/13 signaling in the pathophysiology of asthma rationalizes the effects of pro-contractile stimuli on airway hyperresponsiveness, immune activation and airway remodeling, and suggests new approaches for asthma treatment.
Collapse
Affiliation(s)
- Elizabeth L McDuffie
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, NJ, USA
| | - Charles P Scott
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Coccarelli A, Pant S, Polydoros I, Harraz OF. A new model for evaluating pressure-induced vascular tone in small cerebral arteries. Biomech Model Mechanobiol 2024; 23:271-286. [PMID: 37925376 PMCID: PMC10901969 DOI: 10.1007/s10237-023-01774-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/17/2023] [Indexed: 11/06/2023]
Abstract
The capacity of small cerebral arteries (SCAs) to adapt to pressure fluctuations has a fundamental physiological role and appears to be relevant in different pathological conditions. Here, we present a new computational model for quantifying the link, and its contributors, between luminal pressure and vascular tone generation in SCAs. This is assembled by combining a chemical sub-model, representing pressure-induced smooth muscle cell (SMC) signalling, with a mechanical sub-model for the tone generation and its transduction at tissue level. The devised model can accurately reproduce the impact of luminal pressure on different cytoplasmic components involved in myogenic signalling, both in the control case and when combined with some specific pharmacological interventions. Furthermore, the model is also able to capture and predict experimentally recorded pressure-outer diameter relationships obtained for vessels under control conditions, both in a Ca2 + -free bath and under drug inhibition. The modularity of the proposed framework allows the integration of new components for the study of a broad range of processes involved in the vascular function.
Collapse
Affiliation(s)
- Alberto Coccarelli
- Zienkiewicz Institute for Modelling, Data and AI, Faculty of Science and Engineering, Swansea University, Swansea, UK.
| | - Sanjay Pant
- Zienkiewicz Institute for Modelling, Data and AI, Faculty of Science and Engineering, Swansea University, Swansea, UK
| | - Ioannis Polydoros
- Zienkiewicz Institute for Modelling, Data and AI, Faculty of Science and Engineering, Swansea University, Swansea, UK
| | - Osama F Harraz
- Department of Pharmacology, Larner College of Medicine, and Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, USA
| |
Collapse
|
3
|
Wang Y, Liao G, Wu Y, Wang R, Tang DD. The intermediate filament protein nestin serves as a molecular hub for smooth muscle cytoskeletal signaling. Respir Res 2023; 24:157. [PMID: 37316833 DOI: 10.1186/s12931-023-02473-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/08/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND The recruitment of the actin-regulatory proteins cortactin and profilin-1 (Pfn-1) to the membrane is important for the regulation of actin cytoskeletal reorganization and smooth muscle contraction. Polo-like kinase 1 (Plk1) and the type III intermediate filament protein vimentin are involved in smooth muscle contraction. Regulation of complex cytoskeletal signaling is not entirely elucidated. The aim of this study was to evaluate the role of nestin (a type VI intermediate filament protein) in cytoskeletal signaling in airway smooth muscle. METHODS Nestin expression in human airway smooth muscle (HASM) was knocked down by specific shRNA or siRNA. The effects of nestin knockdown (KD) on the recruitment of cortactin and Pfn-1, actin polymerization, myosin light chain (MLC) phosphorylation, and contraction were evaluated by cellular and physiological approaches. Moreover, we assessed the effects of non-phosphorylatable nestin mutant on these biological processes. RESULTS Nestin KD reduced the recruitment of cortactin and Pfn-1, actin polymerization, and HASM contraction without affecting MLC phosphorylation. Moreover, contractile stimulation enhanced nestin phosphorylation at Thr-315 and the interaction of nestin with Plk1. Nestin KD also diminished phosphorylation of Plk1 and vimentin. The expression of T315A nestin mutant (alanine substitution at Thr-315) reduced the recruitment of cortactin and Pfn-1, actin polymerization, and HASM contraction without affecting MLC phosphorylation. Furthermore, Plk1 KD diminished nestin phosphorylation at this residue. CONCLUSIONS Nestin is an essential macromolecule that regulates actin cytoskeletal signaling via Plk1 in smooth muscle. Plk1 and nestin form an activation loop during contractile stimulation.
Collapse
Affiliation(s)
- Yinna Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, MC-8, Albany, NY, 12208, USA
| | - Guoning Liao
- Department of Molecular and Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, MC-8, Albany, NY, 12208, USA
| | - Yidi Wu
- Department of Molecular and Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, MC-8, Albany, NY, 12208, USA
| | - Ruping Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, MC-8, Albany, NY, 12208, USA
| | - Dale D Tang
- Department of Molecular and Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, MC-8, Albany, NY, 12208, USA.
| |
Collapse
|
4
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
5
|
Lubomirov LT, Schroeter MM, Hasse V, Frohn M, Metzler D, Bust M, Pryymachuk G, Hescheler J, Grisk O, Chalovich JM, Smyth NR, Pfitzer G, Papadopoulos S. Dual thick and thin filament linked regulation of stretch- and L-NAME-induced tone in young and senescent murine basilar artery. Front Physiol 2023; 14:1099278. [PMID: 37057180 PMCID: PMC10088910 DOI: 10.3389/fphys.2023.1099278] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/09/2023] [Indexed: 03/30/2023] Open
Abstract
Stretch-induced vascular tone is an important element of autoregulatory adaptation of cerebral vasculature to maintain cerebral flow constant despite changes in perfusion pressure. Little is known as to the regulation of tone in senescent basilar arteries. We tested the hypothesis, that thin filament mechanisms in addition to smooth muscle myosin-II regulatory-light-chain-(MLC20)-phosphorylation and non-muscle-myosin-II, contribute to regulation of stretch-induced tone. In young BAs (y-BAs) mechanical stretch does not lead to spontaneous tone generation. Stretch-induced tone in y-BAs appeared only after inhibition of NO-release by L-NAME and was fully prevented by treatment with 3 μmol/L RhoA-kinase (ROK) inhibitor Y27632. L-NAME-induced tone was reduced in y-BAs from heterozygous mice carrying a point mutation of the targeting-subunit of the myosin phosphatase, MYPT1 at threonine696 (MYPT1-T696A/+). In y-BAs, MYPT1-T696A-mutation also blunted the ability of L-NAME to increase MLC20-phosphorylation. In contrast, senescent BAs (s-BAs; >24 months) developed stable spontaneous stretch-induced tone and pharmacological inhibition of NO-release by L-NAME led to an additive effect. In s-BAs the MYPT1-T696A mutation also blunted MLC20-phosphorylation, but did not prevent development of stretch-induced tone. In s-BAs from both lines, Y27632 completely abolished stretch- and L-NAME-induced tone. In s-BAs phosphorylation of non-muscle-myosin-S1943 and PAK1-T423, shown to be down-stream effectors of ROK was also reduced by Y27632 treatment. Stretch- and L-NAME tone were inhibited by inhibition of non-muscle myosin (NM-myosin) by blebbistatin. We also tested whether the substrate of PAK1 the thin-filament associated protein, caldesmon is involved in the regulation of stretch-induced tone in advanced age. BAs obtained from heterozygotes Cald1+/− mice generated stretch-induced tone already at an age of 20–21 months old BAs (o-BA). The magnitude of stretch-induced tone in Cald1+/− o-BAs was similar to that in s-BA. In addition, truncation of caldesmon myosin binding Exon2 (CaD-▵Ex2−/−) did not accelerate stretch-induced tone. Our study indicates that in senescent cerebral vessels, mechanisms distinct from MLC20 phosphorylation contribute to regulation of tone in the absence of a contractile agonist. While in y-and o-BA the canonical pathways, i.e., inhibition of MLCP by ROK and increase in pMLC20, predominate, tone regulation in senescence involves ROK regulated mechanisms, involving non-muscle-myosin and thin filament linked mechanisms involving caldesmon.
Collapse
Affiliation(s)
- Lubomir T. Lubomirov
- Center of Physiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
- Research Cluster, Molecular Mechanisms of Cardiovascular Diseases, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
- *Correspondence: Lubomir T. Lubomirov,
| | - Mechthild M. Schroeter
- Center of Physiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
- Center of Physiology, Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Veronika Hasse
- Center of Physiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Marina Frohn
- Center of Physiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Doris Metzler
- Center of Physiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Maria Bust
- Center of Physiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Galyna Pryymachuk
- Institute of Anatomy, University of Cologne, Cologne, Germany
- Institute of Anatomy, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Jürgen Hescheler
- Center of Physiology, Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Olaf Grisk
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
- Research Cluster, Molecular Mechanisms of Cardiovascular Diseases, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Joseph M. Chalovich
- Department of Biochemistry and Molecular Biology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Neil R. Smyth
- Biological Sciences, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
| | - Gabriele Pfitzer
- Center of Physiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Symeon Papadopoulos
- Center of Physiology, Institute of Neurophysiology, University of Cologne, Cologne, Germany
| |
Collapse
|
6
|
Myosin light chain phosphorylation exhibits a gradient across the wall of cerebellar arteries under sustained ex vivo vascular tone. Sci Rep 2023; 13:909. [PMID: 36650375 PMCID: PMC9845333 DOI: 10.1038/s41598-023-28092-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Small blood vessel diseases are often associated with impaired regulation of vascular tone. The current understanding of resistance arteries often focuses on how a level of vascular tone is achieved in the acute phase, while less emphasis is placed on mechanisms that maintain vascular tone. In this study, cannulated rat superior cerebellar arteries (SCA) developed spontaneous myogenic tone and showed a marked and sustained constriction in the presence of diluted serum (10%), a stimulus relevant to cerebrovascular disease. Both phosphorylated myosin light chain (MLC-p) and smooth muscle alpha actin (SM-α-actin) aligned with phalloidin-stained actin filaments in the vessel wall, while exhibiting a 'high to low' gradient across the layers of vascular smooth muscle cells (VSMC), peaking in the outer layer. The MLC-p distribution profile shifted towards the adventitia in serum treated vessels, while removal of the serum reversed it. Furthermore, a positive correlation between the MLC-p signal and vessel wall tension was also evident. The gradients of phosphorylated MLC and SM-α-actin are consistent with a spatial regulation of the myosin-actin apparatus in the vessel wall during the maintenance of vascular tone. Further, the changing profiles of MLC-p and SM-α-actin are consistent with SCA vasoconstriction being accompanied by VSMC cytoskeletal reorganization.
Collapse
|
7
|
Oburn SM, Huss S, Cox J, Gerthoffer MC, Wu S, Biswas A, Murphy M, Crespi VH, Badding JV, Lopez SA, Elacqua E. Photochemically Mediated Polymerization of Molecular Furan and Pyridine: Synthesis of Nanothreads at Reduced Pressures. J Am Chem Soc 2022; 144:22026-22034. [PMID: 36417898 DOI: 10.1021/jacs.2c09204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Nanothreads are emerging one-dimensional sp3-hybridized materials with high predicted tensile strength and a tunable band gap. They can be synthesized by compressing aromatic or nonaromatic small molecules to pressures ranging from 15-30 GPa. Recently, new avenues are being sought that reduce the pressure required to afford nanothreads; the focus has been placed on the polymerization of molecules with reduced aromaticity, favorable stacking, and/or the use of higher reaction temperatures. Herein, we report the photochemically mediated polymerization of pyridine and furan aromatic precursors, which achieves nanothread formation at reduced pressures. In the case of pyridine, it was found that a combination of slow compression/decompression with broadband UV light exposure yielded a crystalline product featuring a six-fold diffraction pattern with similar interplanar spacings to previously synthesized pyridine-derived nanothreads at a reduced pressure. When furan is compressed to 8 GPa and exposed to broadband UV light, a crystalline solid is recovered that similarly demonstrates X-ray diffraction with an interplanar spacing akin to that of the high-pressure synthesized furan-derived nanothreads. Our method realizes a 1.9-fold reduction in the maximum pressure required to afford furan-derived nanothreads and a 1.4-fold reduction in pressure required for pyridine-derived nanothreads. Density functional theory and multiconfigurational wavefunction-based computations were used to understand the photochemical activation of furan and subsequent cascade thermal cycloadditions. The reduction of the onset pressure is caused by an initial [4+4] cycloaddition followed by increasingly facile thermal [4+2]-cycloadditions during polymerization.
