1
|
Mironova GY, Kowalewska PM, El-Lakany M, Tran CHT, Sancho M, Zechariah A, Jackson WF, Welsh DG. The conducted vasomotor response and the principles of electrical communication in resistance arteries. Physiol Rev 2024; 104:33-84. [PMID: 37410448 PMCID: PMC11918294 DOI: 10.1152/physrev.00035.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 06/26/2023] [Accepted: 07/01/2023] [Indexed: 07/07/2023] Open
Abstract
Biological tissues are fed by arterial networks whose task is to set blood flow delivery in accordance with energetic demand. Coordinating vasomotor activity among hundreds of neighboring segments is an essential process, one dependent upon electrical information spreading among smooth muscle and endothelial cells. The "conducted vasomotor response" is a functional expression of electrical spread, and it is this process that lies at the heart of this critical review. Written in a narrative format, this review first highlights historical manuscripts and then characterizes the conducted response across a range of preparations. Trends are highlighted and used to guide subsequent sections, focused on cellular foundations, biophysical underpinnings, and regulation in health and disease. Key information has been tabulated; figures reinforce grounding concepts and reveal a framework within which theoretical and experimental work can be rationalized. This summative review highlights that despite 30 years of concerted experimentation, key aspects of the conducted response remain ill defined. Of note is the need to rationalize the regulation and deterioration of conduction in pathobiological settings. New quantitative tools, along with transgenic technology, are discussed as a means of propelling this investigative field forward.
Collapse
Affiliation(s)
- Galina Yu Mironova
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Paulina M Kowalewska
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Mohammed El-Lakany
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Cam Ha T Tran
- Department of Physiology, Faculty of Medicine, University of Nevada (Reno), Reno, Nevada, United States
| | - Maria Sancho
- Department of Physiology, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Anil Zechariah
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland and Labrador, St. John's, Newfoundland, Canada
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Donald G Welsh
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
2
|
Sedovy MW, Leng X, Leaf MR, Iqbal F, Payne LB, Chappell JC, Johnstone SR. Connexin 43 across the Vasculature: Gap Junctions and Beyond. J Vasc Res 2022; 60:101-113. [PMID: 36513042 PMCID: PMC11073551 DOI: 10.1159/000527469] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/26/2022] [Indexed: 12/15/2022] Open
Abstract
Connexin 43 (Cx43) is essential to the function of the vasculature. Cx43 proteins form gap junctions that allow for the exchange of ions and molecules between vascular cells to facilitate cell-to-cell signaling and coordinate vasomotor activity. Cx43 also has intracellular signaling functions that influence vascular cell proliferation and migration. Cx43 is expressed in all vascular cell types, although its expression and function vary by vessel size and location. This includes expression in vascular smooth muscle cells (vSMC), endothelial cells (EC), and pericytes. Cx43 is thought to coordinate homocellular signaling within EC and vSMC. Cx43 gap junctions also function as conduits between different cell types (heterocellular signaling), between EC and vSMC at the myoendothelial junction, and between pericyte and EC in capillaries. Alterations in Cx43 expression, localization, and post-translational modification have been identified in vascular disease states, including atherosclerosis, hypertension, and diabetes. In this review, we discuss the current understanding of Cx43 localization and function in healthy and diseased blood vessels across all vascular beds.
Collapse
Affiliation(s)
- Meghan W. Sedovy
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
- Translational Biology, Medicine, And Health Graduate Program, Virginia Tech, Blacksburg, VA, USA
| | - Xinyan Leng
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
| | - Melissa R. Leaf
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | - Farwah Iqbal
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | - Laura Beth Payne
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
| | - John C. Chappell
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
| | - Scott R. Johnstone
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
3
|
Lu B, Shen L, Zhu H, Xi L, Wang W, Ouyang X. Association between serum homocysteine and sarcopenia among hospitalized older Chinese adults: a cross-sectional study. BMC Geriatr 2022; 22:896. [PMID: 36424548 PMCID: PMC9685861 DOI: 10.1186/s12877-022-03632-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE Hyperhomocysteinemia (HHcy) is considered to increase the risk of sarcopenia (S) and remains controversial. In this study, we aimed to investigate the prevalence of S among older Chinese adults and explore whether homocysteine (Hcy) was independently associated with S. METHODS This cross-sectional study was performed among older adults hospitalized in the Geriatric Hospital of Nanjing Medical University between June 2017 and December 2021. We measured all participants' serum Hcy levels, hand grip strength, gait speed and appendicular skeletal muscle index(ASMI) using bioelectrical impedance analysis (BIA). S was defined based on the criteria of the Asian Working Group for Sarcopenia 2 (AWGS2), which included muscle mass (ASMI< 7.0 kg/m2 for men and ASMI< 5.7 kg/m2 for women by BIA) and low muscle strength (handgrip strength < 28 kg for men and < 18 kg for women), and/or gait speed < 1.0 m/s. HHcy defined as Hcy ≥10 μmol/L. The strength of the association between Hcy and the risk of S was analyzed by multivariate logistic regression using three models that adjusted for possible confounding variables to calculate the odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS Among the 441 subjects, 161 (36.5%) were diagnosed with S, and 343 (77.8%) were diagnosed with HHcy. A significant association was detected between S and serum Hcy per 1-μmol/L increase after adjustment for age, gender, education, smoking, body mass index (BMI), Mini Nutritional Assessment Short Form (MNA-SF), alanine aminotransferase (ALT), C-reactive protein (CRP), hemoglobin (Hb), albumin (ALB), diabetes, kidney disease, and statin use (OR = 1.07, 95% CI = 1.03-1.12, P = 0.002). The OR for S in the HHcy group (≥10 μmol/L) was nearly 5-fold that in the normal Hcy group (OR 4.96, 95% CI 2.67-9.24, P < 0.001). In a gender-based subgroup analysis that adjusted for age, education, smoking, BMI, MNA-SF, ALT, CRP, Hb, and ALB, female subjects with HHcy had an increased risk of S (OR 10.35, 95% CI 2.84-37.68, P < 0.001). CONCLUSIONS Our results demonstrated that elevated Hcy levels have an independent association with S in older adults. This suggests that the downward adjustment of HHcy (cutoff value < 10 μmol/l) might decrease the risk of S.
Collapse
Affiliation(s)
- Bing Lu
- grid.89957.3a0000 0000 9255 8984Department of Geriatrics, Geriatric Hospital of Nanjing Medical University, 65 Jiangsu Road, Nanjing, 210024 China
| | - Lingyu Shen
- grid.89957.3a0000 0000 9255 8984Chronic Disease and Health Management Research Center, Geriatric Hospital of Nanjing Medical University, 65 Jiangsu Road, Nanjing, 210024 China
| | - Haiqiong Zhu
- grid.89957.3a0000 0000 9255 8984Department of Geriatrics, Geriatric Hospital of Nanjing Medical University, 65 Jiangsu Road, Nanjing, 210024 China
| | - Ling Xi
- grid.89957.3a0000 0000 9255 8984Department of Geriatrics, Geriatric Hospital of Nanjing Medical University, 65 Jiangsu Road, Nanjing, 210024 China
| | - Wei Wang
- grid.89957.3a0000 0000 9255 8984Chronic Disease and Health Management Research Center, Geriatric Hospital of Nanjing Medical University, 65 Jiangsu Road, Nanjing, 210024 China
| | - Xiaojun Ouyang
- grid.89957.3a0000 0000 9255 8984Department of Geriatrics, Geriatric Hospital of Nanjing Medical University, 65 Jiangsu Road, Nanjing, 210024 China
| |
Collapse
|
4
|
Murrant CL, Fletcher NM. Capillary communication: the role of capillaries in sensing the tissue environment, coordinating the microvascular, and controlling blood flow. Am J Physiol Heart Circ Physiol 2022; 323:H1019-H1036. [PMID: 36149771 DOI: 10.1152/ajpheart.00088.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Historically, capillaries have been viewed as the microvascular site for flux of nutrients to cells and removal of waste products. Capillaries are the most numerous blood vessel segment within the tissue, whose vascular wall consists of only a single layer of endothelial cells and are situated within microns of each cell of the tissue, all of which optimizes capillaries for the exchange of nutrients between the blood compartment and the interstitial space of tissues. There is, however, a growing body of evidence to support that capillaries play an important role in sensing the tissue environment, coordinating microvascular network responses, and controlling blood flow. Much of our growing understanding of capillaries stems from work in skeletal muscle and more recent work in the brain, where capillaries can be stimulated by products released from cells of the tissue during increased activity and are able to communicate with upstream and downstream vascular segments, enabling capillaries to sense the activity levels of the tissue and send signals to the microvascular network to coordinate the blood flow response. This review will focus on the emerging role that capillaries play in communication between cells of the tissue and the vascular network required to direct blood flow to active cells in skeletal muscle and the brain. We will also highlight the emerging central role that disruptions in capillary communication may play in blood flow dysregulation, pathophysiology, and disease.
Collapse
Affiliation(s)
- Coral L Murrant
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Nicole M Fletcher
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
5
|
King DR, Sedovy MW, Eaton X, Dunaway LS, Good ME, Isakson BE, Johnstone SR. Cell-To-Cell Communication in the Resistance Vasculature. Compr Physiol 2022; 12:3833-3867. [PMID: 35959755 DOI: 10.1002/cphy.c210040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The arterial vasculature can be divided into large conduit arteries, intermediate contractile arteries, resistance arteries, arterioles, and capillaries. Resistance arteries and arterioles primarily function to control systemic blood pressure. The resistance arteries are composed of a layer of endothelial cells oriented parallel to the direction of blood flow, which are separated by a matrix layer termed the internal elastic lamina from several layers of smooth muscle cells oriented perpendicular to the direction of blood flow. Cells within the vessel walls communicate in a homocellular and heterocellular fashion to govern luminal diameter, arterial resistance, and blood pressure. At rest, potassium currents govern the basal state of endothelial and smooth muscle cells. Multiple stimuli can elicit rises in intracellular calcium levels in either endothelial cells or smooth muscle cells, sourced from intracellular stores such as the endoplasmic reticulum or the extracellular space. In general, activation of endothelial cells results in the production of a vasodilatory signal, usually in the form of nitric oxide or endothelial-derived hyperpolarization. Conversely, activation of smooth muscle cells results in a vasoconstriction response through smooth muscle cell contraction. © 2022 American Physiological Society. Compr Physiol 12: 1-35, 2022.
Collapse
Affiliation(s)
- D Ryan King
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA
| | - Meghan W Sedovy
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA.,Translational Biology, Medicine, and Health Graduate Program, Virginia Tech, Blacksburg, Virginia, USA
| | - Xinyan Eaton
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA
| | - Luke S Dunaway
- Robert M. Berne Cardiovascular Research Centre, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Miranda E Good
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Centre, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Scott R Johnstone
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
6
|
Do skeletal muscle motor units and microvascular units align to help match blood flow to metabolic demand? Eur J Appl Physiol 2021; 121:1241-1254. [PMID: 33538852 DOI: 10.1007/s00421-021-04598-4] [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: 09/23/2020] [Accepted: 01/10/2021] [Indexed: 10/22/2022]
Abstract
PURPOSE We explore the motor unit recruitment and control of perfusion of microvascular units in skeletal muscle to determine whether they coordinate to match blood flow to metabolic demand. METHODS The PubMed database was searched for historical, current and relevant literature. RESULTS A microvascular, or capillary unit consists of 2-20 individual capillaries. Individual capillaries within a capillary unit cannot increase perfusion independently of other capillaries within the unit. Capillary units perfuse a short segment of approx. 12 muscle fibres located beside each other. Motor units consist of muscle fibres that can be dispersed widely within the muscle volume. During a contraction, where not all motor units are recruited, muscle fibre contraction will result in increased perfusion of associated capillaries as well as all capillaries within that capillary unit. Perfusion of the entire capillary unit will result in an increased blood flow delivery to muscle fibres associated with active motor unit plus approximately 11 other inactive muscle fibres within the same region. This will result in an overperfusion of the muscle resulting in blood flow in excess of the muscle fibre needs. CONCLUSIONS Given the architecture of the capillary units and the dispersed nature of muscle fibres within a motor unit, during submaximal contractions, where not all motor units are recruited, there will be a greater perfusion to the muscle than that predicted by the number of active muscle fibres. Such overperfusion brings into question if blood flow and metabolic demand are as tightly matched as previously assumed.
