1
|
Negri S, Sanford M, Shi H, Tarantini S. The role of endothelial TRP channels in age-related vascular cognitive impairment and dementia. Front Aging Neurosci 2023; 15:1149820. [PMID: 37020858 PMCID: PMC10067599 DOI: 10.3389/fnagi.2023.1149820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/28/2023] [Indexed: 04/07/2023] Open
Abstract
Transient receptor potential (TRP) proteins are part of a superfamily of polymodal cation channels that can be activated by mechanical, physical, and chemical stimuli. In the vascular endothelium, TRP channels regulate two fundamental parameters: the membrane potential and the intracellular Ca2+ concentration [(Ca2+)i]. TRP channels are widely expressed in the cerebrovascular endothelium, and are emerging as important mediators of several brain microvascular functions (e.g., neurovascular coupling, endothelial function, and blood-brain barrier permeability), which become impaired with aging. Aging is the most significant risk factor for vascular cognitive impairment (VCI), and the number of individuals affected by VCI is expected to exponentially increase in the coming decades. Yet, there are currently no preventative or therapeutic treatments available against the development and progression of VCI. In this review, we discuss the involvement of endothelial TRP channels in diverse physiological processes in the brain as well as in the pathogenesis of age-related VCI to explore future potential neuroprotective strategies.
Collapse
Affiliation(s)
- Sharon Negri
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Madison Sanford
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Helen Shi
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- *Correspondence: Stefano Tarantini,
| |
Collapse
|
2
|
Nam YW, Pala R, El-Sayed NS, Larin-Henriquez D, Amirrad F, Yang G, Rahman MA, Orfali R, Downey M, Parang K, Nauli SM, Zhang M. Subtype-Selective Positive Modulation of K Ca2.3 Channels Increases Cilia Length. ACS Chem Biol 2022; 17:2344-2354. [PMID: 35947779 PMCID: PMC9396613 DOI: 10.1021/acschembio.2c00469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Small-conductance Ca2+-activated potassium
(KCa2.x) channels are gated exclusively by intracellular
Ca2+. The activation of KCa2.3 channels induces
hyperpolarization,
which augments Ca2+ signaling in endothelial cells. Cilia
are specialized Ca2+ signaling compartments. Here, we identified
compound 4 that potentiates human KCa2.3 channels
selectively. The subtype selectivity of compound 4 for
human KCa2.3 over rat KCa2.2a channels relies
on an isoleucine residue in the HA/HB helices. Positive modulation
of KCa2.3 channels by compound 4 increased
flow-induced Ca2+ signaling and cilia length, while negative
modulation by AP14145 reduced flow-induced Ca2+ signaling
and cilia length. These findings were corroborated by the increased
cilia length due to the expression of Ca2+-hypersensitive
KCa2.3_G351D mutant channels and the reduced cilia length
resulting from the expression of Ca2+-hyposensitive KCa2.3_I438N channels. Collectively, we were able to associate
functions of KCa2.3 channels and cilia, two crucial components
in the flow-induced Ca2+ signaling of endothelial cells,
with potential implications in vasodilation and ciliopathic hypertension.
Collapse
Affiliation(s)
- Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Rajasekharreddy Pala
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Naglaa Salem El-Sayed
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Denisse Larin-Henriquez
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Farideh Amirrad
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Grace Yang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Mohammad Asikur Rahman
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Razan Orfali
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Myles Downey
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Keykavous Parang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Surya M Nauli
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, USA
| |
Collapse
|
3
|
Kolski-Andreaco A, Balut CM, Bertuccio CA, Wilson AS, Rivers WM, Liu X, Gandley RE, Straub AC, Butterworth MB, Binion D, Devor DC. Histone deacetylase inhibitors (HDACi) increase expression of KCa2.3 (SK3) in primary microvascular endothelial cells. Am J Physiol Cell Physiol 2022; 322:C338-C353. [PMID: 35044858 PMCID: PMC8858676 DOI: 10.1152/ajpcell.00409.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The small conductance calcium-activated potassium channel (KCa2.3) has long been recognized for its role in mediating vasorelaxation through the endothelium-derived hyperpolarization (EDH) response. Histone deacetylases (HDACs) have been implicated as potential modulators of blood pressure and histone deacetylase inhibitors (HDACi) are being explored as therapeutics for hypertension. Herein, we show that HDACi increase KCa2.3 expression when heterologously expressed in HEK cells and endogenously expressed in primary cultures of human umbilical vein endothelial cells (HUVECs) and human intestinal microvascular endothelial cells (HIMECs). When primary endothelial cells were exposed to HDACi, KCa2.3 transcripts, subunits, and functional current are increased. Quantitative RT-PCR (qPCR) demonstrated increased KCa2.3 mRNA following HDACi, confirming transcriptional regulation of KCa2.3 by HDACs. By using pharmacological agents selective for different classes of HDACs, we discriminated between cytoplasmic and epigenetic modulation of KCa2.3. Biochemical analysis revealed an association between the cytoplasmic HDAC6 and KCa2.3 in immunoprecipitation studies. Specifically inhibiting HDAC6 increases expression of KCa2.3. In addition to increasing the expression of KCa2.3, we show that nonspecific inhibition of HDACs causes an increase in the expression of the molecular chaperone Hsp70 in endothelial cells. When Hsp70 is inhibited in the presence of HDACi, the magnitude of the increase in KCa2.3 expression is diminished. Finally, we show a slower rate of endocytosis of KCa2.3 as a result of exposure of primary endothelial cells to HDACi. These data provide the first demonstrated approach to increase KCa2.3 channel number in endothelial cells and may partially account for the mechanism by which HDACi induce vasorelaxation.