Collapse
Affiliation(s)
- Shalisa M Oburn
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Steven Huss
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Jordan Cox
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Margaret C Gerthoffer
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Sikai Wu
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Arani Biswas
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Morgan Murphy
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Vincent H Crespi
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States.,Department of Physics, The Pennsylvania State University, University Park, Pennsylvania 16802, United States.,Materials Research Institute, The Pennsylvania State University, University Park, Pennsylvania 16802, United States.,Department of Materials Science and Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - John V Badding
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States.,Department of Physics, The Pennsylvania State University, University Park, Pennsylvania 16802, United States.,Materials Research Institute, The Pennsylvania State University, University Park, Pennsylvania 16802, United States.,Department of Materials Science and Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Steven A Lopez
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Elizabeth Elacqua
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
8
|
Cao R, Yang ZS, Hu SL, Liang SJ, Zhang SM, Zhu SQ, Lu L, Long CH, Yao ST, Ma YJ, Liang XH. Molecular Mechanism of Mouse Uterine Smooth Muscle Regulation on Embryo Implantation. Int J Mol Sci 2022; 23:ijms232012494. [PMID: 36293350 PMCID: PMC9604262 DOI: 10.3390/ijms232012494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/01/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Abstract
Myometrium plays critical roles in multiple processes such as embryo spacing through peristalsis during mouse implantation, indicating vital roles of smooth muscle in the successful establishment and quality of implantation. Actin, a key element of cytoskeleton structure, plays an important role in the movement and contraction of smooth muscle cells (SMCs). However, the function of peri-implantation uterine smooth muscle and the regulation mechanism of muscle tension are still unclear. This study focused on the molecular mechanism of actin assembly regulation on implantation in smooth muscle. Phalloidin is a highly selective bicyclic peptide used for staining actin filaments (also known as F-actin). Phalloidin staining showed that F-actin gradually weakened in the CD-1 mouse myometrium from day 1 to day 4 of early pregnancy. More than 3 mice were studied for each group. Jasplakinolide (Jasp) used to inhibit F-actin depolymerization promotes F-actin polymerization in SMCs during implantation window and consequently compromises embryo implantation quality. Transcriptome analysis following Jasp treatment in mouse uterine SMCs reveals significant molecular changes associated with actin assembly. Tagln is involved in the regulation of the cell cytoskeleton and promotes the polymerization of G-actin to F-actin. Our results show that Tagln expression is gradually reduced in mouse uterine myometrium from day 1 to 4 of pregnancy. Furthermore, progesterone inhibits the expression of Tagln through the progesterone receptor. Using siRNA to knock down Tagln in day 3 SMCs, we found that phalloidin staining is decreased, which confirms the critical role of Tagln in F-actin polymerization. In conclusion, our data suggested that decreases in actin assembly in uterine smooth muscle during early pregnancy is critical to optimal embryo implantation. Tagln, a key molecule involved in actin assembly, regulates embryo implantation by controlling F-actin aggregation before implantation, suggesting moderate uterine contractility is conducive to embryo implantation. This study provides new insights into how the mouse uterus increases its flexibility to accommodate implanting embryos in the early stage of pregnancy.
Collapse
|
9
|
Erdener ŞE, Küreli G, Dalkara T. Contractile apparatus in CNS capillary pericytes. NEUROPHOTONICS 2022; 9:021904. [PMID: 35106320 PMCID: PMC8785978 DOI: 10.1117/1.nph.9.2.021904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/22/2021] [Indexed: 06/14/2023]
Abstract
Significance: Whether or not capillary pericytes contribute to blood flow regulation in the brain and retina has long been debated. This was partly caused by failure of detecting the contractile protein α -smooth muscle actin ( α -SMA) in capillary pericytes. Aim: The aim of this review is to summarize recent developments in detecting α -SMA and contractility in capillary pericytes and the relevant literature on the biology of actin filaments. Results: Evidence suggests that for visualization of the small amounts of α -SMA in downstream mid-capillary pericytes, actin depolymerization must be prevented during tissue processing. Actin filaments turnover is mainly based on de/re-polymerization rather than transcription of the monomeric form, hence, small amounts of α -SMA mRNA may evade detection by transcriptomic studies. Similarly, transgenic mice expressing fluorescent reporters under the α -SMA promoter may yield low fluorescence due to limited transcriptional activity in mid-capillary pericytes. Recent studies show that pericytes including mid-capillary ones express several actin isoforms and myosin heavy chain type 11, the partner of α -SMA in mediating contraction. Emerging evidence also suggests that actin polymerization in pericytes may have a role in regulating the tone of downstream capillaries. Conclusions: With guidance of actin biology, innovative labeling and imaging techniques can reveal the molecular machinery of contraction in pericytes.
Collapse
Affiliation(s)
- Şefik E. Erdener
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey
| | - Gülce Küreli
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey
| | - Turgay Dalkara
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey
| |
Collapse
|
10
|
Sancho M, Fletcher J, Welsh DG. Inward Rectifier Potassium Channels: Membrane Lipid-Dependent Mechanosensitive Gates in Brain Vascular Cells. Front Cardiovasc Med 2022; 9:869481. [PMID: 35419431 PMCID: PMC8995785 DOI: 10.3389/fcvm.2022.869481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Cerebral arteries contain two primary and interacting cell types, smooth muscle (SMCs) and endothelial cells (ECs), which are each capable of sensing particular hemodynamic forces to set basal tone and brain perfusion. These biomechanical stimuli help confer tone within arterial networks upon which local neurovascular stimuli function. Tone development is intimately tied to arterial membrane potential (VM) and changes in intracellular [Ca2+] driven by voltage-gated Ca2+ channels (VGCCs). Arterial VM is in turn set by the dynamic interplay among ion channel species, the strongly inward rectifying K+ (Kir) channel being of special interest. Kir2 channels possess a unique biophysical signature in that they strongly rectify, display negative slope conductance, respond to elevated extracellular K+ and are blocked by micromolar Ba2+. While functional Kir2 channels are expressed in both smooth muscle and endothelium, they lack classic regulatory control, thus are often viewed as a simple background conductance. Recent literature has provided new insight, with two membrane lipids, phosphatidylinositol 4,5-bisphosphate (PIP2) and cholesterol, noted to (1) stabilize Kir2 channels in a preferred open or closed state, respectively, and (2) confer, in association with the cytoskeleton, caveolin-1 (Cav1) and syntrophin, hemodynamic sensitivity. It is these aspects of vascular Kir2 channels that will be the primary focus of this review.
Collapse
Affiliation(s)
- Maria Sancho
- Department of Pharmacology, University of Vermont, Burlington, VT, United States
- Department of Physiology, Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- *Correspondence: Maria Sancho,
| | - Jacob Fletcher
- Department of Physiology and Pharmacology, Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Donald G. Welsh
- Department of Physiology and Pharmacology, Robarts Research Institute, University of Western Ontario, London, ON, Canada
- Donald G. Welsh,
| |
Collapse
|
11
|
Brandt MM, Cheng C, Merkus D, Duncker DJ, Sorop O. Mechanobiology of Microvascular Function and Structure in Health and Disease: Focus on the Coronary Circulation. Front Physiol 2022; 12:771960. [PMID: 35002759 PMCID: PMC8733629 DOI: 10.3389/fphys.2021.771960] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022] Open
Abstract
The coronary microvasculature plays a key role in regulating the tight coupling between myocardial perfusion and myocardial oxygen demand across a wide range of cardiac activity. Short-term regulation of coronary blood flow in response to metabolic stimuli is achieved via adjustment of vascular diameter in different segments of the microvasculature in conjunction with mechanical forces eliciting myogenic and flow-mediated vasodilation. In contrast, chronic adjustments in flow regulation also involve microvascular structural modifications, termed remodeling. Vascular remodeling encompasses changes in microvascular diameter and/or density being largely modulated by mechanical forces acting on the endothelium and vascular smooth muscle cells. Whereas in recent years, substantial knowledge has been gathered regarding the molecular mechanisms controlling microvascular tone and how these are altered in various diseases, the structural adaptations in response to pathologic situations are less well understood. In this article, we review the factors involved in coronary microvascular functional and structural alterations in obstructive and non-obstructive coronary artery disease and the molecular mechanisms involved therein with a focus on mechanobiology. Cardiovascular risk factors including metabolic dysregulation, hypercholesterolemia, hypertension and aging have been shown to induce microvascular (endothelial) dysfunction and vascular remodeling. Additionally, alterations in biomechanical forces produced by a coronary artery stenosis are associated with microvascular functional and structural alterations. Future studies should be directed at further unraveling the mechanisms underlying the coronary microvascular functional and structural alterations in disease; a deeper understanding of these mechanisms is critical for the identification of potential new targets for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Maarten M Brandt
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Caroline Cheng
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Walter Brendel Center of Experimental Medicine (WBex), LMU Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Oana Sorop
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
12
|
Effect of Vibration on Alleviating Foot Pressure-Induced Ischemia under Occlusive Compression. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:6208499. [PMID: 34733455 PMCID: PMC8560250 DOI: 10.1155/2021/6208499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 10/08/2021] [Indexed: 12/03/2022]
Abstract
Objectives Foot ulcers often occur in people with diabetes because of pressure-induced tissue ischemia. Vibration has been reported to be helpful in alleviating mechanical damage and promoting wound healing. The objective of this study is to explore whether vibration can relieve reactive hyperemia in foot tissue under occlusive compression. Methods Thirteen healthy adults participated in the study. Each foot was placed under occlusive compression without or with vibration intervention, which was randomly assigned every other day. The dorsal foot skin blood flow (SBF) was measured pre- and postintervention for each subject in each test. Temporal variations and spectral features of SBF were recorded for comparison. Results The results showed that subjects displayed an obvious reactive hyperemia in the foot tissue after pressure occlusion, whereas they displayed a more regular SBF when vibration was applied along with occlusive compression. Moreover, the amplitude of metabolic, neurogenic, and myogenic pathways for SBF was significantly reduced during the hyperemia process when vibration was applied. Conclusions This study demonstrated that vibration can effectively reduce the level of hyperemia in foot tissue under occlusive compression and also induce less protective physiological regulatory activities. This is helpful for protecting foot tissue from pressure-induced ischemic injury and foot ulcers.
Collapse
|
13
|
Cheon S, Tomcho JC, Edwards JM, Bearss NR, Waigi E, Joe B, McCarthy CG, Wenceslau CF. Opioids Cause Sex-Specific Vascular Changes via Cofilin-Extracellular Signal-Regulated Kinase Signaling: Female Mice Present Higher Risk of Developing Morphine-Induced Vascular Dysfunction than Male Mice. J Vasc Res 2021; 58:392-402. [PMID: 34521095 PMCID: PMC8612963 DOI: 10.1159/000517555] [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: 03/17/2021] [Accepted: 06/02/2021] [Indexed: 11/19/2022] Open
Abstract
Recent studies have shown that chronic use of prescription or illicit opioids leads to an increased risk of cardiovascular events and pulmonary arterial hypertension. Indices of vascular age and arterial stiffness are also shown to be increased in opioid-dependent patients, with the effects being more marked in women. There are currently no studies investigating sex-specific vascular dysfunction in opioid use, and the mechanisms leading to opioid-induced vascular damage remain unknown. We hypothesized that exposure to exogenous opioids causes sex-specific vascular remodeling that will be more pronounced in female. Acknowledging the emerging roles of cofilins and extracellular signal-regulated kinases (ERKs) in mediating actin dynamics, we investigated the effects of morphine on these molecules. Twenty-four hour exposure to morphine increased inactivated cofilin and activated ERKs in resistance arteries from female mice, which may promote stress fiber over-assembly. We also performed continuous intraluminal infusion of morphine in pressurized resistance arteries from male and female mice using culture pressure myographs. We observed that morphine reduced the vascular diameter in resistance arteries from female, but not male mice. These results have significant implications for the previously unexplored role of exogenous opioids as a modifiable cardiovascular risk factor, especially in women.
Collapse
MESH Headings
- Actin Depolymerizing Factors/metabolism
- Analgesics, Opioid/toxicity
- Animals
- Cell Proliferation/drug effects
- Cells, Cultured
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Female
- Hemodynamics/drug effects
- Male
- Mesenteric Arteries/drug effects
- Mesenteric Arteries/enzymology
- Mesenteric Arteries/pathology
- Mesenteric Arteries/physiopathology
- Mice, Inbred C57BL
- Morphine/toxicity
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phosphorylation
- Rats, Sprague-Dawley
- Sex Factors
- Signal Transduction
- Vascular Remodeling/drug effects
- Mice
- Rats
Collapse
Affiliation(s)
- Soyoung Cheon
- Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, Ohio, USA
| | - Jeremy C Tomcho
- Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, Ohio, USA
| | - Jonnelle M Edwards
- Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, Ohio, USA
| | - Nicole R Bearss
- Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, Ohio, USA
| | - Emily Waigi
- Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, Ohio, USA
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Bina Joe
- Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, Ohio, USA
| | - Cameron G McCarthy
- Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, Ohio, USA
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Camilla F Wenceslau
- Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, Ohio, USA
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| |
Collapse
|
14
|
Wang Y, Liao G, Wang R, Tang DD. Acetylation of Abelson interactor 1 at K416 regulates actin cytoskeleton and smooth muscle contraction. FASEB J 2021; 35:e21811. [PMID: 34369620 PMCID: PMC8800440 DOI: 10.1096/fj.202100415r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/28/2021] [Accepted: 07/07/2021] [Indexed: 11/21/2022]
Abstract
Actin cytoskeletal reorganization plays an important role in regulating smooth muscle contraction, which is essential for the modulation of various physiological functions including airway tone. The adapter protein Abi1 (Abelson interactor 1) participates in the control of smooth muscle contraction. The mechanisms by which Abi1 coordinates smooth muscle function are not fully understood. Here, we found that contractile stimulation elicited Abi1 acetylation in human airway smooth muscle (HASM) cells. Mutagenesis analysis identified lysine‐416 (K416) as a major acetylation site. Replacement of K416 with Q (glutamine) enhanced the interaction of Abi1 with neuronal Wiskott‐Aldrich syndrome protein (N‐WASP), an important actin‐regulatory protein. Moreover, the expression of K416Q Abi1 promoted actin polymerization and smooth muscle contraction without affecting myosin light chain phosphorylation at Ser‐19 and vimentin phosphorylation at Ser‐56. Furthermore, p300 is a lysine acetyltransferase that catalyzes acetylation of histone and non‐histone proteins in various cell types. Here, we discovered that a portion of p300 was localized in the cytoplasm of HASM cells. Knockdown of p300 reduced the agonist‐induced Abi1 acetylation in HASM cells and in mouse airway smooth muscle tissues. Smooth muscle conditional knockout of p300 inhibited actin polymerization and the contraction of airway smooth muscle tissues without affecting myosin light chain phosphorylation and vimentin phosphorylation. Together, our results suggest that contractile stimulation induces Abi1 acetylation via p300 in smooth muscle. Acetylation at K416 promotes the coupling of Abi1 with N‐WASP, which facilitates actin polymerization and smooth muscle contraction. This is a novel acetylation‐dependent regulation of the actin cytoskeleton in smooth muscle.