Collapse
|
7
|
Kumar G, Dey SK, Kundu S. Functional implications of vascular endothelium in regulation of endothelial nitric oxide synthesis to control blood pressure and cardiac functions. Life Sci 2020; 259:118377. [PMID: 32898526 DOI: 10.1016/j.lfs.2020.118377] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/24/2020] [Accepted: 08/31/2020] [Indexed: 11/29/2022]
Abstract
The endothelium is the innermost vascular lining performing significant roles all over the human body while maintaining the blood pressure at physiological levels. Malfunction of endothelium is thus recognized as a biomarker linked with many vascular diseases including but not limited to atherosclerosis, hypertension and thrombosis. Alternatively, prevention of endothelial malfunctioning or regulating the functions of its associated physiological partners like endothelial nitric oxide synthase can prevent the associated vascular disorders which account for the highest death toll worldwide. While many anti-hypertensive drugs are available commercially, a comprehensive description of the key physiological roles of the endothelium and its regulation by endothelial nitric oxide synthase or vice versa is the need of the hour to understand its contribution in vascular homeostasis. This, in turn, will help in designing new therapeutics targeting endothelial nitric oxide synthase or its interacting partners present in the cellular pool. This review describes the central role of vascular endothelium in the regulation of endothelial nitric oxide synthase while outlining the emerging drug targets present in the vasculature with potential to treat vascular disorders including hypertension.
Collapse
Affiliation(s)
- Gaurav Kumar
- Department of Biochemistry, University of Delhi, South Campus, New Delhi 110021, India
| | - Sanjay Kumar Dey
- Department of Biochemistry, University of Delhi, South Campus, New Delhi 110021, India; Center for Advanced Biotechnology and Medicine, Rutgers University, NJ 08854, USA
| | - Suman Kundu
- Department of Biochemistry, University of Delhi, South Campus, New Delhi 110021, India.
| |
Collapse
|
8
|
Abstract
Of the 21 members of the connexin family, 4 (Cx37, Cx40, Cx43, and Cx45) are expressed in the endothelium and/or smooth muscle of intact blood vessels to a variable and dynamically regulated degree. Full-length connexins oligomerize and form channel structures connecting the cytosol of adjacent cells (gap junctions) or the cytosol with the extracellular space (hemichannels). The different connexins vary mainly with regard to length and sequence of their cytosolic COOH-terminal tails. These COOH-terminal parts, which in the case of Cx43 are also translated as independent short isoforms, are involved in various cellular signaling cascades and regulate cell functions. This review focuses on channel-dependent and -independent effects of connexins in vascular cells. Channels play an essential role in coordinating and synchronizing endothelial and smooth muscle activity and in their interplay, in the control of vasomotor actions of blood vessels including endothelial cell reactivity to agonist stimulation, nitric oxide-dependent dilation, and endothelial-derived hyperpolarizing factor-type responses. Further channel-dependent and -independent roles of connexins in blood vessel function range from basic processes of vascular remodeling and angiogenesis to vascular permeability and interactions with leukocytes with the vessel wall. Together, these connexin functions constitute an often underestimated basis for the enormous plasticity of vascular morphology and function enabling the required dynamic adaptation of the vascular system to varying tissue demands.
Collapse
Affiliation(s)
- Ulrich Pohl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Planegg-Martinsried, Germany; Biomedical Centre, Cardiovascular Physiology, LMU Munich, Planegg-Martinsried, Germany; German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany; and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
9
|
Javed S, Mitchell K, Sidsworth D, Sellers SL, Reutens-Hernandez J, Massicotte HB, Egger KN, Lee CH, Payne GW. Inonotus obliquus attenuates histamine-induced microvascular inflammation. PLoS One 2019; 14:e0220776. [PMID: 31437163 PMCID: PMC6706056 DOI: 10.1371/journal.pone.0220776] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 07/23/2019] [Indexed: 01/14/2023] Open
Abstract
Cell-to-cell communication is a key element of microvascular blood flow control, including rapidly carrying signals through the vascular endothelium in response to local stimuli. This cell-to-cell communication is negatively impacted during inflammation through the disruption of junctional integrity. Such disruption is associated with promoting the onset of cardiovascular diseases as a result of altered microvascular blood flow regulation. Therefore, understanding the mechanisms how inflammation drives microvascular dysfunction and compounds that mitigate such inflammation and dysfunction are of great interest for development. As such we aimed to investigate extracts of mushrooms as potential novel compounds. Using intravital microscopy, the medicinal mushroom, Inonotus obliquus was observed, to attenuate histamine-induced inflammation conducted vasodilation in second-order arterioles in the gluteus maximus muscle of C57BL/6 mice. Mast cell activation by C48/80 similarly disrupted endothelial junctions and conducted vasodilation but only histamine was blocked by the histamine antagonist, pyrilamine not C48/80 suggesting the importance of mast cell activation. Data presented here supports that histamine induced inflammation is a major disruptor of junctional integrity, and highlights the important anti-inflammatory properties of Inonotus obliquus focusing future assessment of mast cells as putative target for Inonotus obliquus.
Collapse
Affiliation(s)
- Sumreen Javed
- Biochemistry and Molecular Biology Program, University of Northern British Columbia, Prince George, Canada
| | - Kevin Mitchell
- Northern Medical Program, University of Northern British Columbia, Prince George, Canada
| | - Danielle Sidsworth
- Northern Medical Program, University of Northern British Columbia, Prince George, Canada
| | - Stephanie L. Sellers
- Centre for Heart Lung Innovation & Department of Radiology, University of British Columba & St. Paul’s Hospital, Vancouver, Canada
| | - Jennifer Reutens-Hernandez
- Biochemistry and Molecular Biology Program, University of Northern British Columbia, Prince George, Canada
| | - Hugues B. Massicotte
- Ecosystem Science and Management Program, University of Northern British Columbia, Prince George, Canada
| | - Keith N. Egger
- Ecosystem Science and Management Program, University of Northern British Columbia, Prince George, Canada
| | - Chow H. Lee
- Biochemistry and Molecular Biology Program, University of Northern British Columbia, Prince George, Canada
| | - Geoffrey W. Payne
- Northern Medical Program, University of Northern British Columbia, Prince George, Canada
- * E-mail:
| |
Collapse
|
10
|
Socha MJ, Segal SS. Microvascular mechanisms limiting skeletal muscle blood flow with advancing age. J Appl Physiol (1985) 2018; 125:1851-1859. [PMID: 30412030 PMCID: PMC6737458 DOI: 10.1152/japplphysiol.00113.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 10/22/2018] [Accepted: 11/06/2018] [Indexed: 02/08/2023] Open
Abstract
Effective oxygen delivery to active muscle fibers requires that vasodilation initiated in distal arterioles, which control flow distribution and capillary perfusion, ascends the resistance network into proximal arterioles and feed arteries, which govern total blood flow into the muscle. With exercise onset, ascending vasodilation reflects initiation and conduction of hyperpolarization along endothelium from arterioles into feed arteries. Electrical coupling of endothelial cells to smooth muscle cells evokes the rapid component of ascending vasodilation, which is sustained by ensuing release of nitric oxide during elevated luminal shear stress. Concomitant sympathetic neural activation inhibits ascending vasodilation by stimulating α-adrenoreceptors on smooth muscle cells to constrict the resistance vasculature. We hypothesized that compromised muscle blood flow in advanced age reflects impaired ascending vasodilation through actions on both cell layers of the resistance network. In the gluteus maximus muscle of old (24 mo) vs. young (4 mo) male mice (corresponding to mid-60s vs. early 20s in humans) inhibition of α-adrenoreceptors in old mice restored ascending vasodilation, whereas even minimal activation of α-adrenoreceptors in young mice attenuated ascending vasodilation in the manner seen with aging. Conduction of hyperpolarization along the endothelium is impaired in old vs. young mice because of "leaky" membranes resulting from the activation of potassium channels by hydrogen peroxide released from endothelial cells. Exposing the endothelium of young mice to hydrogen peroxide recapitulates this effect of aging. Thus enhanced α-adrenoreceptor activation of smooth muscle in concert with electrically leaky endothelium restricts muscle blood flow by impairing ascending vasodilation in advanced age.
Collapse
Affiliation(s)
- Matthew J Socha
- Biology Department, University of Scranton , Scranton, Pennsylvania
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri , Columbia, Missouri
- Dalton Cardiovascular Research Center , Columbia, Missouri
| |
Collapse
|
11
|
Rosenblum WI. Endothelium-dependent responses in the microcirculation observed in vivo. Acta Physiol (Oxf) 2018; 224:e13111. [PMID: 29873936 DOI: 10.1111/apha.13111] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/19/2018] [Accepted: 06/04/2018] [Indexed: 12/17/2022]
Abstract
Endothelium-dependent responses were first demonstrated 40 years ago in the aorta. Since then, extensive research has been conducted in vitro using conductance vessels and materials derived from them. However, the microcirculation controls blood flow to vital organs and has been the focus of in vivo studies of endothelium-dependent dilation beginning immediately after the first in vitro report. Initial in vivo studies employed a light/dye technique for selectively damaging the endothelium to unequivocally prove, in vivo, the existence of endothelium-dependent dilation and in the microvasculature. Endothelium-dependent constriction was similarly proven. Endothelium-dependent agonists include acetylcholine (ACh), bradykinin, arachidonic acid, calcium ionophore A-23187, calcitonin gene-related peptide (CGRP), serotonin, histamine and endothelin-1. Normal and disease states have been studied. Endothelial nitric oxide synthase, cyclooxygenase and cytochrome P450 have been shown to generate the mediators of the responses. Some of the key enzyme systems generate reactive oxygen species (ROS) like superoxide which may prevent EDR. However, one ROS, namely H2 O2 , is one of a number of hyperpolarizing factors that cause dilation initiated by endothelium. Depending upon microvascular bed, a single agonist may use different pathways to elicit an endothelium-dependent response. Interpretation of studies using inhibitors of eNOS is complicated by the fact that these inhibitors may also inhibit ATP-sensitive potassium channels. Other in vivo observations of brain arterioles failed to establish nitric oxide as the mediator of responses elicited by CGRP or by ACh and suggest that a nitrosothiol may be a better fit for the latter.
Collapse
Affiliation(s)
- W. I. Rosenblum
- Department of Pathology; Icahn School of Medicine at Mt Sinai NYC; New York NY USA
| |
Collapse
|
12
|
Castorena-Gonzalez JA, Zawieja SD, Li M, Srinivasan RS, Simon AM, de Wit C, de la Torre R, Martinez-Lemus LA, Hennig GW, Davis MJ. Mechanisms of Connexin-Related Lymphedema. Circ Res 2018; 123:964-985. [PMID: 30355030 PMCID: PMC6771293 DOI: 10.1161/circresaha.117.312576] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
RATIONALE Mutations in GJC2 and GJA1, encoding Cxs (connexins) 47 and 43, respectively, are linked to lymphedema, but the underlying mechanisms are unknown. Because efficient lymph transport relies on the coordinated contractions of lymphatic muscle cells (LMCs) and their electrical coupling through Cxs, Cx-related lymphedema is proposed to result from dyssynchronous contractions of lymphatic vessels. OBJECTIVE To determine which Cx isoforms in LMCs and lymphatic endothelial cells are required for the entrainment of lymphatic contraction waves and efficient lymph transport. METHODS AND RESULTS We developed novel methods to quantify the spatiotemporal entrainment of lymphatic contraction waves and used optogenetic techniques to analyze calcium signaling within and between the LMC and the lymphatic endothelial cell layers. Genetic deletion of the major lymphatic endothelial cell Cxs (Cx43, Cx47, or Cx37) revealed that none were necessary for the synchronization of the global calcium events that triggered propagating contraction waves. We identified Cx45 in human and mouse LMCs as the critical Cx mediating the conduction of pacemaking signals and entrained contractions. Smooth muscle-specific Cx45 deficiency resulted in 10- to 18-fold reduction in conduction speed, partial-to-severe loss of contractile coordination, and impaired lymph pump function ex vivo and in vivo. Cx45 deficiency resulted in profound inhibition of lymph transport in vivo, but only under an imposed gravitational load. CONCLUSIONS Our results (1) identify Cx45 as the Cx isoform mediating the entrainment of the contraction waves in LMCs; (2) show that major endothelial Cxs are dispensable for the entrainment of contractions; (3) reveal a lack of coupling between lymphatic endothelial cells and LMCs, in contrast to arterioles; (4) point to lymphatic valve defects, rather than contraction dyssynchrony, as the mechanism underlying GJC2- or GJA1-related lymphedema; and (5) show that a gravitational load exacerbates lymphatic contractile defects in the intact mouse hindlimb, which is likely critical for the development of lymphedema in the adult mouse.