Collapse
Affiliation(s)
| | - Corina M. Balut
- 1Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Annette S. Wilson
- 2Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - William M. Rivers
- 2Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xiaoning Liu
- 1Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robin E. Gandley
- 3Department of Obstetrics and Gynecology and Reproductive Sciences, Magee Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adam C. Straub
- 4Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - David Binion
- 2Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Daniel C. Devor
- 1Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
4
|
Jackson WF. Calcium-Dependent Ion Channels and the Regulation of Arteriolar Myogenic Tone. Front Physiol 2021; 12:770450. [PMID: 34819877 PMCID: PMC8607693 DOI: 10.3389/fphys.2021.770450] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/11/2021] [Indexed: 11/25/2022] Open
Abstract
Arterioles in the peripheral microcirculation regulate blood flow to and within tissues and organs, control capillary blood pressure and microvascular fluid exchange, govern peripheral vascular resistance, and contribute to the regulation of blood pressure. These important microvessels display pressure-dependent myogenic tone, the steady state level of contractile activity of vascular smooth muscle cells (VSMCs) that sets resting arteriolar internal diameter such that arterioles can both dilate and constrict to meet the blood flow and pressure needs of the tissues and organs that they perfuse. This perspective will focus on the Ca2+-dependent ion channels in the plasma and endoplasmic reticulum membranes of arteriolar VSMCs and endothelial cells (ECs) that regulate arteriolar tone. In VSMCs, Ca2+-dependent negative feedback regulation of myogenic tone is mediated by Ca2+-activated K+ (BKCa) channels and also Ca2+-dependent inactivation of voltage-gated Ca2+ channels (VGCC). Transient receptor potential subfamily M, member 4 channels (TRPM4); Ca2+-activated Cl− channels (CaCCs; TMEM16A/ANO1), Ca2+-dependent inhibition of voltage-gated K+ (KV) and ATP-sensitive K+ (KATP) channels; and Ca2+-induced-Ca2+ release through inositol 1,4,5-trisphosphate receptors (IP3Rs) participate in Ca2+-dependent positive-feedback regulation of myogenic tone. Calcium release from VSMC ryanodine receptors (RyRs) provide negative-feedback through Ca2+-spark-mediated control of BKCa channel activity, or positive-feedback regulation in cooperation with IP3Rs or CaCCs. In some arterioles, VSMC RyRs are silent. In ECs, transient receptor potential vanilloid subfamily, member 4 (TRPV4) channels produce Ca2+ sparklets that activate IP3Rs and intermediate and small conductance Ca2+ activated K+ (IKCa and sKCa) channels causing membrane hyperpolarization that is conducted to overlying VSMCs producing endothelium-dependent hyperpolarization and vasodilation. Endothelial IP3Rs produce Ca2+ pulsars, Ca2+ wavelets, Ca2+ waves and increased global Ca2+ levels activating EC sKCa and IKCa channels and causing Ca2+-dependent production of endothelial vasodilator autacoids such as NO, prostaglandin I2 and epoxides of arachidonic acid that mediate negative-feedback regulation of myogenic tone. Thus, Ca2+-dependent ion channels importantly contribute to many aspects of the regulation of myogenic tone in arterioles in the microcirculation.