Collapse
Affiliation(s)
- Yinna Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Guoning Liao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Ruping Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Dale D Tang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| |
Collapse
|
15
|
Lee SG, Lee SN, Baek J, Yoon JH, Lee H. Mechanical compression enhances ciliary beating through cytoskeleton remodeling in human nasal epithelial cells. Acta Biomater 2021; 128:346-356. [PMID: 33882353 DOI: 10.1016/j.actbio.2021.04.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 03/27/2021] [Accepted: 04/14/2021] [Indexed: 01/25/2023]
Abstract
Nasal inflammatory diseases, including nasal polyps and acute/chronic sinusitis, are characterized by impaired mucociliary clearance and eventually inflammation and infection. Contact of nasal polyps with adjacent nasal mucosa or stagnated mucus within the maxillary sinus produces compressive mechanical stresses on the apical surface of epithelium which can induce cytoskeleton remodeling in epithelial cells. In this study, we hypothesized that compressive stress modulates ciliary beating by altering the mechanical properties of the cytoskeleton of ciliated cell basal bodies. For the primary human nasal epithelial cells, we found that the applied compressive stress higher than the critical value of 1.0 kPa increased the stroke speed of cilia leading to the enhancement of ciliary beating frequency and mucociliary transportability. Immunostained images of the cytoskeleton showed reorganization and compactness of the actin filaments in the presence of compressive stress. Analysis of beating trajectory with the computational modeling for ciliary beating revealed that the stroke speed of cilium increased as the relative elasticity to viscosity of the surrounding cytoskeleton increases. These results suggest that the compressive stress on epithelial cells increases the ciliary beating speed through cytoskeleton remodeling to prevent mucus stagnation at the early stage of airway obstruction. Our study provides an insight into the defensive mechanism of airway epithelium against pathological conditions. STATEMENT OF SIGNIFICANCE: Cilia dynamics of the nasal epithelium is critical for not only maintaining normal breathing but preventing inflammatory diseases. It has been shown that mechanical compressive stresses can alter the shape and phenotype of epithelial cells. However, the effect of compressive stress on cilia dynamics is unclear. In this study, we demonstrated that the oscillation speed of cilia in human nasal epithelial cells was increased by the applied compressive stress experimentally. The computational simulation revealed that the change of ciliary beating dynamics was attributed to the viscoelastic properties of the reorganized cytoskeleton in response to compressive stress. Our results will be beneficial in understanding the defensive mechanism of airway epithelium against pathological conditions.
Collapse
|
16
|
Szenasi A, Amrein K, Czeiter E, Szarka N, Toth P, Koller A. Molecular Pathomechanisms of Impaired Flow-Induced Constriction of Cerebral Arteries Following Traumatic Brain Injury: A Potential Impact on Cerebral Autoregulation. Int J Mol Sci 2021; 22:6624. [PMID: 34205652 PMCID: PMC8234931 DOI: 10.3390/ijms22126624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 01/21/2023] Open
Abstract
(1) Background: Traumatic brain injury (TBI) frequently occurs worldwide, resulting in high morbidity and mortality. Here, we hypothesized that TBI impairs an autoregulatory mechanism, namely the flow-induced constriction of isolated rat middle cerebral arteries (MCAs). (2) Methods: TBI was induced in anaesthetized rats by weight drop model, and then MCAs were isolated and transferred into a pressure-flow chamber. The internal diameter was measured by a video-microscopy. (3) Results: In MCAs from intact rats, increases in flow and pressure + flow elicited constrictions (-26 ± 1.9 µm and -52 ± 2.8 µm, p < 0.05), which were significantly reduced after TBI or in the presence of thromboxane-prostanoid (TP receptor) antagonist SQ 29,548. Flow-induced constrictions were significantly reduced by HET0016, inhibitor of cytochrome P450 4A (CYP450 4A). Arachidonic acid, (AA, 10-7 M), and CYP-450 4A metabolite 20-hydroxyeicosatetraenoic acid (20-HETE) elicited constrictions of intact MCA (-26 ± 2.3% and -31 ± 3.6%), which were significantly reduced after TBI (to 11 ± 1.3% and -16 ±2.5%). The TP receptor agonist U46619 (10-7 M) elicited substantial constrictions of MCA from intact rats (-21 ± 3.3%), which were also significantly reduced, after TBI (to -16 ± 2.4%). (4) Conclusions: Flow-induced constrictor response of MCA is impaired by traumatic brain injury, likely due to the reduced ability of cytochrome P450 4A to convert arachidonic acid to constrictor prostaglandins and the mitigated sensitivity of thromboxane-prostanoid receptors.
Collapse
Affiliation(s)
- Annamaria Szenasi
- Department of Translational Medicine, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary;
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 1085 Budapest, Hungary
| | - Krisztina Amrein
- Department of Neurosurgery and Szentágothai Research Center, University of Pecs, Medical School, 7623 Pecs, Hungary; (K.A.); (E.C.); (P.T.)
| | - Endre Czeiter
- Department of Neurosurgery and Szentágothai Research Center, University of Pecs, Medical School, 7623 Pecs, Hungary; (K.A.); (E.C.); (P.T.)
| | - Nikolett Szarka
- MTA-PTE Clinical Neuroscience MR Research Group, 7623 Pecs, Hungary;
| | - Peter Toth
- Department of Neurosurgery and Szentágothai Research Center, University of Pecs, Medical School, 7623 Pecs, Hungary; (K.A.); (E.C.); (P.T.)
- MTA-PTE Clinical Neuroscience MR Research Group, 7623 Pecs, Hungary;
| | - Akos Koller
- Department of Translational Medicine, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary;
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 1085 Budapest, Hungary
- Research Center for Sports Physiology, University of Physical Education, 1123 Budapest, Hungary
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|
17
|
Brain capillary pericytes exert a substantial but slow influence on blood flow. Nat Neurosci 2021; 24:633-645. [PMID: 33603231 PMCID: PMC8102366 DOI: 10.1038/s41593-020-00793-2] [Citation(s) in RCA: 197] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 12/23/2020] [Indexed: 01/30/2023]
Abstract
The majority of the brain's vasculature is composed of intricate capillary networks lined by capillary pericytes. However, it remains unclear whether capillary pericytes influence blood flow. Using two-photon microscopy to observe and manipulate brain capillary pericytes in vivo, we find that their optogenetic stimulation decreases lumen diameter and blood flow, but with slower kinetics than similar stimulation of mural cells on upstream pial and precapillary arterioles. This slow vasoconstriction was inhibited by the clinically used vasodilator fasudil, a Rho-kinase inhibitor that blocks contractile machinery. Capillary pericytes were also slower to constrict back to baseline following hypercapnia-induced dilation, and slower to dilate towards baseline following optogenetically induced vasoconstriction. Optical ablation of single capillary pericytes led to sustained local dilation and a doubling of blood cell flux selectively in capillaries lacking pericyte contact. These data indicate that capillary pericytes contribute to basal blood flow resistance and slow modulation of blood flow throughout the brain.
Collapse
|
18
|
Edwards JM, Roy S, Galla SL, Tomcho JC, Bearss NR, Waigi EW, Mell B, Cheng X, Saha P, Vijay-Kumar M, McCarthy CG, Joe B, Wenceslau CF. FPR-1 (Formyl Peptide Receptor-1) Activation Promotes Spontaneous, Premature Hypertension in Dahl Salt-Sensitive Rats. Hypertension 2021; 77:1191-1202. [PMID: 33641367 DOI: 10.1161/hypertensionaha.120.16237] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jonnelle M Edwards
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Shaunak Roy
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Sarah L Galla
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Jeremy C Tomcho
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Nicole R Bearss
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Emily W Waigi
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Blair Mell
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Xi Cheng
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Piu Saha
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Matam Vijay-Kumar
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Cameron G McCarthy
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Bina Joe
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Camilla F Wenceslau
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| |
Collapse
|
19
|
Alarcon-Martinez L, Yemisci M, Dalkara T. Pericyte morphology and function. Histol Histopathol 2021; 36:633-643. [PMID: 33595091 DOI: 10.14670/hh-18-314] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The proper delivery of blood is essential for healthy neuronal function. The anatomical substrate for this precise mechanism is the neurovascular unit, which is formed by neurons, glial cells, endothelia, smooth muscle cells, and pericytes. Based on their particular location on the vessel wall, morphology, and protein expression, pericytes have been proposed as cells capable of regulating capillary blood flow. Pericytes are located around the microvessels, wrapping them with their processes. Their morphology and protein expression substantially vary along the vascular tree. Their contractibility is mediated by a unique cytoskeleton organization formed by filaments of actin that allows pericyte deformability with the consequent mechanical force transferred to the extracellular matrix for changing the diameter. Pericyte ultrastructure is characterized by large mitochondria likely to provide energy to regulate intracellular calcium concentration and fuel contraction. Accordingly, pericytes with compromised energy show a sustained intracellular calcium increase that leads to persistent microvascular constriction. Pericyte morphology is highly plastic and adapted for varying contractile capability along the microvascular tree, making pericytes ideal cells to regulate the capillary blood flow in response to local neuronal activity. Besides the vascular regulation, pericytes also play a role in the maintenance of the blood-brain/retina barrier, neovascularization and angiogenesis, and leukocyte transmigration. Here, we review the morphological and functional features of the pericytes as well as potential specific markers for the study of pericytes in the brain and retina.
Collapse
Affiliation(s)
- Luis Alarcon-Martinez
- Department of Neuroscience and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Université de Montréal, Montréal, QC, Canada.
| | - Muge Yemisci
- Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, Turkey.,Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| |
Collapse
|
20
|
Valisno JAC, May J, Singh K, Helm EY, Venegas L, Budbazar E, Goodman JB, Nicholson CJ, Avram D, Cohen RA, Mitchell GF, Morgan KG, Seta F. BCL11B Regulates Arterial Stiffness and Related Target Organ Damage. Circ Res 2021; 128:755-768. [PMID: 33530702 PMCID: PMC7969164 DOI: 10.1161/circresaha.120.316666] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Supplemental Digital Content is available in the text. BCL11B (B-cell leukemia 11b) is a transcription factor known as an essential regulator of T lymphocytes and neuronal development during embryogenesis. A genome-wide association study showed that a gene desert region downstream of BCL11B, known to function as a BCL11B enhancer, harbors single nucleotide polymorphisms associated with increased arterial stiffness. However, a role for BCL11B in the adult cardiovascular system is unknown.
Collapse
Affiliation(s)
- Jeff Arni C Valisno
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, MA (J.A.C.V., J.M., L.V., E.B., J.B.G., R.A.C., F.S.)
| | - Joel May
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, MA (J.A.C.V., J.M., L.V., E.B., J.B.G., R.A.C., F.S.)
| | - Kuldeep Singh
- Department of Health Sciences, Sargent College, Boston University, MA (K.S., C.J.N., K.G.M.)
| | - Eric Y Helm
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville (E.Y.H., D.A.)
| | - Lisia Venegas
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, MA (J.A.C.V., J.M., L.V., E.B., J.B.G., R.A.C., F.S.)
| | - Enkhjargal Budbazar
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, MA (J.A.C.V., J.M., L.V., E.B., J.B.G., R.A.C., F.S.)
| | - Jena B Goodman
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, MA (J.A.C.V., J.M., L.V., E.B., J.B.G., R.A.C., F.S.)
| | - Christopher J Nicholson
- Department of Health Sciences, Sargent College, Boston University, MA (K.S., C.J.N., K.G.M.)
| | - Dorina Avram
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville (E.Y.H., D.A.).,Department of Immunology, Moffitt Cancer Center, Tampa, FL (D.A.)
| | - Richard A Cohen
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, MA (J.A.C.V., J.M., L.V., E.B., J.B.G., R.A.C., F.S.)
| | | | - Kathleen G Morgan
- Department of Health Sciences, Sargent College, Boston University, MA (K.S., C.J.N., K.G.M.)
| | - Francesca Seta
- Vascular Biology Section, Department of Medicine, Boston University School of Medicine, MA (J.A.C.V., J.M., L.V., E.B., J.B.G., R.A.C., F.S.)
| |
Collapse
|
21
|
Kureli G, Yilmaz-Ozcan S, Erdener SE, Donmez-Demir B, Yemisci M, Karatas H, Dalkara T. F-actin polymerization contributes to pericyte contractility in retinal capillaries. Exp Neurol 2020; 332:113392. [DOI: 10.1016/j.expneurol.2020.113392] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/15/2020] [Accepted: 06/25/2020] [Indexed: 01/24/2023]
|
22
|
Enomoto U, Imashiro C, Takemura K. Collective cell migration of fibroblasts is affected by horizontal vibration of the cell culture dish. Eng Life Sci 2020; 20:402-411. [PMID: 32944015 PMCID: PMC7481772 DOI: 10.1002/elsc.202000013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/25/2020] [Accepted: 06/18/2020] [Indexed: 01/08/2023] Open
Abstract
Regulating the collective migration of cells is an important issue in bioengineering. Enhancing or suppressing cell migration and controlling the migration direction is useful for various physiological phenomena such as wound healing. Several methods of migration regulation based on different mechanical stimuli have been reported. While vibrational stimuli, such as sound waves, show promise for regulating migration, the effect of the vibration direction on collective cell migration has not been studied in depth. Therefore, we fabricated a vibrating system that can apply horizontal vibration to a cell culture dish. Here, we evaluated the effect of the vibration direction on the collective migration of fibroblasts in a wound model comprising two culture areas separated by a gap. Results showed that the vibration direction affects the cell migration distance: vibration orthogonal to the gap enhances the collective cell migration distance while vibration parallel to the gap suppresses it. Results also showed that conditions leading to enhanced migration distance were also associated with elevated glucose consumption. Furthermore, under conditions promoting cell migration, the cell nuclei become elongated and oriented orthogonal to the gap. In contrast, under conditions that reduce the migration distance, cell nuclei were oriented to the direction parallel to the gap.