Collapse
Affiliation(s)
| | - Scott D. Zawieja
- Dept. of Medical Pharmacology and Physiology and University of Missouri School of Medicine
| | - Min Li
- Dept. of Medical Pharmacology and Physiology and University of Missouri School of Medicine
| | - R. Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City OK
| | | | - Cor de Wit
- Institute of Physiology, University of Luebeck, Luebeck Germany
| | | | - Luis A. Martinez-Lemus
- Dept. of Medical Pharmacology and Physiology and University of Missouri School of Medicine
| | | | - Michael J. Davis
- Dept. of Medical Pharmacology and Physiology and University of Missouri School of Medicine
| |
Collapse
|
13
|
Boedtkjer E. Acid-base regulation and sensing: Accelerators and brakes in metabolic regulation of cerebrovascular tone. J Cereb Blood Flow Metab 2018; 38:588-602. [PMID: 28984162 PMCID: PMC5888856 DOI: 10.1177/0271678x17733868] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 08/10/2017] [Accepted: 09/06/2017] [Indexed: 12/29/2022]
Abstract
Metabolic regulation of cerebrovascular tone directs blood flow to areas of increased neuronal activity and during disease states partially compensates for insufficient perfusion by enhancing blood flow in collateral blood vessels. Acid-base disturbances frequently occur as result of enhanced metabolism or insufficient blood supply, but despite definitive evidence that acid-base disturbances alter arterial tone, effects of individual acid-base equivalents and the underlying signaling mechanisms are still being debated. H+ is an important intra- and extracellular messenger that modifies cerebrovascular tone. In addition, low extracellular [HCO3-] promotes cerebrovascular contraction through an endothelium-dependent mechanism. CO2 alters arterial tone development via changes in intra- and extracellular pH but it is still controversial whether CO2 also has direct vasomotor effects. Vasocontractile responses to low extracellular [HCO3-] and acute CO2-induced decreases in intracellular pH can counteract H+-mediated vasorelaxation during metabolic and respiratory acidosis, respectively, and may thereby reduce the risk of capillary damage and cerebral edema that could be consequences of unopposed vasodilation. In this review, the signaling mechanisms for acid-base equivalents in cerebral arteries and the mechanisms of intracellular pH control in the arterial wall are discussed in the context of metabolic regulation of cerebrovascular tone and local perfusion.
Collapse
Affiliation(s)
- Ebbe Boedtkjer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
14
|
Behringer EJ. Calcium and electrical signaling in arterial endothelial tubes: New insights into cellular physiology and cardiovascular function. Microcirculation 2018; 24. [PMID: 27801542 DOI: 10.1111/micc.12328] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/25/2016] [Indexed: 12/23/2022]
Abstract
The integral role of the endothelium during the coordination of blood flow throughout vascular resistance networks has been recognized for several decades now. Early examination of the distinct anatomy and physiology of the endothelium as a signaling conduit along the vascular wall has prompted development and application of an intact endothelial "tube" study model isolated from rodent skeletal muscle resistance arteries. Vasodilatory signals such as increased endothelial cell (EC) Ca2+ ([Ca2+ ]i ) and hyperpolarization take place in single ECs while shared between electrically coupled ECs through gap junctions up to distances of millimeters (≥2 mm). The small- and intermediate-conductance Ca2+ activated K+ (SKCa /IKCa or KCa 2.3/KCa 3.1) channels function at the interface of Ca2+ signaling and hyperpolarization; a bidirectional relationship whereby increases in [Ca2+ ]i activate SKCa /IKCa channels to produce hyperpolarization and vice versa. Further, the spatial domain of hyperpolarization among electrically coupled ECs can be finely tuned via incremental modulation of SKCa /IKCa channels to balance the strength of local and conducted electrical signals underlying vasomotor activity. Multifunctional properties of the voltage-insensitive SKCa /IKCa channels of resistance artery endothelium may be employed for therapy during the aging process and development of vascular disease.
Collapse
Affiliation(s)
- Erik J Behringer
- Department of Basic Sciences, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
15
|
Hill MA, Meininger GA. Small artery mechanobiology: Roles of cellular and non-cellular elements. Microcirculation 2018; 23:611-613. [PMID: 27681605 DOI: 10.1111/micc.12323] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 09/20/2016] [Indexed: 12/15/2022]
Abstract
Small arteries and their component cellular and non-cellular elements are continually subjected to, and interact with, mechanical forces. Such interactions are key in both short- and long-term adaptation of the structure and function of the microcirculation to its local environment and metabolic requirements. Following this brief introduction is a series of papers presented as a symposium (Small Artery Mechanobiology: Roles of Cellular and Non-Cellular Elements) at the World Congress for Microcirculation, Kyoto 2015.
Collapse
Affiliation(s)
- Michael A Hill
- Dalton Cardiovascular Research Center and Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Gerald A Meininger
- Dalton Cardiovascular Research Center and Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
16
|
Behringer EJ, Segal SS. Impact of Aging on Calcium Signaling and Membrane Potential in Endothelium of Resistance Arteries: A Role for Mitochondria. J Gerontol A Biol Sci Med Sci 2017; 72:1627-1637. [PMID: 28510636 DOI: 10.1093/gerona/glx079] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 04/21/2017] [Indexed: 12/20/2022] Open
Abstract
Impaired blood flow to peripheral tissues during advanced age is associated with endothelial dysfunction and diminished bioavailability of nitric oxide (NO). However, it is unknown whether aging impacts coupling between intracellular calcium ([Ca2+]i) signaling and small- and intermediate K+ channel (SKCa/IKCa) activity during endothelium-derived hyperpolarization (EDH), a signaling pathway integral to dilation of the resistance vasculature. To address the potential impact of aging on EDH, Fura-2 photometry and intracellular recording were applied to evaluate [Ca2+]i and membrane potential of intact endothelial tubes (width, 60 µm; length, 1-3 mm) freshly isolated from superior epigastric arteries of young (4-6 mo) and old (24-26 mo) male C57BL/6 mice. In response to acetylcholine, intracellular release of Ca2+ from the endoplasmic reticulum (ER) was enhanced with aging. Further, treatment with the mitochondrial uncoupler FCCP evoked a significant increase of [Ca2+]i with membrane hyperpolarization in an SKCa/IKCa-dependent manner in the endothelium of old but not young mice. We conclude that the ability of resistance artery endothelium to release Ca2+ from intracellular stores (ie, ER and mitochondria) and hyperpolarize Vm via SKCa/IKCa activation is augmented as compensation for reduced NO bioavailability during advanced age.
Collapse
Affiliation(s)
- Erik J Behringer
- Department of Basic Sciences, Loma Linda University, California.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia.,Dalton Cardiovascular Research Center, Columbia, Missouri
| |
Collapse
|
17
|
Namani R, Kassab GS, Lanir Y. Integrative model of coronary flow in anatomically based vasculature under myogenic, shear, and metabolic regulation. J Gen Physiol 2017; 150:145-168. [PMID: 29196421 PMCID: PMC5749109 DOI: 10.1085/jgp.201711795] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 08/23/2017] [Accepted: 10/25/2017] [Indexed: 12/26/2022] Open
Abstract
Coronary blood flow is regulated to match the oxygen demand of myocytes in the heart wall. Flow regulation is essential to meet the wide range of cardiac workload. The blood flows through a complex coronary vasculature of elastic vessels having nonlinear wall properties, under transmural heterogeneous myocardial extravascular loading. To date, there is no fully integrative flow analysis that incorporates global and local passive and flow control determinants. Here, we provide an integrative model of coronary flow regulation that considers the realistic asymmetric morphology of the coronary network, the dynamic myocardial loading on the vessels embedded in it, and the combined effects of local myogenic effect, local shear regulation, and conducted metabolic control driven by venous O2 saturation level. The model predicts autoregulation (approximately constant flow over a wide range of coronary perfusion pressures), reduced heterogeneity of regulated flow, and presence of flow reserve, in agreement with experimental observations. Furthermore, the model shows that the metabolic and myogenic regulations play a primary role, whereas shear has a secondary one. Regulation was found to have a significant effect on the flow except under extreme (high and low) inlet pressures and metabolic demand. Novel outcomes of the model are that cyclic myocardial loading on coronary vessels enhances the coronary flow reserve except under low inlet perfusion pressure, increases the pressure range of effective autoregulation, and reduces the network flow in the absence of metabolic regulation. Collectively, these findings demonstrate the utility of the present biophysical model, which can be used to unravel the underlying mechanisms of coronary physiopathology.
Collapse
Affiliation(s)
- Ravi Namani
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | | | - Yoram Lanir
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
18
|
Abstract
Hypoxic pulmonary vasoconstriction (HPV) in combination with hypercapnic pulmonary vasoconstriction redistributes pulmonary blood flow from poorly aerated to better ventilated lung regions by an active process of local vasoconstriction. Impairment of HPV results in ventilation-perfusion mismatch and is commonly associated with various lung diseases including pneumonia, sepsis, or cystic fibrosis. Although several regulatory pathways have been identified, considerable knowledge gaps persist, and a unifying concept of the signaling pathways that underlie HPV and their impairment in lung diseases has not yet emerged. In the past, conceptual models of HPV have focused on pulmonary arterial smooth muscle cells (PASMC) acting as sensor and effector of hypoxia in the pulmonary vasculature. In contrast, the endothelium was considered a modulating bystander in this scenario. For an ideal design, however, the oxygen sensor in HPV should be located in the region of gas exchange, i.e., in the alveolar capillary network. This concept requires the retrograde propagation of the hypoxic signal along the endothelial layer of the vascular wall and subsequent contraction of PASMC in upstream arterioles that is elicited via temporospatially tightly controlled endothelial-smooth muscle cell crosstalk. The present review summarizes recent work that provides proof-of-principle for the existence and functional relevance of such signaling pathway in HPV that involves important roles for connexin 40, epoxyeicosatrienoic acids, sphingolipids, and cystic fibrosis transmembrane conductance regulator. Of translational relevance, implication of these molecules provides for novel mechanistic explanations for impaired ventilation/perfusion matching in patients with pneumonia, sepsis, cystic fibrosis, and presumably various other lung diseases.