Collapse
Affiliation(s)
- William F Jackson
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
5
|
Hakim MA, Behringer EJ. Methyl-Beta-Cyclodextrin Restores K IR Channel Function in Brain Endothelium of Female Alzheimer's Disease Mice. J Alzheimers Dis Rep 2021; 5:693-703. [PMID: 34755043 PMCID: PMC8543374 DOI: 10.3233/adr-210016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
Background: As the sixth-leading cause of death in the United States, Alzheimer’s disease (AD) entails deteriorating endothelial control of blood flow throughout the brain. In particular, reduced inward-rectifying K+ (KIR) channel function in animal models of aging and AD compromises endothelial function and optimal perfusion of brain parenchyma. Deficient endothelial KIR channels may result from aberrant interaction with plasma membrane cholesterol as a primary regulator of membrane fluidity and ion channels. Objective: We tested the hypothesis that mild methyl-β-cyclodextrin (MβCD) treatment to reduce membrane cholesterol may restore endothelial KIR channel function in brain endothelium of old AD mice. Methods: Membrane potential was continuously measured in isolated endothelial tubes from posterior cerebral arteries of young (1 to 3 months) and old (16 to 19 months) female 3xTg-AD mice before and after mild treatment with the cholesterol-removing agent MβCD (1 mmol/L). Elevated extracellular potassium ([K+]E; 15 mmol/L) and NS309 (1μmol/L) activated KIR and Ca2+-activated K+ (SKCa/IKCa) channels respectively before and after MβCD treatment. Results: SKCa/IKCa channel function for producing hyperpolarization remained stable regardless of age group and MβCD treatment (ΔVm: ∼–33 mV). However, as deficient during AD, KIR channel function was restored (ΔVm: –9±1 mV) versus pre-MβCD conditions (–5±1 mV); a progressive effect that reached –14±1 mV hyperpolarization at 60 min following MβCD washout. Conclusion: In female animals, MβCD treatment of brain endothelium selectively restores KIR versus SKCa/IKCa channel function during AD. Thus, the endothelial cholesterol-KIR channel interface is a novel target for ameliorating perfusion of the AD brain.
Collapse
Affiliation(s)
- Md A Hakim
- Basic Sciences, Loma Linda University, Loma Linda, CA, USA
| | | |
Collapse
|
6
|
Hakim MA, Behringer EJ. Development of Alzheimer's Disease Progressively Alters Sex-Dependent KCa and Sex-Independent KIR Channel Function in Cerebrovascular Endothelium. J Alzheimers Dis 2021; 76:1423-1442. [PMID: 32651315 DOI: 10.3233/jad-200085] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Development of Alzheimer's disease (AD) pathology is associated with impaired blood flow delivery of oxygen and nutrients throughout the brain. Cerebrovascular endothelium regulates vasoreactivity of blood vessel networks for optimal cerebral blood flow. OBJECTIVE We tested the hypothesis that cerebrovascular endothelial Gq-protein-coupled receptor (GPCR; purinergic and muscarinic) and K+ channel [Ca2+-activated (KCa2.3/SK3 and KCa3.1/IK1) and inward-rectifying (KIR2.x)] function declines during progressive AD pathology. METHODS We applied simultaneous measurements of intracellular Ca2+ ([Ca2+]i) and membrane potential (Vm) in freshly isolated endothelium from posterior cerebral arteries of 3×Tg-AD mice [young, no pathology (1- 2 mo), cognitive impairment (CI; 4- 5 mo), extracellular Aβ plaques (Aβ; 6- 8 mo), and Aβ plaques + neurofibrillary tangles (AβT; 12- 15 mo)]. RESULTS The coupling of ΔVm-to-Δ[Ca2+]i during AβT pathology was lowest for both sexes but, overall, ATP-induced purinergic receptor function was stable throughout AD pathology. SKCa/IKCa channel function itself was enhanced by ∼20% during AD (Aβ+ AβT) versus pre-AD (Young + CI) in males while steady in females. Accordingly, hyperpolarization-induced [Ca2+]i increases following SKCa/IKCa channel activation and Δ[Ca2+]i-to-ΔVm coupling was enhanced by ≥two-fold during AD pathology in males but not females. Further, KIR channel function decreased by ∼50% during AD conditions versus young regardless of sex. Finally, other than a ∼40% increase in females versus males during Aβ pathology, [Ca2+]i responses to the mitochondrial uncoupler FCCP were similar among AD versus pre-AD conditions. CONCLUSION Altogether, AD pathology represents a condition of altered KCa and KIR channel function in cerebrovascular endothelium in a sex-dependent and sex-independent manner respectively.