Collapse
Affiliation(s)
- Umi Enomoto
- School of Science for Open and Environmental SystemsGraduate School of Science and TechnologyKeio UniversityYokohamaKanagawaJapan
| | - Chikahiro Imashiro
- Department of Mechanical EngineeringKeio UniversityYokohamaKanagawaJapan
- Institute of Advanced Biomedical Engineering and ScienceTokyo Women's Medical UniversityTokyoJapan
| | - Kenjiro Takemura
- Department of Mechanical EngineeringKeio UniversityYokohamaKanagawaJapan
| |
Collapse
|
23
|
Hashmi SK, Barka V, Yang C, Schneider S, Svitkina TM, Heuckeroth RO. Pseudo-obstruction-inducing ACTG2R257C alters actin organization and function. JCI Insight 2020; 5:140604. [PMID: 32814715 PMCID: PMC7455133 DOI: 10.1172/jci.insight.140604] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/15/2020] [Indexed: 12/14/2022] Open
Abstract
Actin γ 2, smooth muscle (ACTG2) R257C mutation is the most common genetic cause of visceral myopathy. Individuals with ACTG2 mutations endure prolonged hospitalizations and surgical interventions, become dependent on intravenous nutrition and bladder catheterization, and often die in childhood. Currently, we understand little about how ACTG2 mutations cause disease, and there are no mechanism-based treatments. Our goal was to characterize the effects of ACTG2R257C on actin organization and function in visceral smooth muscle cells. We overexpressed ACTG2WT or ACTG2R257C in primary human intestinal smooth muscle cells (HISMCs) and performed detailed quantitative analyses to examine effects of ACTG2R257C on (a) actin filament formation and subcellular localization, (b) actin-dependent HISMC functions, and (c) smooth muscle contractile gene expression. ACTG2R257C resulted in 41% fewer, 13% thinner, 33% shorter, and 40% less branched ACTG2 filament bundles compared with ACTG2WT. Curiously, total F-actin probed by phalloidin and a pan-actin antibody was unchanged between ACTG2WT- and ACTG2R257C-expressing HISMCs, as was ultrastructural F-actin organization. ACTG2R257C-expressing HISMCs contracted collagen gels similar to ACTG2WT-expressing HISMCs but spread 21% more and were 11% more migratory. In conclusion, ACTG2R257C profoundly affects ACTG2 filament bundle structure, without altering global actin cytoskeleton in HISMCs.
Collapse
Affiliation(s)
- Sohaib Khalid Hashmi
- Department of Pediatrics, Children's Hospital of Philadelphia Research Institute, and Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, Pennsylvania, USA.,Department of Bioengineering, University of Pennsylvania School of Engineering and Applied Science, Philadelphia, Pennsylvania, USA
| | - Vasia Barka
- Department of Pediatrics, Children's Hospital of Philadelphia Research Institute, and Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, Pennsylvania, USA
| | - Changsong Yang
- Department of Biology, University of Pennsylvania School of Arts and Sciences, Philadelphia, Pennsylvania, USA
| | - Sabine Schneider
- Department of Pediatrics, Children's Hospital of Philadelphia Research Institute, and Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, Pennsylvania, USA
| | - Tatyana M Svitkina
- Department of Biology, University of Pennsylvania School of Arts and Sciences, Philadelphia, Pennsylvania, USA
| | - Robert O Heuckeroth
- Department of Pediatrics, Children's Hospital of Philadelphia Research Institute, and Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
24
|
Wang S, Lv W, Zhang H, Liu Y, Li L, Jefferson JR, Guo Y, Li M, Gao W, Fang X, Paul IA, Rajkowska G, Shaffery JP, Mosley TH, Hu X, Liu R, Wang Y, Yu H, Roman RJ, Fan F. Aging exacerbates impairments of cerebral blood flow autoregulation and cognition in diabetic rats. GeroScience 2020; 42:1387-1410. [PMID: 32696219 DOI: 10.1007/s11357-020-00233-w] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus (DM) is a leading risk factor for aging-related dementia; however, the underlying mechanisms are not well understood. The present study, utilizing a non-obese T2DN diabetic model, demonstrates that the myogenic response of the middle cerebral artery (MCA) and parenchymal arteriole (PA) and autoregulation of cerebral blood flow (CBF) in the surface and deep cortex were impaired at both young and old ages. The impaired CBF autoregulation was more severe in old than young DM rats, and in the deep than the surface cortex. The myogenic tone of the MCA was enhanced at perfusion pressure in the range of 40-100 mmHg in young DM rats but was reduced at 140-180 mmHg in old DM rats. No change of the myogenic tone of the PA was observed in young DM rats, whereas it was significantly reduced at 30-60 mmHg in old DM rats. Old DM rats had enhanced blood-brain barrier (BBB) leakage and neurodegeneration, reduced vascular density, tight junction, and pericyte coverage on cerebral capillaries in the CA3 region in the hippocampus. Additionally, DM rats displayed impaired functional hyperemia and spatial learning and short- and long-term memory at both young and old ages. Old DM rats had impaired non-spatial short-term memory. These results revealed that impaired CBF autoregulation and enhanced BBB leakage plays an essential role in the pathogenesis of age- and diabetes-related dementia. These findings will lay the foundations for the discovery of anti-diabetic therapies targeting restoring CBF autoregulation to prevent the onset and progression of dementia in elderly DM.
Collapse
Affiliation(s)
- Shaoxun Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Wenshan Lv
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.,Department of Endocrinology and Metabolic, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Huawei Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Yedan Liu
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Longyang Li
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Joshua R Jefferson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Ya Guo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Man Li
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Wenjun Gao
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Xing Fang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Ian A Paul
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Grazyna Rajkowska
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - James P Shaffery
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Thomas H Mosley
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, 39216, USA.,Department of Medicine (Geriatrics), University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Xinlin Hu
- Department of Endocrinology and Metabolic, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Ruen Liu
- Department of Neurosurgery, Peking University People's Hospital, Beijing, 100044, China
| | - Yangang Wang
- Department of Endocrinology and Metabolic, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
25
|
Wang Y, Wang R, Tang DD. Ste20-like Kinase-mediated Control of Actin Polymerization Is a New Mechanism for Thin Filament-associated Regulation of Airway Smooth Muscle Contraction. Am J Respir Cell Mol Biol 2020; 62:645-656. [PMID: 31913659 PMCID: PMC7193783 DOI: 10.1165/rcmb.2019-0310oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 01/07/2020] [Indexed: 12/26/2022] Open
Abstract
It has been reported that actin polymerization is regulated by protein tyrosine phosphorylation in smooth muscle on contractile stimulation. The role of protein serine/threonine phosphorylation in modulating actin dynamics is underinvestigated. SLK (Ste20-like kinase) is a serine/threonine protein kinase that plays a role in apoptosis, cell cycle, proliferation, and migration. The function of SLK in smooth muscle is mostly unknown. Here, SLK knockdown (KD) inhibited acetylcholine (ACh)-induced actin polymerization and contraction without affecting myosin light chain phosphorylation at Ser-19 in human airway smooth muscle. Stimulation with ACh induced paxillin phosphorylation at Ser-272, which was reduced in SLK KD cells. However, SLK did not catalyze paxillin Ser-272 phosphorylation in vitro. But, SLK KD attenuated Plk1 (polo-like kinase 1) phosphorylation at Thr-210. Plk1 mediated paxillin phosphorylation at Ser-272 in vitro. Expression of the nonphosphorylatable paxillin mutant S272A (substitution of alanine at Ser-272) attenuated the agonist-enhanced F-actin/G-actin ratios without affecting myosin light chain phosphorylation. Because N-WASP (neuronal Wiskott-Aldrich Syndrome Protein) phosphorylation at Tyr-256 (an indication of its activation) promotes actin polymerization, we also assessed the role of paxillin phosphorylation in N-WASP activation. S272A paxillin inhibited the ACh-enhanced N-WASP phosphorylation at Tyr-256. Together, these results suggest that SLK regulates paxillin phosphorylation at Ser-272 via Plk1, which modulates N-WASP activation and actin polymerization in smooth muscle. SLK-mediated actin cytoskeletal reorganization may facilitate force transmission between the contractile units and the extracellular matrix.
Collapse
Affiliation(s)
- Yinna Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Ruping Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Dale D Tang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| |
Collapse
|
26
|
Accelerated cerebral vascular injury in diabetes is associated with vascular smooth muscle cell dysfunction. GeroScience 2020; 42:547-561. [PMID: 32166556 DOI: 10.1007/s11357-020-00179-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/04/2020] [Indexed: 12/20/2022] Open
Abstract
Individuals with diabetes are more susceptible to cerebral vascular aging. However, the underlying mechanisms are not well elucidated. The present study examined whether the myogenic response of the middle cerebral artery (MCA) is impaired in diabetic rats due to high glucose (HG)-induced cerebral vascular smooth muscle cell (CVSMC) dysfunction, and whether this is associated with ATP depletion and changes in mitochondrial dynamics and membrane potential. The diameters of the MCA of diabetic rats increased to 135.3 ± 11.3% when perfusion pressure was increased from 40 to 180 mmHg, while it fell to 85.1 ± 3.1% in non-diabetic controls. The production of ROS and mitochondrial-derived superoxide were enhanced in cerebral arteries of diabetic rats. Levels of mitochondrial superoxide were significantly elevated in HG-treated primary CVSMCs, which was associated with decreased ATP production, mitochondrial respiration, and membrane potential. The expression of OPA1 was reduced, and MFF was elevated in HG-treated CVSMCs in association with fragmented mitochondria. Moreover, HG-treated CVSMCs displayed lower contractile and proliferation capabilities. These results demonstrate that imbalanced mitochondrial dynamics (increased fission and decreased fusion) and membrane depolarization contribute to ATP depletion in HG-treated CVSMCs, which promotes CVSMC dysfunction and may play an essential role in exacerbating the impaired myogenic response in the cerebral circulation in diabetes and accelerating vascular aging.
Collapse
|
27
|
Zhang L, Wu JH, Huang TQ, Nepliouev I, Brian L, Zhang Z, Wertman V, Rudemiller NP, McMahon TJ, Shenoy SK, Miller FJ, Crowley SD, Freedman NJ, Stiber JA. Drebrin regulates angiotensin II-induced aortic remodelling. Cardiovasc Res 2019; 114:1806-1815. [PMID: 29931051 DOI: 10.1093/cvr/cvy151] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 06/14/2018] [Indexed: 01/07/2023] Open
Abstract
Aims The actin-binding protein Drebrin is up-regulated in response to arterial injury and reduces smooth muscle cell (SMC) migration and proliferation through its interaction with the actin cytoskeleton. We, therefore, tested the hypothesis that SMC Drebrin inhibits angiotensin II-induced remodelling of the proximal aorta. Methods and results Angiotensin II was administered via osmotic minipumps at 1000 ng/kg/min continuously for 28 days in SM22-Cre+/Dbnflox/flox (SMC-Dbn-/-) and control mice. Blood pressure responses to angiotensin II were assessed by telemetry. After angiotensin II infusion, we assessed remodelling in the proximal ascending aorta by echocardiography and planimetry of histological cross sections. Although the degree of hypertension was equivalent in SMC-Dbn-/- and control mice, SMC-Dbn-/- mice nonetheless exhibited 60% more proximal aortic medial thickening and two-fold more outward aortic remodelling than control mice in response to angiotensin II. Proximal aortas demonstrated greater cellular proliferation and matrix deposition in SMC-Dbn-/- mice than in control mice, as evidenced by a higher prevalence of proliferating cell nuclear antigen-positive nuclei and higher levels of collagen I. Compared with control mouse aortas, SMC-Dbn-/- aortas demonstrated greater angiotensin II-induced NADPH oxidase activation and inflammation, evidenced by higher levels of Ser-536-phosphorylated NFκB p65 subunits and higher levels of vascular cell adhesion molecule-1, matrix metalloproteinase-9, and adventitial macrophages. Conclusions We conclude that SMC Drebrin deficiency augments angiotensin II-induced inflammation and adverse aortic remodelling.