Collapse
Affiliation(s)
- Benjamin Grimmer
- Institute of Physiology, Charité Universitätsmedizin Berlin, Berlin , Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité Universitätsmedizin Berlin, Berlin , Germany
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital , Toronto, Ontario , Canada
- Departments of Surgery and Physiology, University of Toronto , Toronto, Ontario , Canada
| |
Collapse
|
19
|
Sinkler SY, Segal SS. Rapid versus slow ascending vasodilatation: intercellular conduction versus flow-mediated signalling with tetanic versus rhythmic muscle contractions. J Physiol 2017; 595:7149-7165. [PMID: 28981145 DOI: 10.1113/jp275186] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 09/28/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS In response to exercise, vasodilatation ascends from downstream arterioles into upstream feed arteries (FAs). We hypothesized that the signalling events underlying ascending vasodilatation variy with the intensity and duration of skeletal muscle contraction. In the gluteus maximus muscle of C57BL/6 mice, brief tetanic contraction evoked rapid onset vasodilatation (ROV) (<1 s) throughout the resistance network. Selective damage to endothelium midway between FAs and primary arterioles eliminated ROV only in FAs. Blocking SKCa and IKCa channels attenuated ROV, implicating hyperpolarization as the underlying signal. During rhythmic twitch contractions, slow onset vasodilatation (10-15 s) in FAs remained intact following loss of ROV and was eliminated following nitric oxide synthase inhibition. Tetanic contraction initiates hyperpolarization that conducts along endothelium into FAs. Rhythmic twitch contractions stimulate FA endothelium to release nitric oxide in response to elevated shear stress secondary to metabolic dilatation of arterioles. Complementary endothelial signalling pathways for ascending vasodilatation ensure increased oxygen delivery to active skeletal muscle. ABSTRACT In response to exercise, vasodilatation initiated within the microcirculation of skeletal muscle ascends the resistance network into upstream feed arteries (FAs) located external to the tissue. Ascending vasodilatation (AVD) is essential for reducing FA resistance that otherwise restricts blood flow into the microcirculation. In the present study, we tested the hypothesis that signalling events underlying AVD vary with the intensity and duration of muscle contraction. In the gluteus maximus muscle of anaesthetized male C57BL/6 mice (aged 3-4 months), brief tetanic contraction (100 Hz for 500 ms) evoked rapid onset vasodilatation (ROV) in FAs that peaked within 4 s. By contrast, during rhythmic twitch contractions (4 Hz), slow onset vasodilatation (SOV) of FAs began after ∼10 s and plateaued within 30 s. Selectively damaging the endothelium with light-dye treatment midway between a FA and its primary arteriole eliminated ROV in the FA along with conducted vasodilatation of the FA initiated on the arteriole using ACh microiontophoresis. Superfusion of SKCa and IKCa channel blockers UCL 1684 + TRAM 34 attenuated ROV, implicating endothelial hyperpolarization as the underlying signal. Nevertheless, the SOV of FAs during rhythmic contractions persisted until inhibition of nitric oxide synthase with Nω -nitro-l-arginine methyl ester. Thus, ROV of FAs reflects hyperpolarization of downstream arterioles that conducts along the endothelium into proximal FAs. By contrast, SOV of FAs reflects the local production of nitric oxide by the endothelium in response to luminal shear stress, which increases secondary to arteriolar dilatation downstream. Thus, AVD ensures increased oxygen delivery to active muscle fibres by reducing upstream resistance via complementary signalling pathways that reflect the intensity and duration of muscle contraction.
Collapse
Affiliation(s)
- Shenghua Y Sinkler
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA.,Dalton Cardiovascular Research Center, Columbia, MO, USA
| |
Collapse
|
20
|
Welsh DG, Tran CHT, Hald BO, Sancho M. The Conducted Vasomotor Response: Function, Biophysical Basis, and Pharmacological Control. Annu Rev Pharmacol Toxicol 2017; 58:391-410. [PMID: 28968190 DOI: 10.1146/annurev-pharmtox-010617-052623] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Arterial tone is coordinated among vessel segments to optimize nutrient transport and organ function. Coordinated vasomotor activity is remarkable to observe and depends on stimuli, sparsely generated in tissue, eliciting electrical responses that conduct lengthwise among electrically coupled vascular cells. The conducted response is the focus of this topical review, and in this regard, the authors highlight literature that advances an appreciation of functional significance, cellular mechanisms, and biophysical principles. Of particular note, this review stresses that conduction is enabled by a defined pattern of charge movement along the arterial wall as set by three key parameters (tissue structure, gap junctional resistivity, and ion channel activity). The impact of disease on conduction is carefully discussed, as are potential strategies to restore this key biological response and, along with it, the match of blood flow delivery with tissue energetic demand.
Collapse
Affiliation(s)
- Donald G Welsh
- Robarts Research Institute, Department of Physiology and Pharmacology, Schulich School of Medicine, University of Western Ontario, London, Ontario N6A 5B7, Canada;
| | - Cam Ha T Tran
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Bjorn O Hald
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Maria Sancho
- Robarts Research Institute, Department of Physiology and Pharmacology, Schulich School of Medicine, University of Western Ontario, London, Ontario N6A 5B7, Canada;
| |
Collapse
|
21
|
Komarova YA, Kruse K, Mehta D, Malik AB. Protein Interactions at Endothelial Junctions and Signaling Mechanisms Regulating Endothelial Permeability. Circ Res 2017; 120:179-206. [PMID: 28057793 DOI: 10.1161/circresaha.116.306534] [Citation(s) in RCA: 336] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 12/31/2022]
Abstract
The monolayer of endothelial cells lining the vessel wall forms a semipermeable barrier (in all tissue except the relatively impermeable blood-brain and inner retinal barriers) that regulates tissue-fluid homeostasis, transport of nutrients, and migration of blood cells across the barrier. Permeability of the endothelial barrier is primarily regulated by a protein complex called adherens junctions. Adherens junctions are not static structures; they are continuously remodeled in response to mechanical and chemical cues in both physiological and pathological settings. Here, we discuss recent insights into the post-translational modifications of junctional proteins and signaling pathways regulating plasticity of adherens junctions and endothelial permeability. We also discuss in the context of what is already known and newly defined signaling pathways that mediate endothelial barrier leakiness (hyperpermeability) that are important in the pathogenesis of cardiovascular and lung diseases and vascular inflammation.
Collapse
Affiliation(s)
- Yulia A Komarova
- From the Department of Pharmacology and the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Kevin Kruse
- From the Department of Pharmacology and the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Dolly Mehta
- From the Department of Pharmacology and the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Asrar B Malik
- From the Department of Pharmacology and the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago.
| |
Collapse
|
22
|
Dora KA. Conducted dilatation to ATP and K + in rat skeletal muscle arterioles. Acta Physiol (Oxf) 2017; 219:202-218. [PMID: 26804547 PMCID: PMC5215486 DOI: 10.1111/apha.12656] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/25/2015] [Accepted: 01/19/2016] [Indexed: 11/28/2022]
Abstract
AIM During exercise in humans, circulating levels of ATP and K+ increase at a time when blood flow increases to satisfy metabolic demand. Both molecules can activate arteriolar K+ channels to stimulate vasodilatation; here, it is established whether conducted dilatation is observed in a skeletal muscle bed. METHODS Isolated and cannulated rat cremaster arterioles were used to assess both local and conducted responses. Agents were either added to the bath, focally pulse-ejected to the downstream end of arterioles, or in triple-cannulated arterioles, luminally perfused into the downstream branches to assess both local and conducted responses. RESULTS The endothelium-dependent agonist ACh and the KATP channel opener levcromakalim each stimulated both local and conducted vasodilatation. Focal, bolus delivery of ATP (10 μm) or KCl (33 mm) to the outside of arterioles stimulated a biphasic vasomotor response: rapid vasoconstriction followed by dilatation as each washed away. At lower concentrations of KCl (19 mm), constriction was avoided, and instead, Ba2+ -sensitive local dilatation and conducted dilatation were both observed. Luminal perfusion of ATP avoided constriction and activated P2Y1 receptors stimulating vasodilatation secondary to opening of KCa channels. In triple-cannulated arterioles, either ATP (10 μm) or K+ (15 mm) luminally perfused into daughter branches of a bifurcation stimulated local dilatation which conducted into the parent arteriole. CONCLUSION The recognized physiological autocrine and paracrine mediators ATP and K+ each act to evoke both local and conducted vasodilatation in rat cremaster arterioles. Therefore, in situations when circulating levels are raised, such as during exercise, these agents can act as important regulators of blood flow.
Collapse
Affiliation(s)
- K. A. Dora
- Department of PharmacologyUniversity of OxfordOxfordUK
| |
Collapse
|
23
|
Schmidt K, Windler R, de Wit C. Communication Through Gap Junctions in the Endothelium. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 77:209-40. [PMID: 27451099 DOI: 10.1016/bs.apha.2016.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A swarm of fish displays a collective behavior (swarm behavior) and moves "en masse" despite the huge number of individual animals. In analogy, organ function is supported by a huge number of cells that act in an orchestrated fashion and this applies also to vascular cells along the vessel length. It is obvious that communication is required to achieve this vital goal. Gap junctions with their modular bricks, connexins (Cxs), provide channels that interlink the cytosol of adjacent cells by a pore sealed against the extracellular space. This allows the transfer of ions and charge and thereby the travel of membrane potential changes along the vascular wall. The endothelium provides a low-resistance pathway that depends crucially on connexin40 which is required for long-distance conduction of dilator signals in the microcirculation. The experimental evidence for membrane potential changes synchronizing vascular behavior is manifold but the functional verification of a physiologic role is still open. Other molecules may also be exchanged that possibly contribute to the synchronization (eg, Ca(2+)). Recent data suggest that vascular Cxs have more functions than just facilitating communication. As pharmacological tools to modulate gap junctions are lacking, Cx-deficient mice provide currently the standard to unravel their vascular functions. These include arteriolar dilation during functional hyperemia, hypoxic pulmonary vasoconstriction, vascular collateralization after ischemia, and feedback inhibition on renin secretion in the kidney.
Collapse
Affiliation(s)
- K Schmidt
- Institut für Physiologie, Universität zu Lübeck, Lübeck, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - R Windler
- Institut für Physiologie, Universität zu Lübeck, Lübeck, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - C de Wit
- Institut für Physiologie, Universität zu Lübeck, Lübeck, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany.
| |
Collapse
|
24
|
Longden TA, Hill-Eubanks DC, Nelson MT. Ion channel networks in the control of cerebral blood flow. J Cereb Blood Flow Metab 2016; 36:492-512. [PMID: 26661232 PMCID: PMC4794103 DOI: 10.1177/0271678x15616138] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 09/17/2015] [Accepted: 10/14/2015] [Indexed: 12/26/2022]
Abstract
One hundred and twenty five years ago, Roy and Sherrington made the seminal observation that neuronal stimulation evokes an increase in cerebral blood flow.(1) Since this discovery, researchers have attempted to uncover how the cells of the neurovascular unit-neurons, astrocytes, vascular smooth muscle cells, vascular endothelial cells and pericytes-coordinate their activity to control this phenomenon. Recent work has revealed that ionic fluxes through a diverse array of ion channel species allow the cells of the neurovascular unit to engage in multicellular signaling processes that dictate local hemodynamics.In this review we center our discussion on two major themes: (1) the roles of ion channels in the dynamic modulation of parenchymal arteriole smooth muscle membrane potential, which is central to the control of arteriolar diameter and therefore must be harnessed to permit changes in downstream cerebral blood flow, and (2) the striking similarities in the ion channel complements employed in astrocytic endfeet and endothelial cells, enabling dual control of smooth muscle from either side of the blood-brain barrier. We conclude with a discussion of the emerging roles of pericyte and capillary endothelial cell ion channels in neurovascular coupling, which will provide fertile ground for future breakthroughs in the field.
Collapse
Affiliation(s)
- Thomas A Longden
- Department of Pharmacology, University of Vermont, Burlington, VT, USA
| | | | - Mark T Nelson
- Department of Pharmacology, University of Vermont, Burlington, VT, USA Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
25
|
Li L, Zhang W, Shi WY, Ma KT, Zhao L, Wang Y, Zhang L, Li XZ, Zhu H, Zhang ZS, Liu WD, Si JQ. The enhancement of Cx45 expression and function in renal interlobar artery of spontaneously hypertensive rats at different age. Kidney Blood Press Res 2016; 40:52-65. [PMID: 25791497 DOI: 10.1159/000368482] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS This study was designed to investigate the expression and function of gap junction protein connexin 45 (Cx45) in renal interlobar artery (RIA) of spontaneously hypertensive rats (SHR), and the association between hypertension and enhanced vasoconstrictive response in SHR. METHODS Western blot analysis and pressure myography were used to examine the differences in expression and function of Cx45 in vascular smooth muscle cells (VSMCs) of RIA between SHR and normotensive Wistar-Kyoto (WKY) rats. RESULTS Our results demonstrated that 1) whole-cell patch clamp measurements showed that the membrane capacitance and conductance of in-situ RIA VSMCs of SHR were significantly greater than those of WKY rats (p<0.05, n=6), suggesting that the coupling of gap junction between VSMCs of RIA was enhanced in SHR; 2) the KCl or phenylephrine (PE)-stimulated RIA constriction was more pronounced in SHR than that in WKY rats (p<0.05, n=10). After applying a gap junction inhibitor 18β-glycyrrhetintic acid (18β-GA), the inhibitory effect of 18β-GA on KCl or PE-induced vasoconstriction was greater in SHR (p<0.05, n=10); and 3) the expression of Cx45 in RIA of SHR was greater than that in WKY rats (p<0.05, n=3) at 4, 12 and 48 wks of age. CONCLUSIONS The hypertension-induced elevation of Cx45 may affect communication between VSMCs and coupling between VSMCs and endothelium, which results in an increased vasoconstrictive response in renal artery and might contribute to the development of hypertension.