Collapse
Affiliation(s)
- Md A Hakim
- Basic Sciences, Loma Linda University, Loma Linda, CA, USA
| | | |
Collapse
|
7
|
Hansen FB, Secher N, Mattson T, Løfgren B, Simonsen U, Granfeldt A. Effect of the KCa3.1 blocker, senicapoc, on cerebral edema and cardiovascular function after cardiac arrest - A randomized experimental rat study. Resusc Plus 2021; 6:100111. [PMID: 34223371 PMCID: PMC8244250 DOI: 10.1016/j.resplu.2021.100111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 11/30/2022] Open
Abstract
Senicapoc was successfully administered intravenously. Senicapoc did not reduce cerebral edema 4 h after cardiac arrest. Senicapoc did not increase mean arterial pressure within 4 h from resuscitation.
Aim Formation of cerebral edema and cardiovascular dysfunction may worsen brain injury following cardiac arrest. We hypothesized that administration of the intermediate calcium-activated potassium (KCa3.1) channel blocker, senicapoc, would reduce cerebral edema and augment mean arterial pressure in the early post-resuscitation period. Method Male Sprague-Dawley rats, aged 11–15 weeks, were utilized in the study. Rats were exposed to 8 min of asphyxial cardiac arrest. Shortly after resuscitation, rats were randomized to receive either vehicle or senicapoc (10 mg/kg) intravenously. The primary outcome was cerebral wet to dry weight ratio 4 h after resuscitation. Secondary outcomes included mean arterial pressure, cardiac output, norepinephrine dose, inflammatory cytokines and neuron specific enolase levels. Additionally, a sub-study was conducted to validate intravenous administration of senicapoc. Results The sub-study revealed that senicapoc-treated rats maintained a significantly higher mean arterial pressure during administration of SKA-31 (a KCa3.1 channel opener). The plasma concentration of senicapoc was 1060 ± 303 ng/ml 4 h after administration. Senicapoc did not reduce cerebral edema or augment mean arterial pressure 4 h after resuscitation. Likewise, cardiac function and norepinephrine dose did not vary between groups. Inflammatory cytokines and neuron specific enolase levels increased in both groups after resuscitation with no difference between groups. Senicapoc enhanced the PaO2/FiO2 ratio significantly 4 h after resuscitation. Conclusion Senicapoc was successfully administered intravenously after resuscitation, but did not reduce cerebral edema or increase mean arterial pressure in the early post-resuscitation period.
Collapse
Affiliation(s)
- Frederik Boe Hansen
- Department of Biomedicine, Aarhus University, Ole Worms Allé 4, 8000 Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Blvd. 82, 8200 Aarhus N, Denmark
| | - Niels Secher
- Department of Anesthesiology and Intensive Care, Aarhus University Hospital, Palle Juul-Jensens Blvd. 99, 8200 Aarhus N, Denmark
| | - Thomas Mattson
- Department of Anesthesiology and Intensive Care, Aarhus University Hospital, Palle Juul-Jensens Blvd. 99, 8200 Aarhus N, Denmark
| | - Bo Løfgren
- Department of Internal Medicine, Randers Regional Hospital, Skovlyvej 15, 8930 Randers NE, Denmark.,Research Center for Emergency Medicine, Aarhus University Hospital, Palle Juul-Jensens Blvd. 161, 8200 Aarhus N, Denmark
| | - Ulf Simonsen
- Department of Biomedicine, Aarhus University, Ole Worms Allé 4, 8000 Aarhus, Denmark
| | - Asger Granfeldt
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Blvd. 82, 8200 Aarhus N, Denmark.,Department of Anesthesiology and Intensive Care, Aarhus University Hospital, Palle Juul-Jensens Blvd. 99, 8200 Aarhus N, Denmark
| |
Collapse
|
8
|
Nam YW, Kong D, Wang D, Orfali R, Sherpa RT, Totonchy J, Nauli SM, Zhang M. Differential modulation of SK channel subtypes by phosphorylation. Cell Calcium 2021; 94:102346. [PMID: 33422768 PMCID: PMC8415101 DOI: 10.1016/j.ceca.2020.102346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 01/01/2023]
Abstract
Small-conductance Ca2+-activated K+ (SK) channels are voltage-independent and are activated by Ca2+ binding to the calmodulin constitutively associated with the channels. Both the pore-forming subunits and the associated calmodulin are subject to phosphorylation. Here, we investigated the modulation of different SK channel subtypes by phosphorylation, using the cultured endothelial cells as a tool. We report that casein kinase 2 (CK2) negatively modulates the apparent Ca2+ sensitivity of SK1 and IK channel subtypes by more than 5-fold, whereas the apparent Ca2+ sensitivity of the SK3 and SK2 subtypes is only reduced by ∼2-fold, when heterologously expressed on the plasma membrane of cultured endothelial cells. The SK2 channel subtype exhibits limited cell surface expression in these cells, partly as a result of the phosphorylation of its C-terminus by cyclic AMP-dependent protein kinase (PKA). SK2 channels expressed on the ER and mitochondria membranes may protect against cell death. This work reveals the subtype-specific modulation of the apparent Ca2+ sensitivity and subcellular localization of SK channels by phosphorylation in cultured endothelial cells.