Collapse
Affiliation(s)
- Lisheng Zhang
- Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, USA
| | - Jiao-Hui Wu
- Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, USA
| | - Tai-Qin Huang
- Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, USA
| | - Igor Nepliouev
- Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, USA
| | - Leigh Brian
- Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, USA
| | - Zhushan Zhang
- Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, USA
| | - Virginia Wertman
- Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, USA
| | - Nathan P Rudemiller
- Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, USA
| | - Timothy J McMahon
- Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, USA
| | - Sudha K Shenoy
- Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, USA
| | - Francis J Miller
- Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, USA
| | - Steven D Crowley
- Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, USA
| | - Neil J Freedman
- Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, USA
| | - Jonathan A Stiber
- Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, USA
| |
Collapse
|
28
|
Zhang W, Gunst SJ. Molecular Mechanisms for the Mechanical Modulation of Airway Responsiveness. ACTA ACUST UNITED AC 2019; 2. [PMID: 32270135 PMCID: PMC7141576 DOI: 10.1115/1.4042775] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The smooth muscle of the airways is exposed to continuously changing mechanical
forces during normal breathing. The mechanical oscillations that occur during
breathing have profound effects on airway tone and airway responsiveness both in
experimental animals and humans in vivo and in isolated airway tissues in vitro.
Experimental evidence suggests that alterations in the contractile and
mechanical properties of airway smooth muscle tissues caused by mechanical
perturbations result from adaptive changes in the organization of the
cytoskeletal architecture of the smooth muscle cell. The cytoskeleton is a
dynamic structure that undergoes rapid reorganization in response to external
mechanical and pharmacologic stimuli. Contractile stimulation initiates the
assembly of cytoskeletal/extracellular matrix adhesion complex proteins into
large macromolecular signaling complexes (adhesomes) that undergo activation to
mediate the polymerization and reorganization of a submembranous network of
actin filaments at the cortex of the cell. Cortical actin polymerization is
catalyzed by Neuronal-Wiskott–Aldrich syndrome protein (N-WASP) and the
Arp2/3 complex, which are activated by pathways regulated by paxillin and the
small GTPase, cdc42. These processes create a strong and rigid cytoskeletal
framework that may serve to strengthen the membrane for the transmission of
force generated by the contractile apparatus to the extracellular matrix, and to
enable the adaptation of smooth muscle cells to mechanical stresses. This model
for the regulation of airway smooth muscle function can provide novel
perspectives to explain the normal physiologic behavior of the airways and
pathophysiologic properties of the airways in asthma.
Collapse
Affiliation(s)
- Wenwu Zhang
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Susan J Gunst
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
29
|
Mahavadi S, Grider JR, Murthy KS. Muscarinic m2 receptor-mediated actin polymerization via PI3 kinase γ and integrin-linked kinase in gastric smooth muscle. Neurogastroenterol Motil 2019; 31:e13495. [PMID: 30393912 PMCID: PMC6347515 DOI: 10.1111/nmo.13495] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/07/2018] [Accepted: 09/25/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Actin polymerization plays an important role in smooth muscle contraction. Integrin-linked kinase (ILK) was shown to mediate actin polymerization in airway smooth muscle. The role of ILK in actin polymerization in response to m2 receptor activation was not in gastric smooth muscle. METHODS Phosphorylation of paxillin, neuronal Wiskott-Aldrich syndrome protein (N-WASp), and association of paxillin with GEF proteins (Cool2/αPix [Cool2/PAK-interacting exchange factor alpha], Cool1/βPix [Cool1/PAK-interacting exchange factor beta], and DOCK 180 [Dedicator of cytokinesis]) and N-WASp with Arp2/3 complex were measured by western blot. Activation of Cdc42 was determined using an antibody for activated Cdc42. Actin polymerization was measured as an increase in F-actin/G-actin ratio. RESULTS Phosphorylation of paxillin, an association of paxillin with GEF proteins, Cdc42 activity, and actin polymerization were increased in response to m2 receptor activation in gastric smooth muscle cells. The increases in paxillin phosphorylation, Cdc42 activity, and actin polymerization were inhibited by a PI3Kγ inhibitor (AS-605240), ILK siRNA, and ILK dominant negative mutant (ILK [R211]). Increase in actin polymerization was also inhibited by Cdc42 dominant negative mutant (Cdc42 [T17N]). Increases in the association of paxillin with GEF proteins, phosphorylation of N-WASp and its association with Arp2/3 complex were inhibited by ILK (R211). CONCLUSION In gastric smooth muscle cells, activation of PI3Kγ by muscarinic m2 receptors causes ILK-dependent phosphorylation of paxillin, an association of paxillin with Cdc42 GEF proteins and activation of Cdc42, which, in turn, causes phosphorylation of N-WASp and its association with Arp2/3 complex leading to actin polymerization.
Collapse
Affiliation(s)
- Sunila Mahavadi
- Department of Physiology and Biophysics VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University Richmond Virginia
| | - John R. Grider
- Department of Physiology and Biophysics VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University Richmond Virginia
| | - Karnam S. Murthy
- Department of Physiology and Biophysics VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University Richmond Virginia
| |
Collapse
|
30
|
Wenceslau CF, McCarthy CG, Szasz T, Calmasini FB, Mamenko M, Webb RC. Formyl peptide receptor-1 activation exerts a critical role for the dynamic plasticity of arteries via actin polymerization. Pharmacol Res 2019; 141:276-290. [PMID: 30639374 DOI: 10.1016/j.phrs.2019.01.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 12/20/2022]
Abstract
Several human diseases, include cancer and stroke are characterized by changes in immune system activation and vascular contractility. However, the mechanistic foundation of a vascular immuno-physiology network is still largely unknown. Formyl peptide receptor-1 (FPR-1), which plays a vital role in the function of the innate immune system, is widely expressed in arteries, but its role in vascular plasticity is unclear. We questioned why a receptor that is crucial for immune defense, and cell motility in leukocytes, would be expressed in vascular smooth muscle cells (VSMCs). We hypothesized that activation of FPR-1 in arteries is important for the temporal reorganization of actin filaments, and consequently, changes in vascular function, similar to what is observed in neutrophils. To address our hypothesis, we used FPR-1 knockout and VSMCs lacking FPR-1. We observed that FPR-1 activation induces actin polymerization in wild type VSMCs. Absence of FPR-1 in the vasculature significantly decreased vascular contraction and induced loss of myogenic tone to elevated intraluminal pressures via disruption of actin polymerization. Actin polymerization activator ameliorated these responses. In conclusion, we have established a novel role for FPR-1 in VSMC contractility and motility, similar to the one observed in sentinel cells of the innate immune system. This discovery is fundamental for vascular immuno-pathophysiology, given that FPR-1 in VSMCs not only functions as an immune system receptor, but it also has an important role for the dynamic plasticity of arteries.
Collapse
Affiliation(s)
- Camilla F Wenceslau
- Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA; Department of Physiology, Augusta University, Augusta, GA, USA.
| | - Cameron G McCarthy
- Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA; Department of Physiology, Augusta University, Augusta, GA, USA
| | - Theodora Szasz
- Department of Physiology, Augusta University, Augusta, GA, USA
| | | | - Mykola Mamenko
- Department of Physiology, Augusta University, Augusta, GA, USA
| | - R Clinton Webb
- Department of Physiology, Augusta University, Augusta, GA, USA
| |
Collapse
|
31
|
Martinez-Quinones P, McCarthy CG, Watts SW, Klee NS, Komic A, Calmasini FB, Priviero F, Warner A, Chenghao Y, Wenceslau CF. Hypertension Induced Morphological and Physiological Changes in Cells of the Arterial Wall. Am J Hypertens 2018; 31:1067-1078. [PMID: 29788246 DOI: 10.1093/ajh/hpy083] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 05/14/2018] [Indexed: 12/17/2022] Open
Abstract
Morphological and physiological changes in the vasculature have been described in the evolution and maintenance of hypertension. Hypertension-induced vascular dysfunction may present itself as a contributing, or consequential factor, to vascular remodeling caused by chronically elevated systemic arterial blood pressure. Changes in all vessel layers, from the endothelium to the perivascular adipose tissue (PVAT), have been described. This mini-review focuses on the current knowledge of the structure and function of the vessel layers, specifically muscular arteries: intima, media, adventitia, PVAT, and the cell types harbored within each vessel layer. The contributions of each cell type to vessel homeostasis and pathophysiological development of hypertension will be highlighted.
Collapse
Affiliation(s)
- Patricia Martinez-Quinones
- Department of Surgery, Medical College of Georgia at Augusta University, Augusta, Georgia
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Cameron G McCarthy
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Nicole S Klee
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Amel Komic
- Department of Surgery, Medical College of Georgia at Augusta University, Augusta, Georgia
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Fabiano B Calmasini
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Fernanda Priviero
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Alexander Warner
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Yu Chenghao
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Camilla F Wenceslau
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| |
Collapse
|
32
|
Clifford PS, Ferguson BS, Jasperse JL, Hill MA. Arteriolar vasodilation involves actin depolymerization. Am J Physiol Heart Circ Physiol 2018; 315:H423-H428. [PMID: 29727217 DOI: 10.1152/ajpheart.00723.2017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It is generally assumed that relaxation of arteriolar vascular smooth muscle occurs through hyperpolarization of the cell membrane, reduction in intracellular Ca2+ concentration, and activation of myosin light chain phosphatase/inactivation of myosin light chain kinase. We hypothesized that vasodilation is related to depolymerization of F-actin. Cremaster muscles were dissected in rats under pentobarbital sodium anesthesia (50 mg/kg). First-order arterioles were dissected, cannulated on glass micropipettes, pressurized, and warmed to 34°C. Internal diameter was monitored with an electronic video caliper. The concentration of G-actin was determined in flash-frozen intact segments of arterioles by ultracentrifugation and Western blot analyses. Arterioles dilated by ~40% of initial diameter in response to pinacidil (1 × 10-6 mM) and sodium nitroprusside (5 × 10-5 mM). The G-actin-to-smooth muscle 22α ratio was 0.67 ± 0.09 in arterioles with myogenic tone and increased significantly to 1.32 ± 0.34 ( P < 0.01) when arterioles were dilated with pinacidil and 1.14 ± 0.18 ( P < 0.01) with sodium nitroprusside, indicating actin depolymerization. Compared with control vessels (49 ± 5%), the percentage of phosphorylated myosin light chain was significantly reduced by pinacidil (24 ± 2%, P < 0.01) but not sodium nitroprusside (42 ± 4%). These findings suggest that actin depolymerization is an important mechanism for vasodilation of resistance arterioles to external agonists. Furthermore, pinacidil produces smooth muscle relaxation via both decreases in myosin light chain phosphorylation and actin depolymerization, whereas sodium nitroprusside produces smooth muscle relaxation primarily via actin depolymerization. NEW & NOTEWORTHY This article adds to the accumulating evidence on the contribution of the actin cytoskeleton to the regulation of vascular smooth muscle tone in resistance arterioles. Actin depolymerization appears to be an important mechanism for vasodilation of resistance arterioles to pharmacological agonists. Dilation to the K+ channel opener pinacidil is produced by decreases in myosin light chain phosphorylation and actin depolymerization, whereas dilation to the nitric oxide donor sodium nitroprusside occurs primarily via actin depolymerization.
Collapse
Affiliation(s)
- Philip S Clifford
- College of Applied Health Sciences, University of Illinois at Chicago , Chicago, Illinois
| | - Brian S Ferguson
- College of Applied Health Sciences, University of Illinois at Chicago , Chicago, Illinois
| | - Jeffrey L Jasperse
- Department of Sports Medicine, Pepperdine University , Malibu, California
| | - Michael A Hill
- Dalton Cardiovascular Research Center and Department of Medical Pharmacology and Physiology, University of Missouri , Columbia, Missouri
| |
Collapse
|
33
|
Coucha M, Abdelsaid M, Ward R, Abdul Y, Ergul A. Impact of Metabolic Diseases on Cerebral Circulation: Structural and Functional Consequences. Compr Physiol 2018; 8:773-799. [PMID: 29687902 DOI: 10.1002/cphy.c170019] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Metabolic diseases including obesity, insulin resistance, and diabetes have profound effects on cerebral circulation. These diseases not only affect the architecture of cerebral blood arteries causing adverse remodeling, pathological neovascularization, and vasoregression but also alter the physiology of blood vessels resulting in compromised myogenic reactivity, neurovascular uncoupling, and endothelial dysfunction. Coupled with the disruption of blood brain barrier (BBB) integrity, changes in blood flow and microbleeds into the brain rapidly occur. This overview is organized into sections describing cerebrovascular architecture, physiology, and BBB in these diseases. In each section, we review these properties starting with larger arteries moving into smaller vessels. Where information is available, we review in the order of obesity, insulin resistance, and diabetes. We also tried to include information on biological variables such as the sex of the animal models noted since most of the information summarized was obtained using male animals. © 2018 American Physiological Society. Compr Physiol 8:773-799, 2018.