Collapse
|
26
|
Segal SS. Integration and Modulation of Intercellular Signaling Underlying Blood Flow Control. J Vasc Res 2015; 52:136-57. [PMID: 26368324 DOI: 10.1159/000439112] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 07/30/2015] [Indexed: 01/25/2023] Open
Abstract
Vascular resistance networks control tissue blood flow in concert with regulating arterial perfusion pressure. In response to increased metabolic demand, vasodilation arising in arteriolar networks ascends to encompass proximal feed arteries. By reducing resistance upstream, ascending vasodilation (AVD) increases blood flow into the microcirculation. Once initiated, e.g. through local activation of K(+) channels in endothelial cells (ECs), hyperpolarization is conducted through gap junctions along the endothelium. Via EC projections through the internal elastic lamina, hyperpolarization spreads into the surrounding smooth-muscle cells (SMCs) through myoendothelial gap junctions (MEGJs) to promote their relaxation. Intercellular signaling through electrical signal transmission (i.e. cell-to-cell conduction) can thereby coordinate vasodilation along and among the branches of microvascular resistance networks. Perivascular sympathetic nerve fibers course through the adventitia and release norepinephrine to stimulate SMCs via α-adrenoreceptors to produce contraction. In turn, SMCs can signal ECs through MEGJs to activate K(+) channels and attenuate sympathetic vasoconstriction. Activation of K(+) channels along the endothelium will dissipate electrical signal transmission and inhibit AVD, thereby restricting blood flow into the microcirculation while maintaining peripheral resistance and perfusion pressure. This review explores the origins and nature of the intercellular signaling that governs blood flow control in skeletal muscle with respect to the interplay between AVD and sympathetic innervation. Whereas these interactions are integral to daily activity and athletic performance, determining the interplay between respective signaling events provides insight into how selective interventions can improve tissue perfusion and oxygen delivery during vascular disease.
Collapse
|
27
|
Mutchler SM, Straub AC. Compartmentalized nitric oxide signaling in the resistance vasculature. Nitric Oxide 2015; 49:8-15. [PMID: 26028569 DOI: 10.1016/j.niox.2015.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 05/15/2015] [Accepted: 05/20/2015] [Indexed: 01/23/2023]
Abstract
Nitric oxide (NO) was first described as a bioactive molecule through its ability to stimulate soluble guanylate cyclase, but the revelation that NO was the endothelium derived relaxation factor drove the field to its modern state. The wealth of research conducted over the past 30 years has provided us with a picture of how diverse NO signaling can be within the vascular wall, going beyond simple vasodilation to include such roles as signaling through protein S-nitrosation. This expanded view of NO's actions requires highly regulated and compartmentalized production. Importantly, resistance arteries house multiple proteins involved in the production and transduction of NO allowing for efficient movement of the molecule to regulate vascular tone and reactivity. In this review, we focus on the many mechanisms regulating NO production and signaling action in the vascular wall, with a focus on the control of endothelial nitric oxide synthase (eNOS), the enzyme responsible for synthesizing most of the NO within these confines. We also explore how cross talk between the endothelium and smooth muscle in the microcirculation can modulate NO signaling, illustrating that this one small molecule has the capability to produce a plethora of responses.
Collapse
Affiliation(s)
- Stephanie M Mutchler
- Heart, Lung, Blood and Vascular Medicine Institute, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Adam C Straub
- Heart, Lung, Blood and Vascular Medicine Institute, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15216, USA.
| |
Collapse
|
28
|
Straub AC, Zeigler AC, Isakson BE. The myoendothelial junction: connections that deliver the message. Physiology (Bethesda) 2015; 29:242-9. [PMID: 24985328 DOI: 10.1152/physiol.00042.2013] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A vast amount of investigation has centered on how the endothelium and smooth muscle communicate. From this evidence, myoendothelial junctions have emerged as critical anatomical structures to regulate heterocellular cross talk. Indeed, there is now evidence that the myoendothelial junction serves as a signaling microdomain to organize proteins used to facilitate vascular heterocellular communication. This review highlights the evolving role of myoendothelial junctions in the context of vascular cell-cell communication.
Collapse
Affiliation(s)
- Adam C Straub
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Angela C Zeigler
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia; and
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia; and Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
29
|
Rodrigues SF, Granger DN. Blood cells and endothelial barrier function. Tissue Barriers 2015; 3:e978720. [PMID: 25838983 DOI: 10.4161/21688370.2014.978720] [Citation(s) in RCA: 205] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/15/2014] [Indexed: 12/18/2022] Open
Abstract
The barrier properties of endothelial cells are critical for the maintenance of water and protein balance between the intravascular and extravascular compartments. An impairment of endothelial barrier function has been implicated in the genesis and/or progression of a variety of pathological conditions, including pulmonary edema, ischemic stroke, neurodegenerative disorders, angioedema, sepsis and cancer. The altered barrier function in these conditions is often linked to the release of soluble mediators from resident cells (e.g., mast cells, macrophages) and/or recruited blood cells. The interaction of the mediators with receptors expressed on the surface of endothelial cells diminishes barrier function either by altering the expression of adhesive proteins in the inter-endothelial junctions, by altering the organization of the cytoskeleton, or both. Reactive oxygen species (ROS), proteolytic enzymes (e.g., matrix metalloproteinase, elastase), oncostatin M, and VEGF are part of a long list of mediators that have been implicated in endothelial barrier failure. In this review, we address the role of blood borne cells, including, neutrophils, lymphocytes, monocytes, and platelets, in the regulation of endothelial barrier function in health and disease. Attention is also devoted to new targets for therapeutic intervention in disease states with morbidity and mortality related to endothelial barrier dysfunction.
Collapse
Key Words
- AJ, Adherens junctions
- ANG-1, Angiopoietin 1
- AQP, Aquaporins
- BBB, blood brain barrier
- CNS, Central nervous system
- COPD, Chronic obstructive pulmonary disease
- EAE, Experimental autoimmune encephalomyelitis
- EPAC1, Exchange protein activated by cyclic AMP
- ERK1/2, Extracellular signal-regulated kinases 1 and 2
- Endothelial barrier
- FA, Focal adhesions
- FAK, focal adhesion tyrosine kinase
- FoxO1, Forkhead box O1
- GAG, Glycosaminoglycans
- GDNF, Glial cell-derived neurotrophic factor
- GJ, Gap junctions
- GPCR, G-protein coupled receptors
- GTPase, Guanosine 5'-triphosphatase
- HMGB-1, High mobility group box 1
- HRAS, Harvey rat sarcoma viral oncogene homolog
- ICAM-1, Intercellular adhesion molecule 1
- IL-1β, Interleukin 1 beta
- IP3, Inositol 1,4,5-triphosphate
- JAM, Junctional adhesion molecules
- MEK, Mitogen-activated protein kinase kinase
- MLC, Myosin light chain
- MLCK, Myosin light-chain kinase
- MMP, Matrix metalloproteinases
- NO, Nitric oxide
- OSM, Oncostatin M
- PAF, Platelet activating factor
- PDE, Phosphodiesterase
- PKA, Protein kinase A
- PNA, Platelet-neutrophil aggregates
- ROS, Reactive oxygen species
- Rac1, Ras-related C3 botulinum toxin substrate 1
- Rap1, Ras-related protein 1
- RhoA, Ras homolog gene family, member A
- S1P, Sphingosine-1-phosphate
- SCID, Severe combined immunodeficient
- SOCS-3, Suppressors of cytokine signaling 3
- Shp-2, Src homology 2 domain-containing phosphatase 2
- Src, Sarcoma family of protein kinases
- TEER, Transendothelial electrical resistance
- TGF-beta1, Transforming growth factor-beta1
- TJ, Tight junctions
- TNF-, Tumor necrosis factor alpha
- VCAM-1, Vascular cell adhesion molecule 1
- VE, Vascular endothelial
- VE-PTP, Vascular endothelial receptor protein tyrosine phosphatase
- VEGF, Vascular endothelial growth factor
- VVO, Vesiculo-vacuolar organelle
- ZO, Zonula occludens
- cAMP, 3'-5'-cyclic adenosine monophosphate
- erythrocytes
- leukocytes
- pSrc, Phosphorylated Src
- platelets
- vascular permeability
Collapse
Affiliation(s)
- Stephen F Rodrigues
- Department of Clinical and Toxicological Analyses; School of Pharmaceutical Sciences; University of Sao Paulo ; Sao Paulo, Brazil
| | - D Neil Granger
- Department of Molecular and Cellular Physiology; Louisiana State University Health Sciences Center ; Shreveport, LA USA
| |
Collapse
|
30
|
Billaud M, Lohman AW, Johnstone SR, Biwer LA, Mutchler S, Isakson BE. Regulation of cellular communication by signaling microdomains in the blood vessel wall. Pharmacol Rev 2014; 66:513-69. [PMID: 24671377 PMCID: PMC3973613 DOI: 10.1124/pr.112.007351] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
It has become increasingly clear that the accumulation of proteins in specific regions of the plasma membrane can facilitate cellular communication. These regions, termed signaling microdomains, are found throughout the blood vessel wall where cellular communication, both within and between cell types, must be tightly regulated to maintain proper vascular function. We will define a cellular signaling microdomain and apply this definition to the plethora of means by which cellular communication has been hypothesized to occur in the blood vessel wall. To that end, we make a case for three broad areas of cellular communication where signaling microdomains could play an important role: 1) paracrine release of free radicals and gaseous molecules such as nitric oxide and reactive oxygen species; 2) role of ion channels including gap junctions and potassium channels, especially those associated with the endothelium-derived hyperpolarization mediated signaling, and lastly, 3) mechanism of exocytosis that has considerable oversight by signaling microdomains, especially those associated with the release of von Willebrand factor. When summed, we believe that it is clear that the organization and regulation of signaling microdomains is an essential component to vessel wall function.
Collapse
Affiliation(s)
- Marie Billaud
- Dept. of Molecular Physiology and Biophysics, University of Virginia School of Medicine, PO Box 801394, Charlottesville, VA 22902.
| | | | | | | | | | | |
Collapse
|
31
|
Lohman AW, Isakson BE. Differentiating connexin hemichannels and pannexin channels in cellular ATP release. FEBS Lett 2014; 588:1379-88. [PMID: 24548565 DOI: 10.1016/j.febslet.2014.02.004] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Revised: 02/06/2014] [Accepted: 02/06/2014] [Indexed: 12/22/2022]
Abstract
Adenosine triphosphate (ATP) plays a fundamental role in cellular communication, with its extracellular accumulation triggering purinergic signaling cascades in a diversity of cell types. While the roles for purinergic signaling in health and disease have been well established, identification and differentiation of the specific mechanisms controlling cellular ATP release is less well understood. Multiple mechanisms have been proposed to regulate ATP release with connexin (Cx) hemichannels and pannexin (Panx) channels receiving major focus. However, segregating the specific roles of Panxs and Cxs in ATP release in a plethora of physiological and pathological contexts has remained enigmatic. This multifaceted problem has arisen from the selectivity of pharmacological inhibitors for Panxs and Cxs, methodological differences in assessing Panx and Cx function and the potential compensation by other isoforms in gene silencing and genetic knockout models. Consequently, there remains a void in the current understanding of specific contributions of Panxs and Cxs in releasing ATP during homeostasis and disease. Differentiating the distinct signaling pathways that regulate these two channels will advance our current knowledge of cellular communication and aid in the development of novel rationally-designed drugs for modulation of Panx and Cx activity, respectively.
Collapse
Affiliation(s)
- Alexander W Lohman
- Department of Molecular Physiology and Biophysics, University of Virginia, Charlottesville, VA 22098, United States; Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, United States
| | - Brant E Isakson
- Department of Molecular Physiology and Biophysics, University of Virginia, Charlottesville, VA 22098, United States; Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, United States.
| |
Collapse
|
32
|
Veeranki S, Tyagi SC. Defective homocysteine metabolism: potential implications for skeletal muscle malfunction. Int J Mol Sci 2013; 14:15074-91. [PMID: 23873298 PMCID: PMC3742288 DOI: 10.3390/ijms140715074] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 06/24/2013] [Accepted: 07/11/2013] [Indexed: 12/13/2022] Open
Abstract
Hyperhomocysteinemia (HHcy) is a systemic medical condition and has been attributed to multi-organ pathologies. Genetic, nutritional, hormonal, age and gender differences are involved in abnormal homocysteine (Hcy) metabolism that produces HHcy. Homocysteine is an intermediate for many key processes such as cellular methylation and cellular antioxidant potential and imbalances in Hcy production and/or catabolism impacts gene expression and cell signaling including GPCR signaling. Furthermore, HHcy might damage the vagus nerve and superior cervical ganglion and affects various GPCR functions; therefore it can impair both the parasympathetic and sympathetic regulation in the blood vessels of skeletal muscle and affect long-term muscle function. Understanding cellular targets of Hcy during HHcy in different contexts and its role either as a primary risk factor or as an aggravator of certain disease conditions would provide better interventions. In this review we have provided recent Hcy mediated mechanistic insights into different diseases and presented potential implications in the context of reduced muscle function and integrity. Overall, the impact of HHcy in various skeletal muscle malfunctions is underappreciated; future studies in this area will provide deeper insights and improve our understanding of the association between HHcy and diminished physical function.
Collapse
Affiliation(s)
- Sudhakar Veeranki
- Authors to whom correspondence should be addressed; E-Mails: (S.V.); (S.C.T.); Tel.: +1-973-610-1160 (S.V.); +1-502-852-3381 (S.C.T.); Fax: +1-502-852-6239 (S.C.T.)
| | - Suresh C. Tyagi
- Authors to whom correspondence should be addressed; E-Mails: (S.V.); (S.C.T.); Tel.: +1-973-610-1160 (S.V.); +1-502-852-3381 (S.C.T.); Fax: +1-502-852-6239 (S.C.T.)
| |
Collapse
|
33
|
Givvimani S, Narayanan N, Armaghan F, Pushpakumar S, Tyagi SC. Attenuation of conducted vasodilation in skeletal muscle arterioles during hyperhomocysteinemia. Pharmacology 2013; 91:287-96. [PMID: 23736684 DOI: 10.1159/000350394] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 03/03/2013] [Indexed: 11/19/2022]
Abstract
BACKGROUND Vasomotor responses conducted from terminal arterioles to proximal vessels may contribute to match tissue demands and blood supply during skeletal muscle contraction. Conduction of vasodilatation (CVD) from distal resistance arterioles to the proximal arterioles and feeding arteries during metabolic demand is mediated by intercellular gap junctions in the vascular endothelium. The role of hyperhomocysteinemia (HHcy) in the musculoskeletal system during CVD is unclear. We hypothesize that during HHcy, there is impaired CVD due to decreased expression of endothelial-associated connexins and thus decreased tissue perfusion to the contracting skeletal muscles. METHODS CVD studies were performed in a gluteus maximus muscle preparation of wild-type (C57BL6/J) and CBS-/+ (HHcy) mice using intravital microscopy. Expression of connexins and myostatin protein (an antiskeletal muscle statin) was studied by Western blot and immunohistochemistry methods. Tissue perfusion to acetylcholine was assessed by the laser Doppler technique. RESULTS There was decreased CVD and tissue perfusion in response to acetylcholine in CBS-/+ mice compared to wild-type controls. There was decreased expression of connexins 37, 40 and 43 and increased expression of myostatin in CBS-/+ mice compared to wild-type controls. CONCLUSION Our findings suggest that CVD in skeletal muscle is decreased during HHcy due to decreased expression of gap junction connexins.
Collapse
Affiliation(s)
- S Givvimani
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, Ky., USA.
| | | | | | | | | |
Collapse
|
34
|
Behringer EJ, Shaw RL, Westcott EB, Socha MJ, Segal SS. Aging impairs electrical conduction along endothelium of resistance arteries through enhanced Ca2+-activated K+ channel activation. Arterioscler Thromb Vasc Biol 2013; 33:1892-901. [PMID: 23723370 DOI: 10.1161/atvbaha.113.301514] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Intercellular conduction of electrical signals underlies spreading vasodilation of resistance arteries. Small- and intermediate-conductance Ca(2+)-activated K(+) channels of endothelial cells serve a dual function by initiating hyperpolarization and modulating electrical conduction. We tested the hypothesis that regulation of electrical signaling by small- and intermediate-conductance Ca(2+)-activated K(+) channels is altered with advancing age. APPROACH AND RESULTS Intact endothelial tubes (60 µm wide; 1-3 mm long) were freshly isolated from male C57BL/6 mouse (Young: 4-6 months; Intermediate: 12-14 months; Old: 24-26 months) superior epigastric arteries. Using dual intracellular microelectrodes, current was injected (± 0.1-3 nA) at site 1 while recording membrane potential (Vm) at site 2 (separation distance: 50-2000 µm). Across age groups, greatest differences were observed between Young and Old. Resting Vm in Old (-38 ± 1 mV) was more negative (P<0.05) than Young (-30 ± 1 mV). Maximal hyperpolarization to both direct (NS309) and indirect (acetylcholine) activation of small- and intermediate-conductance Ca(2+)-activated K(+) channels was sustained (ΔVm ≈-40 mV) with age. The length constant (λ) for electrical conduction was reduced (P<0.05) from 1630 ± 80 µm (Young) to 1320 ± 80 µm (Old). Inhibiting small- and intermediate-conductance Ca(2+)-activated K(+) channels with apamin+charybdotoxin or scavenging hydrogen peroxide (H2O2) with catalase improved electrical conduction (P<0.05) in Old. Exogenous H2O2 (200 µmol/L) in Young evoked hyperpolarization and impaired electrical conduction; these effects were blocked by apamin+charybdotoxin. CONCLUSIONS Enhanced current loss through Ca2+-activated K+ channel activation impairs electrical conduction along the endothelium of resistance arteries with aging. Attenuating the spatial domain of electrical signaling will restrict the spread of vasodilation and thereby contribute to blood flow limitations associated with advanced age.
Collapse
Affiliation(s)
- Erik J Behringer
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA
| | | | | | | | | |
Collapse
|
35
|
Socha MJ, Domeier TL, Behringer EJ, Segal SS. Coordination of intercellular Ca(2+) signaling in endothelial cell tubes of mouse resistance arteries. Microcirculation 2013; 19:757-70. [PMID: 22860994 DOI: 10.1111/micc.12000] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 08/01/2012] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To test the hypothesis that Ca(2+) responses to GPCR activation are coordinated between neighboring ECs of resistance arteries. METHODS EC tubes were freshly isolated from superior epigastric arteries of C57BL/6 mice. Intercellular coupling was tested using microinjection of propidium iodide. Following loading with fluo-4 dye, intracellular Ca(2+) responses to ACh were imaged with confocal microscopy. RESULTS Cell-to-cell transfer of propidium iodide confirmed functional GJCs. A 1 μm ACh stimulus evoked Ca(2+) responses (9.8 ± 0.8/min, F/F(0) = 3.11 ± 0.2) which pseudo-line-scan analysis revealed as composed of Ca(2+) waves and spatially restricted Ca(2+) release events. A 100 nm ACh stimulus induced Ca(2+) responses of lower frequency (4.5 ± 0.7/min) and amplitude (F/F(0) = 1.95 ± 0.11) composed primarily of spatially restricted events. The time interval between Ca(2+) waves in adjacent cells (0.79 ± 0.12 s) was shorter (p < 0.05) than that between nonadjacent cells (1.56 ± 0.25 s). Spatially restricted Ca(2+) release events had similar frequencies and latencies between adjacent and nonadjacent cells. Inhibiting intracellular Ca(2+) release with 2-APB, Xestospongin C or thapsigargin eliminated Ca(2+) responses. CONCLUSIONS With moderate GPCR stimulation, localized Ca(2+) release events predominate among cells. Greater GPCR stimulation evokes coordinated intercellular Ca(2+) waves via the ER. Calcium signaling during GPCR activation is complex among cells, varying with stimulus intensity and proximity to actively signaling cells.
Collapse
Affiliation(s)
- Matthew J Socha
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri 65212, USA
| | | | | | | |
Collapse
|
36
|
Willcox JM, Summerlee AJS, Murrant CL. Relaxin induces rapid, transient vasodilation in the microcirculation of hamster skeletal muscle. J Endocrinol 2013; 218:179-91. [PMID: 23720398 DOI: 10.1530/joe-13-0115] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Relaxin produces a sustained decrease in total peripheral resistance, but the effects of relaxin on skeletal muscle arterioles, an important contributor to systemic resistance, are unknown. Using the intact, blood-perfused hamster cremaster muscle preparation in situ, we tested the effects of relaxin on skeletal muscle arteriolar microvasculature by applying 10(-10) M relaxin to second-, third- and fourth-order arterioles and capillaries. The mechanisms responsible for relaxin-induced dilations were explored by applying 10(-10) M relaxin to second-order arterioles in the presence of 10(-5) M N(G)-nitro-l-arginine methyl ester (l-NAME, nitric oxide (NO) synthase inhibitor), 10(-5) M glibenclamide (GLIB, ATP-dependent potassium (K(+)) channel inhibitor), 10(-3) M tetraethylammonium (TEA) or 10(-7) M iberiotoxin (IBTX, calcium-associated K(+) channel inhibitor). Relaxin caused second- (peak change in diameter: 8.3 ± 1.7 μm) and third (4.5 ± 1.1 μm)-order arterioles to vasodilate transiently while fourth-order arterioles did not (0.01 ± 0.04 μm). Relaxin-induced vasodilations were significantly inhibited by l-NAME, GLIB, TEA and IBTX. Relaxin stimulated capillaries to induce a vasodilation in upstream fourth-order arterioles (2.1 ± 0.3 μm), indicating that relaxin can induce conducted responses vasodilation that travels through blood vessel walls via gap junctions. We confirmed gap junction involvement by showing that gap junction uncouplers (18-β-glycyrrhetinic acid (40 × 10(-6) M) or 0.07% halothane) inhibited upstream vasodilations to localised relaxin stimulation of second-order arterioles. Therefore, relaxin produces transient NO- and K(+) channel-dependent vasodilations in skeletal muscle arterioles and stimulates capillaries to initiate conducted responses. The transient nature of the arteriolar dilation brings into question the role of skeletal muscle vascular beds in generating the sustained systemic haemodynamic effects induced by relaxin.
Collapse
Affiliation(s)
- Jordan M Willcox
- Department of Biomedical Sciences Human Health, University of Guelph, ANNU Bldg, Room 350, Guelph, Ontario, Canada N1G 2W1
| | | | | |
Collapse
|
37
|
Twynstra J, Ruiz DA, Murrant CL. Functional coordination of the spread of vasodilations through skeletal muscle microvasculature: implications for blood flow control. Acta Physiol (Oxf) 2012; 206:229-41. [PMID: 22726936 DOI: 10.1111/j.1748-1716.2012.02465.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2012] [Revised: 03/30/2012] [Accepted: 06/18/2012] [Indexed: 11/29/2022]
Abstract
AIM We sought to understand the integrated vascular response to muscle contraction by determining how different branch orders of the terminal microvascular network respond to stimulation using a K(ATP) channel opener pinacidil (PIN) as a muscle contraction mimetic. METHODS Using the blood perfused, hamster cremaster preparation in situ, we locally micropipette-applied 10(-5) M PIN on the capillaries, Branch arteriole (third order, two branch orders up from the capillaries) and transverse arterioles (TA) (second order, three branch orders up from the capillaries) and observed different branch orders of the microvasculature to determine where the localized vasodilation spread throughout the terminal microvascular network. RESULTS We observed that PIN stimulation of capillaries caused associated upstream vasodilation of the module inflow arteriole (MI) (fourth order, the terminal arteriole) (2.1 ± 0.4 μm), the associate Branch (1.4 ± 0.5 μm) and in the upstream direction on the TA (2.1 ± 0.5 μm). Vasodilation did not occur in all MIs (-0.2 ± 0.2 μm) from the vasodilated branch and did not go downstream on the TA (0.7 ± 0.4 μm). Branch stimulation caused upstream TA (3.3 ± 1.0 μm) and upstream Branch (1.7 ± 0.3 μm) vasodilation but not downstream TA (1.5 ± 0.6 μm) or downstream Branch (0.2 ± 0.3 μm) vasodilation. TA stimulation caused conducted responses in both directions and into all associated arteriolar Branches and MIs. CONCLUSIONS The spread of the conducted response is dependent on the vascular branch order stimulated: capillary stimulation was most specific in its direction and TA stimulation was the least specific. Our data indicate that vascular branch order is important in determining the vascular response needed to direct blood flow to contracting skeletal muscle cells.