Collapse
Affiliation(s)
- Maha Coucha
- South University, School of Pharmacy, Savannah, Georgia, USA
| | | | - Rebecca Ward
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yasir Abdul
- Charlie Norwood VA Medical Center, Augusta, Georgia, USA.,Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Adviye Ergul
- Charlie Norwood VA Medical Center, Augusta, Georgia, USA.,Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
34
|
Weise-Cross L, Sands MA, Sheak JR, Broughton BRS, Snow JB, Gonzalez Bosc LV, Jernigan NL, Walker BR, Resta TC. Actin polymerization contributes to enhanced pulmonary vasoconstrictor reactivity after chronic hypoxia. Am J Physiol Heart Circ Physiol 2018; 314:H1011-H1021. [PMID: 29373038 DOI: 10.1152/ajpheart.00664.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Chronic hypoxia (CH) augments basal and endothelin-1 (ET-1)-induced pulmonary vasoconstrictor reactivity through reactive oxygen species (ROS) generation and RhoA/Rho kinase (ROCK)-dependent myofilament Ca2+ sensitization. Because ROCK promotes actin polymerization and the actin cytoskeleton regulates smooth muscle tension, we hypothesized that actin polymerization is required for enhanced basal and ET-1-dependent vasoconstriction after CH. To test this hypothesis, both end points were monitored in pressurized, endothelium-disrupted pulmonary arteries (fourth-fifth order) from control and CH (4 wk at 0.5 atm) rats. The actin polymerization inhibitors cytochalasin and latrunculin attenuated both basal and ET-1-induced vasoconstriction only in CH vessels. To test whether CH directly alters the arterial actin profile, we measured filamentous actin (F-actin)-to-globular actin (G-actin) ratios by fluorescent labeling of F-actin and G-actin in fixed pulmonary arteries and actin sedimentation assays using homogenized pulmonary artery lysates. We observed no difference in actin polymerization between groups under baseline conditions, but ET-1 enhanced actin polymerization in pulmonary arteries from CH rats. This response was blunted by the ROS scavenger tiron, the ROCK inhibitor fasudil, and the mDia (RhoA effector) inhibitor small-molecule inhibitor of formin homology domain 2. Immunoblot analysis revealed an effect of CH to increase both phosphorylated (inactive) and total levels of the actin disassembly factor cofilin but not phosphorylated cofilin-to-total cofilin ratios. We conclude that actin polymerization contributes to increased basal pulmonary arterial constriction and ET-1-induced vasoconstrictor reactivity after CH in a ROS- and ROCK-dependent manner. Our results further suggest that enhanced ET-1-mediated actin polymerization after CH is dependent on mDia but independent of changes in the phosphorylated cofilin-to-total cofilin ratio. NEW & NOTEWORTHY This research is the first to demonstrate a role for actin polymerization in chronic hypoxia-induced basal pulmonary arterial constriction and enhanced agonist-induced vasoconstrictor activity. These results suggest that a reactive oxygen species-Rho kinase-actin polymerization signaling pathway mediates this response and may provide a mechanistic basis for the vasoconstrictor component of pulmonary hypertension.
Collapse
Affiliation(s)
- Laura Weise-Cross
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Michelle A Sands
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Joshua R Sheak
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Brad R S Broughton
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Jessica B Snow
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Laura V Gonzalez Bosc
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| |
Collapse
|
35
|
Abstract
Smooth muscle contraction requires both myosin activation and actin cytoskeletal remodeling. Actin cytoskeletal reorganization facilitates smooth muscle contraction by promoting force transmission between the contractile unit and the extracellular matrix (ECM), and by enhancing intercellular mechanical transduction. Myosin may be viewed to serve as an "engine" for smooth muscle contraction whereas the actin cytoskeleton may function as a "transmission system" in smooth muscle. The actin cytoskeleton in smooth muscle also undergoes restructuring upon activation with growth factors or the ECM, which controls smooth muscle cell proliferation and migration. Abnormal smooth muscle contraction, cell proliferation, and motility contribute to the development of vascular and pulmonary diseases. A number of actin-regulatory proteins including protein kinases have been discovered to orchestrate actin dynamics in smooth muscle. In particular, Abelson tyrosine kinase (c-Abl) is an important molecule that controls actin dynamics, contraction, growth, and motility in smooth muscle. Moreover, c-Abl coordinates the regulation of blood pressure and contributes to the pathogenesis of airway hyperresponsiveness and vascular/airway remodeling in vivo. Thus, c-Abl may be a novel pharmacological target for the development of new therapy to treat smooth muscle diseases such as hypertension and asthma.
Collapse
Affiliation(s)
- Dale D Tang
- Albany Medical College, Albany, NY, United States.
| |
Collapse
|
36
|
Xu Q, Huff LP, Fujii M, Griendling KK. Redox regulation of the actin cytoskeleton and its role in the vascular system. Free Radic Biol Med 2017; 109:84-107. [PMID: 28285002 PMCID: PMC5497502 DOI: 10.1016/j.freeradbiomed.2017.03.004] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/17/2017] [Accepted: 03/06/2017] [Indexed: 12/17/2022]
Abstract
The actin cytoskeleton is critical for form and function of vascular cells, serving mechanical, organizational and signaling roles. Because many cytoskeletal proteins are sensitive to reactive oxygen species, redox regulation has emerged as a pivotal modulator of the actin cytoskeleton and its associated proteins. Here, we summarize work implicating oxidants in altering actin cytoskeletal proteins and focus on how these alterations affect cell migration, proliferation and contraction of vascular cells. Finally, we discuss the role of oxidative modification of the actin cytoskeleton in vivo and highlight its importance for vascular diseases.
Collapse
Affiliation(s)
- Qian Xu
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States; Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Lauren P Huff
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States
| | - Masakazu Fujii
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States.
| |
Collapse
|
37
|
Gupta K, Li Q, Fan JJ, Fong ELS, Song Z, Mo S, Tang H, Ng IC, Ng CW, Pawijit P, Zhuo S, Dong CY, Low BC, Wee A, Dan YY, Kanchanawong P, So P, Viasnoff V, Yu H. Actomyosin contractility drives bile regurgitation as an early response during obstructive cholestasis. J Hepatol 2017; 66:1231-1240. [PMID: 28189756 DOI: 10.1016/j.jhep.2017.01.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 01/10/2017] [Accepted: 01/29/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND & AIMS A wide range of liver diseases manifest as biliary obstruction, or cholestasis. However, the sequence of molecular events triggered as part of the early hepatocellular homeostatic response in obstructive cholestasis is poorly elucidated. Pericanalicular actin is known to accumulate during obstructive cholestasis. Therefore, we hypothesized that the pericanalicular actin cortex undergoes significant remodeling as a regulatory response to obstructive cholestasis. METHODS In vivo investigations were performed in a bile duct-ligated mouse model. Actomyosin contractility was assessed using sandwich-cultured rat hepatocytes transfected with various fluorescently labeled proteins and pharmacological inhibitors of actomyosin contractility. RESULTS Actomyosin contractility induces transient deformations along the canalicular membrane, a process we have termed inward blebbing. We show that these membrane intrusions are initiated by local ruptures in the pericanalicular actin cortex; and they typically retract following repair by actin polymerization and actomyosin contraction. However, above a certain osmotic pressure threshold, these inward blebs pinch away from the canalicular membrane into the hepatocyte cytoplasm as large vesicles (2-8μm). Importantly, we show that these vesicles aid in the regurgitation of bile from the bile canaliculi. CONCLUSION Actomyosin contractility induces the formation of bile-regurgitative vesicles, thus serving as an early homeostatic mechanism against increased biliary pressure during cholestasis. LAY SUMMARY Bile canaliculi expand and contract in response to the amount of secreted bile, and resistance from the surrounding actin bundles. Further expansion due to bile duct blockade leads to the formation of inward blebs, which carry away excess bile to prevent bile build up in the canaliculi.
Collapse
Affiliation(s)
- Kapish Gupta
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Qiushi Li
- Mechanobiology Institute, National University of Singapore, Singapore; National University of Singapore Research Institute, Singapore
| | - Jun Jun Fan
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), Singapore; BioSyM, Singapore-MIT Alliance for Research and Technology, Singapore; Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, China
| | - Eliza Li Shan Fong
- Department of Physiology, National University of Singapore, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore
| | - Ziwei Song
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Shupei Mo
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Haoyu Tang
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Inn Chuan Ng
- Department of Physiology, National University of Singapore, Singapore
| | - Chan Way Ng
- Department of Physiology, National University of Singapore, Singapore
| | - Pornteera Pawijit
- Department of Physiology, National University of Singapore, Singapore; NUS Graduate School of Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Shuangmu Zhuo
- BioSyM, Singapore-MIT Alliance for Research and Technology, Singapore; Fujian Normal University, Fuzhou, Fujian, China
| | - Chen-Yuan Dong
- Department of Physics, National Taiwan University, Taiwan
| | - Boon Chuan Low
- Mechanobiology Institute, National University of Singapore, Singapore; Department of Biological Sciences, National University of Singapore, Singapore
| | - Aileen Wee
- Department of Pathology, National University of Singapore, Singapore
| | - Yock Young Dan
- Division of Gastroenterology and Hepatology, National University Hospital, Singapore
| | - Pakorn Kanchanawong
- Mechanobiology Institute, National University of Singapore, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore
| | - Peter So
- BioSyM, Singapore-MIT Alliance for Research and Technology, Singapore
| | - Virgile Viasnoff
- Mechanobiology Institute, National University of Singapore, Singapore; CNRS UMI3639, Singapore
| | - Hanry Yu
- Mechanobiology Institute, National University of Singapore, Singapore; Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), Singapore; BioSyM, Singapore-MIT Alliance for Research and Technology, Singapore; Department of Physiology, National University of Singapore, Singapore; Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
38
|
Bays JL, Campbell HK, Heidema C, Sebbagh M, DeMali KA. Linking E-cadherin mechanotransduction to cell metabolism through force-mediated activation of AMPK. Nat Cell Biol 2017; 19:724-731. [PMID: 28553939 PMCID: PMC5494977 DOI: 10.1038/ncb3537] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 04/24/2017] [Indexed: 02/08/2023]
Abstract
The response of cells to mechanical force is a major determinant of cell behaviour and is an energetically costly event. How cells derive energy to resist mechanical force is unknown. Here, we show that application of force to E-cadherin stimulates liver kinase B1 (LKB1) to activate AMP-activated protein kinase (AMPK), a master regulator of energy homeostasis. LKB1 recruits AMPK to the E-cadherin mechanotransduction complex, thereby stimulating actomyosin contractility, glucose uptake and ATP production. The increase in ATP provides energy to reinforce the adhesion complex and actin cytoskeleton so that the cell can resist physiological forces. Together, these findings reveal a paradigm for how mechanotransduction and metabolism are linked and provide a framework for understanding how diseases involving contractile and metabolic disturbances arise.
Collapse
Affiliation(s)
- Jennifer L Bays
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | - Hannah K Campbell
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | - Christy Heidema
- Interdisciplinary Graduate Program in Molecular and Cellular Biology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | - Michael Sebbagh
- Centre de Recherche en Cancérologie de Marseille, Aix Marseille University UM105, Institut Paoli Calmettes, UMR7258 CNRS, U1068 INSERM, Cell Polarity, Cell signalling and Cancer-Equipe labellisée Ligue Contre le Cancer, Marseille 13273, France
| | - Kris A DeMali
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA.,Interdisciplinary Graduate Program in Molecular and Cellular Biology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| |
Collapse
|
39
|
Wang Y, Zhou Q, Wu B, Zhou H, Zhang X, Jiang W, Wang L, Wang A. Propofol induces excessive vasodilation of aortic rings by inhibiting protein kinase Cβ2 and θ in spontaneously hypertensive rats. Br J Pharmacol 2017; 174:1984-2000. [PMID: 28369981 DOI: 10.1111/bph.13797] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 02/21/2017] [Accepted: 03/19/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Exaggerated hypotension following administration of propofol is strongly predicted in patients with hypertension. Increased PKCs play a crucial role in regulating vascular tone. We studied whether propofol induces vasodilation by inhibiting increased PKC activity in spontaneously hypertensive rats (SHRs) and, if so, whether contractile Ca2+ sensitization pathways and filamentous-globular (F/G) actin dynamics were involved. EXPERIMENTAL APPROACH Rings of thoracic aorta, denuded of endothelium, from normotensive Wistar-Kyoto (WKY) rats and SHR were prepared for functional studies. Expression and activity of PKCs in vascular smooth muscle (VSM) cells were determined by Western blot analysis and elisa respectively. Phosphorylation of the key proteins in PKC Ca2+ sensitization pathways was also examined. Actin polymerization was evaluated by differential centrifugation to probe G- and F-actin content. KEY RESULTS Basal expression and activity of PKCβ2 and PKCθ were increased in aortic VSMs of SHR, compared with those from WKY rats. Vasorelaxation of SHR aortas by propofol was markedly attenuated by LY333531 (a specific PKCβ inhibitor) or the PKCθ pseudo-substrate inhibitor. Furthermore, noradrenaline-enhanced phosphorylation, and the translocation of PKCβ2 and PKCθ, was inhibited by propofol, with decreased actin polymerization and PKCβ2-mediated Ca2+ sensitization pathway in SHR aortas. CONCLUSION AND IMPLICATIONS Propofol suppressed increased PKCβ2 and PKCθ activity, which was partly responsible for exaggerated vasodilation in SHR. This suppression results in inhibition of actin polymerization, as well as that of the PKCβ2- but not PKCθ-mediated, Ca2+ sensitization pathway. These data provide a novel explanation for the unwanted side effects of propofol.