Collapse
Affiliation(s)
- J. Twynstra
- Department of Human Health and Nutritional Science; University of Guelph; Guelph; ON; Canada
| | - D. A. Ruiz
- Department of Human Health and Nutritional Science; University of Guelph; Guelph; ON; Canada
| | - C. L. Murrant
- Department of Human Health and Nutritional Science; University of Guelph; Guelph; ON; Canada
| |
Collapse
|
38
|
Abstract
The mechanism enabling coordination of the resistance of feed arteries with microcirculatory arterioles to rapidly regulate tissue blood flow in line with changes in metabolic demand has preoccupied scientists for a quarter of a century. As experiments uncovered the underlying electrical events, it was frequently questioned how vasodilation could conduct over long distances without appreciable attenuation. This perspective reviews the data pertinent to this phenomenon and provides evidence that this remarkable response could be made possible by a simple mechanism based on the steep relationship between membrane potential and calcium entry demonstrated by the voltage-dependent calcium channels which mediate the control of vascular tone in vivo.
Collapse
Affiliation(s)
- Caryl E Hill
- Department of Neuroscience, The John Curtin School of Medical Research, The Australian National University, Acton, ACT, Australia.
| |
Collapse
|
39
|
Wang L, Yin J, Nickles HT, Ranke H, Tabuchi A, Hoffmann J, Tabeling C, Barbosa-Sicard E, Chanson M, Kwak BR, Shin HS, Wu S, Isakson BE, Witzenrath M, de Wit C, Fleming I, Kuppe H, Kuebler WM. Hypoxic pulmonary vasoconstriction requires connexin 40-mediated endothelial signal conduction. J Clin Invest 2012; 122:4218-30. [PMID: 23093775 PMCID: PMC3484430 DOI: 10.1172/jci59176] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 08/30/2012] [Indexed: 12/21/2022] Open
Abstract
Hypoxic pulmonary vasoconstriction (HPV) is a physiological mechanism by which pulmonary arteries constrict in hypoxic lung areas in order to redirect blood flow to areas with greater oxygen supply. Both oxygen sensing and the contractile response are thought to be intrinsic to pulmonary arterial smooth muscle cells. Here we speculated that the ideal site for oxygen sensing might instead be at the alveolocapillary level, with subsequent retrograde propagation to upstream arterioles via connexin 40 (Cx40) endothelial gap junctions. HPV was largely attenuated by Cx40-specific and nonspecific gap junction uncouplers in the lungs of wild-type mice and in lungs from mice lacking Cx40 (Cx40-/-). In vivo, hypoxemia was more severe in Cx40-/- mice than in wild-type mice. Real-time fluorescence imaging revealed that hypoxia caused endothelial membrane depolarization in alveolar capillaries that propagated to upstream arterioles in wild-type, but not Cx40-/-, mice. Transformation of endothelial depolarization into vasoconstriction involved endothelial voltage-dependent α1G subtype Ca2+ channels, cytosolic phospholipase A2, and epoxyeicosatrienoic acids. Based on these data, we propose that HPV originates at the alveolocapillary level, from which the hypoxic signal is propagated as endothelial membrane depolarization to upstream arterioles in a Cx40-dependent manner.
Collapse
MESH Headings
- Animals
- Calcium Channels/metabolism
- Connexins/genetics
- Connexins/metabolism
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Endothelium, Vascular/physiopathology
- Human Umbilical Vein Endothelial Cells
- Humans
- Hypoxia/genetics
- Hypoxia/metabolism
- Hypoxia/pathology
- Hypoxia/physiopathology
- Lung/blood supply
- Lung/metabolism
- Lung/pathology
- Lung/physiopathology
- Mice
- Mice, Knockout
- Muscle, Smooth/metabolism
- Muscle, Smooth/pathology
- Muscle, Smooth/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phospholipases A2, Cytosolic/metabolism
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Signal Transduction
- Vasoconstriction
- Gap Junction alpha-5 Protein
Collapse
Affiliation(s)
- Liming Wang
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Jun Yin
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Hannah T. Nickles
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Hannes Ranke
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Arata Tabuchi
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Julia Hoffmann
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Christoph Tabeling
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Eduardo Barbosa-Sicard
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Marc Chanson
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Brenda R. Kwak
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Hee-Sup Shin
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Songwei Wu
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Brant E. Isakson
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Martin Witzenrath
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Cor de Wit
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Ingrid Fleming
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Hermann Kuppe
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Wolfgang M. Kuebler
- The Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada.
Institute of Physiology, Department of Internal Medicine, Charité-Universitätsmedizin, Berlin, Germany.
Department of Cardiothoracic Surgery, Affiliated People′s Hospital of Jiangsu University, Zhenjiang, China.
German Heart Institute, Berlin, Germany.
Division of Infectious Diseases and Pulmonary Medicine, Department of Internal Medicine, Charité-Universitätsmedizin Berlin, Germany.
Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
Laboratory of Clinical Investigation III, Hôpitaux Universitaires de Genève (HUG), and
Department of Pathology and Immunology, Université de Genève, Genève, Switzerland.
Center for Neural Science, Korea Institute of Science and Technology, Seoul, Republic of Korea.
Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.
Robert M. Berne Cardiovascular Research Center, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA.
Institute of Physiology, University of Lübeck, Lübeck, Germany.
Department of Surgery and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
40
|
Behringer EJ, Socha MJ, Polo-Parada L, Segal SS. Electrical conduction along endothelial cell tubes from mouse feed arteries: confounding actions of glycyrrhetinic acid derivatives. Br J Pharmacol 2012; 166:774-87. [PMID: 22168386 DOI: 10.1111/j.1476-5381.2011.01814.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Electrical conduction along endothelium of resistance vessels has not been determined independently of the influence of smooth muscle, surrounding tissue or blood. Two interrelated hypotheses were tested: (i) Intercellular conduction of electrical signals is manifest in endothelial cell (EC) tubes; and (ii) Inhibitors of gap junction channels (GJCs) have confounding actions on EC electrical and Ca(2+) signalling. EXPERIMENTAL APPROACH Intact EC tubes were isolated from abdominal muscle feed (superior epigastric) arteries of C57BL/6 mice. Hyperpolarization was initiated with indirect (ACh) and direct (NS309) stimulation of intermediate- and small-conductance Ca(2+) -activated K(+) channels (IK(Ca) /SK(Ca) ). Remote membrane potential (V(m) ) responses to intracellular current injection defined the length constant (λ) for electrical conduction. Dye coupling was evaluated following intracellular microinjection of propidium iodide. Intracellular Ca(2+) dynamics were determined using Fura-2 photometry. Carbenoxolone (CBX) or β-glycyrrhetinic acid (βGA) was used to investigate the role of GJCs. KEY RESULTS Steady-state V(m) of ECs was -25 mV. ACh and NS309 hyperpolarized ECs by -40 and -60 mV respectively. Electrical conduction decayed monoexponentially with distance (λ∼1.4 mm). Propidium iodide injected into one EC spread into surrounding ECs. CBX or βGA inhibited dye transfer, electrical conduction and EC hyperpolarization reversibly. Both agents elevated resting Ca(2+) while βGA inhibited responses to ACh. CONCLUSIONS AND IMPLICATIONS Individual cells were effectively coupled to each other within EC tubes. Inhibiting GJCs with glycyrrhetinic acid derivatives blocked hyperpolarization mediated by IK(Ca) /SK(Ca) channels, regardless of Ca(2+) signalling, obviating use of these agents in distinguishing key determinants of electrical conduction along the endothelium.
Collapse
Affiliation(s)
- Erik J Behringer
- Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA
| | | | | | | |
Collapse
|
41
|
Behringer EJ, Segal SS. Spreading the signal for vasodilatation: implications for skeletal muscle blood flow control and the effects of ageing. J Physiol 2012; 590:6277-84. [PMID: 22890708 DOI: 10.1113/jphysiol.2012.239673] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Blood flow control requires coordinated contraction and relaxation of smooth muscle cells (SMCs) along and among the arterioles and feed arteries that comprise vascular resistance networks. Whereas smooth muscle contraction of resistance vessels is enhanced by noradrenaline release along perivascular sympathetic nerves, the endothelium is integral to coordinating smooth muscle relaxation. Beyond producing nitric oxide in response to agonists and shear stress, endothelial cells (ECs) provide an effective conduit for conducting hyperpolarization along vessel branches and into surrounding SMCs through myoendothelial coupling. In turn, bidirectional signalling from SMCs into ECs enables the endothelium to moderate adrenergic vasoconstriction in response to sympathetic nerve activity. This review focuses on the endothelium as the cellular pathway that coordinates spreading vasodilatation. We discuss the nature and regulation of cell-to-cell coupling through gap junctions, bidirectional signalling between ECs and SMCs, and how oxidative stress during ageing may influence respective signalling pathways. Our recent findings illustrate the role of small (SK(Ca)) and intermediate (IK(Ca)) Ca(2+) activated K(+) channels as modulators of electrical conduction along the endothelium. Gaps in current understanding indicate the need to determine mechanisms that regulate intracellular Ca(2+) homeostasis and ion channel activation in the resistance vasculature with advancing age.
Collapse
Affiliation(s)
- Erik J Behringer
- Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA
| | | |
Collapse
|
42
|
Behringer EJ, Segal SS. Tuning electrical conduction along endothelial tubes of resistance arteries through Ca(2+)-activated K(+) channels. Circ Res 2012; 110:1311-21. [PMID: 22492531 DOI: 10.1161/circresaha.111.262592] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
RATIONALE Electrical conduction through gap junction channels between endothelial cells of resistance vessels is integral to blood flow control. Small and intermediate-conductance Ca(2+)-activated K(+) channels (SK(Ca)/IK(Ca)) initiate electrical signals in endothelial cells, but it is unknown whether SK(Ca)/IK(Ca) activation alters signal transmission along the endothelium. OBJECTIVE We tested the hypothesis that SK(Ca)/IK(Ca) activity regulates electrical conduction along the endothelium of resistance vessels. METHODS AND RESULTS Freshly isolated endothelial cell tubes (60 μm wide; 1-3 mm long; cell length, ≈35 μm) from mouse skeletal muscle feed (superior epigastric) arteries were studied using dual intracellular microelectrodes. Current was injected (±0.1-3 nA) at site 1 while recording membrane potential (V(m)) at site 2 (separation distance=50-2000 μm). SK(Ca)/IK(Ca) activation (NS309, 1 μmol/L) reduced the change in V(m) along endothelial cell tubes by ≥50% and shortened the electrical length constant (λ) from 1380 to 850 μm (P<0.05) while intercellular dye transfer (propidium iodide) was maintained. Activating SK(Ca)/IK(Ca) with acetylcholine or SKA-31 also reduced electrical conduction. These effects of SK(Ca)/IK(Ca) activation persisted when hyperpolarization (>30 mV) was prevented with 60 mmol/L [K(+)](o). Conversely, blocking SK(Ca)/IK(Ca) (apamin+charybdotoxin) depolarized cells by ≈10 mV and enhanced electrical conduction (ie, changes in V(m)) by ≈30% (P<0.05). CONCLUSIONS These findings illustrate a novel role for SK(Ca)/IK(Ca) activity in tuning electrical conduction along the endothelium of resistance vessels by governing signal dissipation through changes in membrane resistance. Voltage-insensitive ion channels can thereby tune intercellular electrical signaling independent from gap junction channels.