Collapse
Affiliation(s)
- Yan Wang
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Quanhong Zhou
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Bin Wu
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Huixuan Zhou
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Xiaoli Zhang
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Wei Jiang
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Li Wang
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Aizhong Wang
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
40
|
El-Yazbi AF, Abd-Elrahman KS. ROK and Arteriolar Myogenic Tone Generation: Molecular Evidence in Health and Disease. Front Pharmacol 2017; 8:87. [PMID: 28280468 PMCID: PMC5322222 DOI: 10.3389/fphar.2017.00087] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/10/2017] [Indexed: 12/13/2022] Open
Abstract
The myogenic response is an inherent property of resistance arteries that warrants a relatively constant blood flow in response to changes in perfusion pressure and protect delicate organs from vascular insufficiencies and excessive blood flow. This fundamental phenomenon has been extensively studied aiming to elucidate the underlying mechanisms triggering smooth muscle contraction in response to intraluminal pressure elevation, particularly, Rho-associated kinase (ROK)-mediated Ca2+-independent mechanisms. The size of the resistance arteries limits the capacity to examine changes in protein phosphorylation/expression levels associated with ROK signaling. A highly sensitive biochemical detection approach was beneficial in examining the role of ROK in different force generation mechanisms along the course of myogenic constriction. In this mini review, we summarize recent results showing direct evidence for the contribution of ROK in development of myogenic response at the level of mechanotransduction, myosin light chain phosphatase inhibition and dynamic actin cytoskeleton reorganization. We will also present evidence that alterations in ROK signaling could underlie the progressive loss in myogenic response in a rat model of type 2 diabetes.
Collapse
Affiliation(s)
- Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of BeirutBeirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria UniversityAlexandria, Egypt
| | - Khaled S Abd-Elrahman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria UniversityAlexandria, Egypt; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of OttawaOttawa, ON, Canada
| |
Collapse
|
41
|
Toth P, Tarantini S, Csiszar A, Ungvari Z. Functional vascular contributions to cognitive impairment and dementia: mechanisms and consequences of cerebral autoregulatory dysfunction, endothelial impairment, and neurovascular uncoupling in aging. Am J Physiol Heart Circ Physiol 2017; 312:H1-H20. [PMID: 27793855 PMCID: PMC5283909 DOI: 10.1152/ajpheart.00581.2016] [Citation(s) in RCA: 330] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 10/10/2016] [Accepted: 10/26/2016] [Indexed: 12/16/2022]
Abstract
Increasing evidence from epidemiological, clinical and experimental studies indicate that age-related cerebromicrovascular dysfunction and microcirculatory damage play critical roles in the pathogenesis of many types of dementia in the elderly, including Alzheimer's disease. Understanding and targeting the age-related pathophysiological mechanisms that underlie vascular contributions to cognitive impairment and dementia (VCID) are expected to have a major role in preserving brain health in older individuals. Maintenance of cerebral perfusion, protecting the microcirculation from high pressure-induced damage and moment-to-moment adjustment of regional oxygen and nutrient supply to changes in demand are prerequisites for the prevention of cerebral ischemia and neuronal dysfunction. This overview discusses age-related alterations in three main regulatory paradigms involved in the regulation of cerebral blood flow (CBF): cerebral autoregulation/myogenic constriction, endothelium-dependent vasomotor function, and neurovascular coupling responses responsible for functional hyperemia. The pathophysiological consequences of cerebral microvascular dysregulation in aging are explored, including blood-brain barrier disruption, neuroinflammation, exacerbation of neurodegeneration, development of cerebral microhemorrhages, microvascular rarefaction, and ischemic neuronal dysfunction and damage. Due to the widespread attention that VCID has captured in recent years, the evidence for the causal role of cerebral microvascular dysregulation in cognitive decline is critically examined.
Collapse
Affiliation(s)
- Peter Toth
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Pecs, Hungary; and
| | - Stefano Tarantini
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Anna Csiszar
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Zoltan Ungvari
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma;
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| |
Collapse
|
42
|
Jernigan NL, Resta TC, Gonzalez Bosc LV. Altered Redox Balance in the Development of Chronic Hypoxia-induced Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:83-103. [PMID: 29047083 DOI: 10.1007/978-3-319-63245-2_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Normally, the pulmonary circulation is maintained in a low-pressure, low-resistance state with little resting tone. Pulmonary arteries are thin-walled and rely heavily on pulmonary arterial distension and recruitment for reducing pulmonary vascular resistance when cardiac output is elevated. Under pathophysiological conditions, however, active vasoconstriction and vascular remodeling lead to enhanced pulmonary vascular resistance and subsequent pulmonary hypertension (PH). Chronic hypoxia is a critical pathological factor associated with the development of PH resulting from airway obstruction (COPD, sleep apnea), diffusion impairment (interstitial lung disease), developmental lung abnormalities, or high altitude exposure (World Health Organization [WHO]; Group III). The rise in pulmonary vascular resistance increases right heart afterload causing right ventricular hypertrophy that can ultimately lead to right heart failure in patients with chronic lung disease. PH is typically characterized by diminished paracrine release of vasodilators, antimitogenic factors, and antithrombotic factors (e.g., nitric oxide and protacyclin) and enhanced production of vasoconstrictors and mitogenic factors (e.g., reactive oxygen species and endothelin-1) from the endothelium and lung parenchyma. In addition, phenotypic changes to pulmonary arterial smooth muscle cells (PASMC), including alterations in Ca2+ homeostasis, Ca2+ sensitivity, and activation of transcription factors are thought to play prominent roles in the development of both vasoconstrictor and arterial remodeling components of hypoxia-associated PH. These changes in PASMC function are briefly reviewed in Sect. 1 and the influence of altered reactive oxygen species homeostasis on PASMC function discussed in Sects. 2-4.
Collapse
Affiliation(s)
- Nikki L Jernigan
- Department Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Thomas C Resta
- Department Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Laura V Gonzalez Bosc
- Department Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA.
| |
Collapse
|
43
|
Abd-Elrahman KS, Colinas O, Walsh EJ, Zhu HL, Campbell CM, Walsh MP, Cole WC. Abnormal myosin phosphatase targeting subunit 1 phosphorylation and actin polymerization contribute to impaired myogenic regulation of cerebral arterial diameter in the type 2 diabetic Goto-Kakizaki rat. J Cereb Blood Flow Metab 2017; 37:227-240. [PMID: 26721393 PMCID: PMC5363741 DOI: 10.1177/0271678x15622463] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 10/26/2015] [Accepted: 11/17/2015] [Indexed: 12/11/2022]
Abstract
The myogenic response of cerebral resistance arterial smooth muscle to intraluminal pressure elevation is a key physiological mechanism regulating blood flow to the brain. Rho-associated kinase plays a critical role in the myogenic response by activating Ca2+ sensitization mechanisms: (i) Rho-associated kinase inhibits myosin light chain phosphatase by phosphorylating its targeting subunit myosin phosphatase targeting subunit 1 (at T855), augmenting 20 kDa myosin regulatory light chain (LC20) phosphorylation and force generation; and (ii) Rho-associated kinase stimulates cytoskeletal actin polymerization, enhancing force transmission to the cell membrane. Here, we tested the hypothesis that abnormal Rho-associated kinase-mediated myosin light chain phosphatase regulation underlies the dysfunctional cerebral myogenic response of the Goto-Kakizaki rat model of type 2 diabetes. Basal levels of myogenic tone, LC20, and MYPT1-T855 phosphorylation were elevated and G-actin content was reduced in arteries of pre-diabetic 8-10 weeks Goto-Kakizaki rats with normal serum insulin and glucose levels. Pressure-dependent myogenic constriction, LC20, and myosin phosphatase targeting subunit 1 phosphorylation and actin polymerization were suppressed in both pre-diabetic Goto-Kakizaki and diabetic (18-20 weeks) Goto-Kakizaki rats, whereas RhoA, ROK2, and MYPT1 expression were unaffected. We conclude that abnormal Rho-associated kinase-mediated Ca2+ sensitization contributes to the dysfunctional cerebral myogenic response in the Goto-Kakizaki model of type 2 diabetes.
Collapse
Affiliation(s)
- Khaled S Abd-Elrahman
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Olaia Colinas
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Emma J Walsh
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Hai-Lei Zhu
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Christine M Campbell
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Michael P Walsh
- The Smooth Muscle Research Group, Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - William C Cole
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| |
Collapse
|
44
|
Milewicz DM, Trybus KM, Guo DC, Sweeney HL, Regalado E, Kamm K, Stull JT. Altered Smooth Muscle Cell Force Generation as a Driver of Thoracic Aortic Aneurysms and Dissections. Arterioscler Thromb Vasc Biol 2017; 37:26-34. [PMID: 27879251 PMCID: PMC5222685 DOI: 10.1161/atvbaha.116.303229] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 11/02/2016] [Indexed: 12/30/2022]
Abstract
The importance of maintaining contractile function in aortic smooth muscle cells (SMCs) is evident by the fact that heterozygous mutations in the major structural proteins or kinases controlling contraction lead to the formation of aneurysms of the ascending thoracic aorta that predispose to life-threatening aortic dissections. Force generation by SMC requires ATP-dependent cyclic interactions between filaments composed of SMC-specific isoforms of α-actin (encoded by ACTA2) and myosin heavy chain (MYH11). ACTA2 and MYH11 mutations are predicted or have been shown to disrupt this cyclic interaction predispose to thoracic aortic disease. Movement of the myosin motor domain is controlled by phosphorylation of the regulatory light chain on the myosin filament, and loss-of-function mutations in the dedicated kinase for this phosphorylation, myosin light chain kinase (MYLK) also predispose to thoracic aortic disease. Finally, a mutation in the cGMP-activated protein kinase (PRKG1) results in constitutive activation of the kinase in the absence of cGMP, thus driving SMC relaxation in part through increased dephosphorylation of the regulatory light chain and predisposes to thoracic aortic disease. Furthermore, SMCs cannot generate force without connections to the extracellular matrix through focal adhesions, and mutations in the major protein in the extracellular matrix, fibrillin-1, linking SMCs to the matrix also cause thoracic aortic disease in individuals with Marfan syndrome. Thus, disruption of the ability of the aortic SMC to generate force through the elastin-contractile units in response to pulsatile blood flow may be a primary driver for thoracic aortic aneurysms and dissections.
Collapse
MESH Headings
- Actins/genetics
- Actins/metabolism
- Aortic Dissection/genetics
- Aortic Dissection/metabolism
- Aortic Dissection/pathology
- Aortic Dissection/physiopathology
- Animals
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/physiopathology
- Calcium-Binding Proteins/genetics
- Calcium-Binding Proteins/metabolism
- Cyclic GMP-Dependent Protein Kinase Type I/genetics
- Cyclic GMP-Dependent Protein Kinase Type I/metabolism
- Dilatation, Pathologic
- Elastin/metabolism
- Genetic Markers
- Genetic Testing
- Heredity
- Humans
- Mechanotransduction, Cellular
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Mutation
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Myosin Heavy Chains/genetics
- Myosin Heavy Chains/metabolism
- Myosin-Light-Chain Kinase/genetics
- Myosin-Light-Chain Kinase/metabolism
- Phenotype
- Pulsatile Flow
- Vasoconstriction/genetics
Collapse
Affiliation(s)
- Dianna M Milewicz
- From the Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (D.M.M., D.-c.G., E.R.); Department of Molecular Physiology and Biophysics, University of Vermont, Burlington (K.M.T.); Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville (H.L.S.); and Department of Physiology, University of Texas Southwestern Medical Center, Dallas (K.K. J.T.S.).
| | - Kathleen M Trybus
- From the Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (D.M.M., D.-c.G., E.R.); Department of Molecular Physiology and Biophysics, University of Vermont, Burlington (K.M.T.); Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville (H.L.S.); and Department of Physiology, University of Texas Southwestern Medical Center, Dallas (K.K. J.T.S.)
| | - Dong-Chuan Guo
- From the Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (D.M.M., D.-c.G., E.R.); Department of Molecular Physiology and Biophysics, University of Vermont, Burlington (K.M.T.); Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville (H.L.S.); and Department of Physiology, University of Texas Southwestern Medical Center, Dallas (K.K. J.T.S.)
| | - H Lee Sweeney
- From the Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (D.M.M., D.-c.G., E.R.); Department of Molecular Physiology and Biophysics, University of Vermont, Burlington (K.M.T.); Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville (H.L.S.); and Department of Physiology, University of Texas Southwestern Medical Center, Dallas (K.K. J.T.S.)
| | - Ellen Regalado
- From the Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (D.M.M., D.-c.G., E.R.); Department of Molecular Physiology and Biophysics, University of Vermont, Burlington (K.M.T.); Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville (H.L.S.); and Department of Physiology, University of Texas Southwestern Medical Center, Dallas (K.K. J.T.S.)
| | - Kristine Kamm
- From the Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (D.M.M., D.-c.G., E.R.); Department of Molecular Physiology and Biophysics, University of Vermont, Burlington (K.M.T.); Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville (H.L.S.); and Department of Physiology, University of Texas Southwestern Medical Center, Dallas (K.K. J.T.S.)
| | - James T Stull
- From the Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (D.M.M., D.-c.G., E.R.); Department of Molecular Physiology and Biophysics, University of Vermont, Burlington (K.M.T.); Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville (H.L.S.); and Department of Physiology, University of Texas Southwestern Medical Center, Dallas (K.K. J.T.S.)