Collapse
Affiliation(s)
- Erik J Behringer
- Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA
| | | |
Collapse
|
43
|
Hald BO, Jensen LJ, Sørensen PG, Holstein-Rathlou NH, Jacobsen JCB. Applicability of cable theory to vascular conducted responses. Biophys J 2012; 102:1352-62. [PMID: 22455918 DOI: 10.1016/j.bpj.2012.01.055] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 01/13/2012] [Accepted: 01/27/2012] [Indexed: 12/19/2022] Open
Abstract
Conduction processes in the vasculature have traditionally been described using cable theory, i.e., locally induced signals decaying passively along the arteriolar wall. The decay is typically quantified using the steady-state length-constant, λ, derived from cable theory. However, the applicability of cable theory to blood vessels depends on assumptions that are not necessarily fulfilled in small arteries and arterioles. We have employed a morphologically and electrophysiologically detailed mathematical model of a rat mesenteric arteriole to investigate if the assumptions hold and whether λ adequately describes simulated conduction profiles. We find that several important cable theory assumptions are violated when applied to small blood vessels. However, the phenomenological use of a length-constant from a single exponential function is a good measure of conduction length. Hence, λ should be interpreted as a descriptive measure and not in light of cable theory. Determination of λ using cable theory assumes steady-state conditions. In contrast, using the model it is possible to probe how conduction behaves before steady state is achieved. As ion channels have time-dependent activation and inactivation, the conduction profile changes considerably during this dynamic period with an initially longer spread of current. This may have implications in relation to explaining why different agonists have different conduction properties. Also, it illustrates the necessity of using and developing models that handle the nonlinearity of ion channels.
Collapse
Affiliation(s)
- Bjørn Olav Hald
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | | | |
Collapse
|
44
|
Bohlen HG. Rapid and slow nitric oxide responses during conducted vasodilation in the in vivo intestine and brain cortex microvasculatures. Microcirculation 2012; 18:623-34. [PMID: 22098301 DOI: 10.1111/j.1549-8719.2011.00127.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Conduction of arteriolar vasodilation is initiated by activation of nitric oxide (NO) mechanisms, but dependent on conduction of hyperpolarization. Most studies have used brief (<1 second) activation of the initial vasodilation to evaluate the fast conduction processes. However, most arteriolar mechanisms involving NO production persist for minutes. In this study, fast and slower components of arteriolar conduction in the in vivo rat brain and small intestine were compared using three-minute stimulation of NO-dependent vasodilation and measurement of [NO] at the distal sites. Within 10-15 seconds, both vasculatures had a rapidly conducted vasodilation and dilation at distance had a fast but small [NO] component. A slower but larger distal vasodilation occurred after 60-90 seconds in the intestine, but not the brain, and was associated with a substantial increase in [NO]. This slowly developed dilation appeared to be caused by flow mediated responses of larger arterioles as smaller arterioles dilated to lower downstream resistance. These results indicate while the intestinal and cerebral arterioles have a fast conducted vasodilation system, the intestinal arterioles also have a slower but larger dilation of major arterioles that is NO related and dependent on the conduction of vasodilation between small arterioles.
Collapse
Affiliation(s)
- H Glenn Bohlen
- Department of Cellular and Integrative Physiology, Indiana University Medical School, Indianapolis, Indiana 46202, USA.
| |
Collapse
|
45
|
Socha MJ, Behringer EJ, Segal SS. Calcium and electrical signalling along endothelium of the resistance vasculature. Basic Clin Pharmacol Toxicol 2011; 110:80-6. [PMID: 21917120 DOI: 10.1111/j.1742-7843.2011.00798.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This MiniReview is focused on the nature of intercellular signalling along the endothelium that helps to co-ordinate blood flow control in vascular resistance networks. Vasodilation initiated by contracting skeletal muscle ascends from arterioles within the tissue to encompass resistance arteries upstream and thereby increase blood flow during exercise. In resistance vessels, acetylcholine microiontophoresis or intracellular current injection initiates hyperpolarization that conducts through gap junction channels (GJCs) along the vessel wall resulting in conducted vasodilation (CVD). Both ascending vasodilation and CVD are eliminated with endothelial cell (EC) disruption, pointing to common signalling events and mutual dependence upon EC integrity. As demonstrated by electrical coupling and dye transfer during intracellular recording, their longitudinal orientation and robust expression of GJCs enable ECs to play a predominant role in CVD. Once conduction is initiated, a major interest centres on whether CVD is purely passive or involves additional 'active' signalling events. Here, we discuss components for Ca²⁺ and electrical signalling with an emphasis on intercellular coupling through endothelial GJCs. We stress the importance of understanding relationships between intracellular Ca²⁺ dynamics, EC hyperpolarization and CVD while integrating findings from isolated ECs into more complex interactions in vivo. Whereas endothelial dysfunction accompanies cardiovascular disease and the components of intra- and inter-cellular signalling are increasingly well defined, little is known of how Ca²⁺ signalling and electrical conduction along microvascular endothelium are altered in diseased states. Thus, greater insight into how these relationships are governed and interact is a key goal for continued research efforts.
Collapse
Affiliation(s)
- Matthew J Socha
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA
| | | | | |
Collapse
|
46
|
Fang JS, Angelov SN, Simon AM, Burt JM. Cx40 is required for, and cx37 limits, postischemic hindlimb perfusion, survival and recovery. J Vasc Res 2011; 49:2-12. [PMID: 21986401 PMCID: PMC3221267 DOI: 10.1159/000329616] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 05/20/2011] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND/AIMS Ischemia induced by large-vessel obstruction or vascular injury induces a complex cascade of vasodilatory, remodeling and inflammatory pathways; coordination of these processes by vascular endothelium is likely to involve endothelial gap junctions. Vascular endothelium predominantly expresses two connexin (Cx) isoforms: Cx37 and Cx40. The relevance of these Cxs to postischemic limb recovery remains unclear. METHODS In this study, we use a well-established, severe femoral-saphenous artery-vein pair resection model of unilateral hindlimb ischemia to test the relevance of Cx37 and Cx40 to postischemic tissue survival and recovery of limb perfusion. RESULTS Cx40-deficient animals (Cx40-/-) experienced a severe reduction in limb perfusion relative to wild-type (WT) animals and exhibited profound and rapid failure of ischemic limb survival. By contrast, the deficit in limb perfusion was less severe in Cx37-ablated (Cx37-/-) animals compared to WT, corresponding with more rapid recovery of limb appearance and use. These results demonstrate that Cx40 is necessary for postischemic limb survival and reperfusion, whereas Cx37 deletion reduces the extent of ischemia in the same model. CONCLUSION In summary, we present evidence demonstrating that Cx37 and Cx40 uniquely regulate postischemic limb perfusion, altering the severity of ischemic insult and consequent postischemic survival.
Collapse
Affiliation(s)
| | | | | | - Janis M. Burt
- *Dr. Janis M. Burt, Department of Physiology, University of Arizona Health Sciences Center, 1656 E Mabel Drive, Room 440, PO Box 245051, Tucson, AZ 85724-5051 (USA), Tel. +1 520 626 6833, E-Mail
| |
Collapse
|
47
|
Bagher P, Davis MJ, Segal SS. Intravital macrozoom imaging and automated analysis of endothelial cell calcium signals coincident with arteriolar dilation in Cx40(BAC) -GCaMP2 transgenic mice. Microcirculation 2011; 18:331-8. [PMID: 21418383 DOI: 10.1111/j.1549-8719.2011.00093.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Calcium signaling is integral to endothelium-dependent vasodilation. Our goal was to develop methods enabling automated analyses for accurately and objectively determining the dynamic relationship between EC Ca(2+) responses and arteriolar diameter in vivo. METHODS User-friendly software (DiaFluor) written in LabView was applied to images acquired at 15fps with a custom macrozoom intravital microscope to evaluate changes in EC Ca(2+) concomitant with arteriolar diameter. Transgenic Cx40(BAC) -GCaMP2 mice expressing a fluorescent Ca(2+) indicator molecule in arteriolar ECs enabled resolution of EC Ca(2+) signaling in response to ACh microiontophoresis (500nA, 100-1000msec pulse) from a micropipette (1μm tip) positioned adjacent to an arteriole in the superfused cremaster muscle preparation. RESULTS A 100-msec pulse of ACh (1M) had little effect on EC Ca(2+) or arteriolar diameter. As pulse duration increased, vasodilation increased with fluorescence intensity (p<0.01). Based upon fluorescence responses (F/F(o)), the effective diffusion distance of ACh along arterioles increased from ∼100μm (250msec pulse) to ∼200μm (1000msec pulse) with a peak velocity of ∼150μm/sec. CONCLUSIONS The novel imaging and software presented here are the first to enable automated simultaneous evaluation of EC Ca(2+) signaling and endothelium-dependent vasodilation in vivo.
Collapse
Affiliation(s)
- Pooneh Bagher
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri 65212, USA
| | | | | |
Collapse
|
48
|
Bagher P, Davis MJ, Segal SS. Visualizing calcium responses to acetylcholine convection along endothelium of arteriolar networks in Cx40BAC-GCaMP2 transgenic mice. Am J Physiol Heart Circ Physiol 2011; 301:H794-802. [PMID: 21666122 PMCID: PMC3191093 DOI: 10.1152/ajpheart.00425.2011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 06/02/2011] [Indexed: 11/22/2022]
Abstract
Acetylcholine evokes endothelium-dependent vasodilation subsequent to a rise in intracellular calcium. Despite widespread application in human and animal studies, calcium responses to intravascular ACh have not been visualized in vivo. Microiontophoresis of ACh in tissue adjacent to an arteriole activates abluminal muscarinic receptors on endothelial cells within a "local" region of diffusion, but it is unknown whether ACh released in such fashion gains access to the flow stream resulting in further actions downstream. To test this hypothesis and provide new insight into calcium signaling in vivo, we studied the cremaster muscle microcirculation of anesthetized male Cx40(BAC)-GCaMP2 transgenic mice (n = 22; 5-9 mo; 33 ± 1 g) expressing the fluorescent calcium sensor GCaMP2 selectively in arteriolar endothelial cells. Submaximal ACh stimuli were delivered using microiontophoresis (1-μm pipette tip, 500 nA). With stimulus duration <500 ms or with the micropipette positioned within one vessel diameter (∼30 μm) away from an arteriole, endothelial cell calcium fluorescence was restricted to the region of ACh diffusion (<200 μm). In contrast, with the micropipette tip positioned immediately adjacent to an arteriole or within its lumen, calcium fluorescence encompassed entire networks downstream. The velocity of downstream calcium signaling in response to ACh increased with centerline velocity of fluorescent tracer microbeads (r(2) > 0.99; range: <1 mm/s to >10 mm/s). Diverting arteriolar blood flow into a side branch redirected downstream fluorescence responses to ACh; occluding flow abolished responses. Blocking luminal muscarinic receptors (intravascular glycopyrrolate; 6 μg/kg) inhibited downstream responses reversibly. Through visualizing the actions of a "local" ACh stimulus on endothelial cell calcium fluorescence in vivo, the present findings illustrate that transmural diffusion and convection of an agonist can activate entire networks of arteriolar endothelial cells concomitant with its delivery in the flow stream.
Collapse
Affiliation(s)
- Pooneh Bagher
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri 65212, USA
| | | | | |
Collapse
|
49
|
Abstract
This review is concerned with understanding how vasodilation initiated from local sites in the tissue can spread to encompass multiple branches of the resistance vasculature. Within tissues, arteriolar networks control the distribution and magnitude of capillary perfusion. Vasodilation arising from the microcirculation can 'ascend' into feed arteries that control blood flow into arteriolar networks. Thus distal segments of the resistance network signal proximal segments to dilate and thereby increase total oxygen supply to parenchymal cells. August Krogh proposed that innervation of capillaries provided the mechanism for a spreading vasodilatory response. With greater understanding of the ultrastructural organization of resistance networks, an alternative explanation has emerged: Electrical signalling from cell to cell along the vessel wall through gap junctions. Hyperpolarization originates from ion channel activation at the site of stimulation with the endothelium serving as the predominant cellular pathway for signal conduction along the vessel wall. As hyperpolarization travels, it is transmitted into surrounding smooth muscle cells through myoendothelial coupling to promote relaxation. Conducted vasodilation (CVD) encompasses greater distances than can be explained by passive decay and understanding such behaviour is the focus of current research efforts. In the context of athletic performance, the ability of vasodilation to ascend into feed arteries is essential to achieving peak levels of muscle blood flow. CVD is tempered by sympathetic neuroeffector signalling when governing muscle blood flow at rest and during exercise. Impairment of conduction during ageing and in diseased states can limit physical work capacity by restricting muscle blood flow.
Collapse
Affiliation(s)
- P Bagher
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA
| | | |
Collapse
|
50
|
Félétou M. The Endothelium, Part I: Multiple Functions of the Endothelial Cells -- Focus on Endothelium-Derived Vasoactive Mediators. ACTA ACUST UNITED AC 2011. [DOI: 10.4199/c00031ed1v01y201105isp019] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|