| |
Collapse
|
45
|
Hong K, Zhao G, Hong Z, Sun Z, Yang Y, Clifford PS, Davis MJ, Meininger GA, Hill MA. Mechanical activation of angiotensin II type 1 receptors causes actin remodelling and myogenic responsiveness in skeletal muscle arterioles. J Physiol 2016; 594:7027-7047. [PMID: 27531064 PMCID: PMC5134373 DOI: 10.1113/jp272834] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 08/09/2016] [Indexed: 12/15/2022] Open
Abstract
KEY POINTS Candesartan, an inverse agonist of the type 1 angiotensin II receptor (AT1 R), causes a concentration-dependent inhibition of pressure-dependent myogenic tone consistent with previous reports of mechanosensitivity of this G protein-coupled receptor. Mechanoactivation of the AT1 R occurs independently of local angiotensin II production and the type 2 angiotensin receptor. Mechanoactivation of the AT1 R stimulates actin polymerization by a protein kinase C-dependent mechanism, but independently of a change in intracellular Ca2+ . Using atomic force microscopy, changes in single vascular smooth muscle cell cortical actin are observed to remodel following mechanoactivation of the AT1 R. ABSTRACT The Gq/11 protein-coupled angiotensin II type 1 receptor (AT1 R) has been shown to be activated by mechanical stimuli. In the vascular system, evidence supports the AT1 R being a mechanosensor that contributes to arteriolar myogenic constriction. The aim of this study was to determine if AT1 R mechanoactivation affects myogenic constriction in skeletal muscle arterioles and to determine underlying cellular mechanisms. Using pressure myography to study rat isolated first-order cremaster muscle arterioles the AT1 R inhibitor candesartan (10-7 -10-5 m) showed partial but concentration-dependent inhibition of myogenic reactivity. Inhibition was demonstrated by a rightward shift in the pressure-diameter relationship over the intraluminal pressure range, 30-110 mmHg. Pressure-induced changes in global vascular smooth muscle intracellular Ca2+ (using Fura-2) were similar in the absence or presence of candesartan, indicating that AT1 R-mediated myogenic constriction relies on Ca2+ -independent downstream signalling. The diacylglycerol analogue 1-oleoyl-2-acetyl-sn-glycerol (OAG) reversed the inhibitory effect of candesartan, while this rescue effect was prevented by the protein kinase C (PKC) inhibitor GF 109203X. Both candesartan and PKC inhibition caused increased G-actin levels, as determined by Western blotting of vessel lysates, supporting involvement of cytoskeletal remodelling. At the single vascular smooth muscle cell level, atomic force microscopy showed that cell swelling (stretch) with hypotonic buffer also caused thickening of cortical actin fibres and this was blocked by candesartan. Collectively, the present studies support growing evidence for novel modes of activation of the AT1 R in arterioles and suggest that mechanically activated AT1 R generates diacylglycerol, which in turn activates PKC which induces the actin cytoskeleton reorganization that is required for pressure-induced vasoconstriction.
Collapse
Affiliation(s)
- Kwangseok Hong
- Dalton Cardiovascular Research CentreUniversity of MissouriColumbiaMO65211USA
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMO65211USA
- Robert M. Berne Cardiovascular Research Centre and Department of Molecular Physiology and Biological PhysicsUniversity of VirginiaCharlottesvilleVA22908USA
| | - Guiling Zhao
- College of Applied Health SciencesUniversity of Illinois at ChicagoChicagoIL60612USA
| | - Zhongkui Hong
- Dalton Cardiovascular Research CentreUniversity of MissouriColumbiaMO65211USA
- Department of Biomedical EngineeringUniversity of South DakotaSioux FallsSD57107USA
| | - Zhe Sun
- Dalton Cardiovascular Research CentreUniversity of MissouriColumbiaMO65211USA
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMO65211USA
| | - Yan Yang
- Dalton Cardiovascular Research CentreUniversity of MissouriColumbiaMO65211USA
| | - Philip S. Clifford
- College of Applied Health SciencesUniversity of Illinois at ChicagoChicagoIL60612USA
| | - Michael J. Davis
- Dalton Cardiovascular Research CentreUniversity of MissouriColumbiaMO65211USA
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMO65211USA
| | - Gerald A. Meininger
- Dalton Cardiovascular Research CentreUniversity of MissouriColumbiaMO65211USA
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMO65211USA
| | - Michael A. Hill
- Dalton Cardiovascular Research CentreUniversity of MissouriColumbiaMO65211USA
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMO65211USA
| |
Collapse
|
46
|
Yao Y, Mak AF. Strengthening of C2C12 mouse myoblasts against compression damage by mild cyclic compressive stimulation. J Biomech 2016; 49:3956-3961. [PMID: 27884430 DOI: 10.1016/j.jbiomech.2016.11.050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 11/10/2016] [Accepted: 11/11/2016] [Indexed: 11/28/2022]
Abstract
Deep tissue injury (DTI) is a severe kind of pressure ulcers formed by sustained deformation of muscle tissues over bony prominences. As a major clinical issue, DTI affects people with physical disabilities, and is obviously related to the load-bearing capacity of muscle cells in various in-vivo conditions. It is important to provide a preventive approach to help muscle cells from being damaged by compressive stress. In this study, we hypothesized that cyclic compressive stimulation could strengthen muscle cells against compressive damage and enhance the cell plasma membrane resealing capability. Monolayer of myoblasts was cultured in the cell culture dish covered by a cylinder 0.5% agarose gel. The platen indenter was applied with 20% strain on the agarose gel in the Mach-1 micromechanical system. The vibration was 1Hz sinusoidal function with amplitude 0.2% strain based on 20% gel strain. Cyclic compressive stimulation for 2h could enhance the compressive stress damage threshold of muscle cells, the muscle cell plasma membrane resealing ratio and viability of muscle cell under static loading as preventive approach. This approach might help to reduce the risk of DTI in clinic.
Collapse
Affiliation(s)
- Yifei Yao
- Division of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Arthur Ft Mak
- Division of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
47
|
Coucha M, Abdelsaid M, Li W, Johnson MH, Orfi L, El-Remessy AB, Fagan SC, Ergul A. Nox4 contributes to the hypoxia-mediated regulation of actin cytoskeleton in cerebrovascular smooth muscle. Life Sci 2016; 163:46-54. [PMID: 27558234 DOI: 10.1016/j.lfs.2016.08.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/12/2016] [Accepted: 08/19/2016] [Indexed: 11/29/2022]
Abstract
UNLABELLED Ischemia/reperfusion and the resulting oxidative/nitrative stress impair cerebral myogenic tone via actin depolymerization. While it is known that NADPH oxidase (Nox) family is a major source of vascular oxidative stress; the extent and mechanisms by which Nox activation contributes to actin depolymerization, and equally important, the relative role of Nox isoforms in this response is not clear. AIM To determine the role of Nox4 in hypoxia-mediated actin depolymerization and myogenic-tone impairment in cerebral vascular smooth muscle. MAIN METHODS Control and Nox4 deficient (siRNA knock-down) human brain vascular smooth muscle cells (HBVSMC) were exposed to 30-min hypoxia/45-min reoxygenation. Nox2, Nox4, inducible and neuronal nitric oxide synthase (iNOS and nNOS) and nitrotyrosine levels as well as F:G actin were determined. Myogenic-tone was measured using pressurized arteriography in middle cerebral artery isolated from rats subjected to sham, 30-min ischemia/45-min reperfusion or ex-vivo oxygen glucose deprivation in the presence and absence of Nox inhibitors. RESULTS Nox4 and iNOS expression were significantly upregulated following hypoxia or ischemia/reperfusion. Hypoxia augmented nitrotyrosine levels while reducing F actin. These effects were nullified by inhibiting nitration with epicatechin or pharmacological or molecular inhibition of Nox4. Ischemia/reperfusion impaired myogenic-tone, which was restored by the selective inhibition of Nox4. CONCLUSION Nox4 activation in VSMCs contributes to actin depolymerization after hypoxia, which could be the underlying mechanism for myogenic-tone impairment following ischemia/reperfusion.
Collapse
Affiliation(s)
- Maha Coucha
- Charlie Norwood VA Medical Center, Augusta University, United States; Departments of Physiology, Augusta University, United States; Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, United States
| | - Mohammed Abdelsaid
- Charlie Norwood VA Medical Center, Augusta University, United States; Departments of Physiology, Augusta University, United States
| | - Weiguo Li
- Charlie Norwood VA Medical Center, Augusta University, United States; Departments of Physiology, Augusta University, United States
| | | | | | - Azza B El-Remessy
- Charlie Norwood VA Medical Center, Augusta University, United States; Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, United States
| | - Susan C Fagan
- Charlie Norwood VA Medical Center, Augusta University, United States; Departments of Neurology, Augusta University, United States; Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, United States
| | - Adviye Ergul
- Charlie Norwood VA Medical Center, Augusta University, United States; Departments of Physiology, Augusta University, United States; Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, United States.
| |
Collapse
|
48
|
Brozovich FV, Nicholson CJ, Degen CV, Gao YZ, Aggarwal M, Morgan KG. Mechanisms of Vascular Smooth Muscle Contraction and the Basis for Pharmacologic Treatment of Smooth Muscle Disorders. Pharmacol Rev 2016; 68:476-532. [PMID: 27037223 PMCID: PMC4819215 DOI: 10.1124/pr.115.010652] [Citation(s) in RCA: 321] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The smooth muscle cell directly drives the contraction of the vascular wall and hence regulates the size of the blood vessel lumen. We review here the current understanding of the molecular mechanisms by which agonists, therapeutics, and diseases regulate contractility of the vascular smooth muscle cell and we place this within the context of whole body function. We also discuss the implications for personalized medicine and highlight specific potential target molecules that may provide opportunities for the future development of new therapeutics to regulate vascular function.
Collapse
Affiliation(s)
- F V Brozovich
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - C J Nicholson
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - C V Degen
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - Yuan Z Gao
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - M Aggarwal
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - K G Morgan
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| |
Collapse
|
49
|
Long Lasting Microvascular Tone Alteration in Rat Offspring Exposed In Utero to Maternal Hyperglycaemia. PLoS One 2016; 11:e0146830. [PMID: 26756337 PMCID: PMC4710502 DOI: 10.1371/journal.pone.0146830] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 12/22/2015] [Indexed: 12/18/2022] Open
Abstract
Epidemiologic studies have demonstrated that cardiovascular risk is not only determined by conventional risk factors in adulthood, but also by early life events which may reprogram vascular function. To evaluate the effect of maternal diabetes on fetal programming of vascular tone in offspring and its evolution during adulthood, we investigated vascular reactivity of third order mesenteric arteries from diabetic mother offspring (DMO) and control mother offspring (CMO) aged 3 and 18 months. In arteries isolated from DMO the relaxation induced by prostacyclin analogues was reduced in both 3- and 18-month old animals although endothelium (acetylcholine)-mediated relaxation was reduced in 18-month old DMO only. Endothelium-independent (sodium nitroprusside) relaxation was not affected. Pressure-induced myogenic tone, which controls local blood flow, was reduced in 18-month old CMO compared to 3-month old CMO. Interestingly, myogenic tone was maintained at a high level in 18-month old DMO even though agonist-induced vasoconstriction was not altered. These perturbations, in 18-months old DMO rats, were associated with an increased pMLC/MLC, pPKA/PKA ratio and an activated RhoA protein. Thus, we highlighted perturbations in the reactivity of resistance mesenteric arteries in DMO, at as early as 3 months of age, followed by the maintenance of high myogenic tone in older rats. These modifications are in favour of excessive vasoconstrictor tone. These results evidenced a fetal programming of vascular functions of resistance arteries in adult rats exposed in utero to maternal diabetes, which could explain a re-setting of vascular functions and, at least in part, the occurrence of hypertension later in life.
Collapse
|
50
|
Abstract
Vascular smooth muscle (VSM; see Table 1 for a list of abbreviations) is a heterogeneous biomaterial comprised of cells and extracellular matrix. By surrounding tubes of endothelial cells, VSM forms a regulated network, the vasculature, through which oxygenated blood supplies specialized organs, permitting the development of large multicellular organisms. VSM cells, the engine of the vasculature, house a set of regulated nanomotors that permit rapid stress-development, sustained stress-maintenance and vessel constriction. Viscoelastic materials within, surrounding and attached to VSM cells, comprised largely of polymeric proteins with complex mechanical characteristics, assist the engine with countering loads imposed by the heart pump, and with control of relengthening after constriction. The complexity of this smart material can be reduced by classical mechanical studies combined with circuit modeling using spring and dashpot elements. Evaluation of the mechanical characteristics of VSM requires a more complete understanding of the mechanics and regulation of its biochemical parts, and ultimately, an understanding of how these parts work together to form the machinery of the vascular tree. Current molecular studies provide detailed mechanical data about single polymeric molecules, revealing viscoelasticity and plasticity at the protein domain level, the unique biological slip-catch bond, and a regulated two-step actomyosin power stroke. At the tissue level, new insight into acutely dynamic stress-strain behavior reveals smooth muscle to exhibit adaptive plasticity. At its core, physiology aims to describe the complex interactions of molecular systems, clarifying structure-function relationships and regulation of biological machines. The intent of this review is to provide a comprehensive presentation of one biomachine, VSM.
Collapse
Affiliation(s)
- Paul H Ratz
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|