Collapse
Affiliation(s)
- Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Dezhi Kong
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
| | - Dong Wang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Razan Orfali
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Rinzhin T Sherpa
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Jennifer Totonchy
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Surya M Nauli
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA.
| |
Collapse
|
9
|
Hakim MA, Chum PP, Buchholz JN, Behringer EJ. Aging Alters Cerebrovascular Endothelial GPCR and K+ Channel Function: Divergent Role of Biological Sex. J Gerontol A Biol Sci Med Sci 2021; 75:2064-2073. [PMID: 31760422 DOI: 10.1093/gerona/glz275] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Indexed: 12/11/2022] Open
Abstract
Age-related dementia entails impaired blood flow to and throughout the brain due, in part, to reduced endothelial nitric oxide signaling. However, it is unknown whether sex affects cerebrovascular Gq-protein-coupled receptors (GPCRs) and K+ channels underlying endothelium-derived hyperpolarization (EDH) during progressive aging. Thus, we simultaneously evaluated intracellular Ca2+ ([Ca2+]i) and membrane potential (Vm) of intact endothelial tubes freshly isolated from posterior cerebral arteries of young (4-6 mo), middle-aged (12-16 mo), and old (24-28 mo) male and female C57BL/6 mice. Purinergic receptor function (vs. muscarinic) was dominant and enhanced for [Ca2+]i increases in old females versus old males. However, Ca2+-sensitive K+ channel function as defined by NS309-evoked Vm hyperpolarization was mildly impaired in females versus males during old age. This sex-based contrast in declined function of GPCRs and K+ channels to produce EDH may support a greater ability for physiological endothelial GPCR function to maintain optimal cerebral blood flow in females versus males during old age. As reflective of the pattern of cerebral blood flow decline in human subjects, inward-rectifying K+ (KIR) channel function decreased with progressive age regardless of sex. Combined age-related analyses masked male versus female aging and, contrary to expectation, hydrogen peroxide played a minimal role. Altogether, we conclude a sex-based divergence in cerebrovascular endothelial GPCR and K+ channel function while highlighting a previously unidentified form of age-related endothelial dysfunction as reduced KIR channel function.
Collapse
Affiliation(s)
- Md A Hakim
- Basic Sciences, Loma Linda University, California
| | | | | | | |
Collapse
|
10
|
Nam YW, Cui M, Orfali R, Viegas A, Nguyen M, Mohammed EHM, Zoghebi KA, Rahighi S, Parang K, Zhang M. Hydrophobic interactions between the HA helix and S4-S5 linker modulate apparent Ca 2+ sensitivity of SK2 channels. Acta Physiol (Oxf) 2021; 231:e13552. [PMID: 32865319 PMCID: PMC7736289 DOI: 10.1111/apha.13552] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/09/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022]
Abstract
AIM Small-conductance Ca2+ -activated potassium (SK) channels are activated exclusively by increases in intracellular Ca2+ that binds to calmodulin constitutively associated with the channel. Wild-type SK2 channels are activated by Ca2+ with an EC50 value of ~0.3 μmol/L. Here, we investigate hydrophobic interactions between the HA helix and the S4-S5 linker as a major determinant of channel apparent Ca2+ sensitivity. METHODS Site-directed mutagenesis, electrophysiological recordings and molecular dynamic (MD) simulations were utilized. RESULTS Mutations that decrease hydrophobicity at the HA-S4-S5 interface lead to Ca2+ hyposensitivity of SK2 channels. Mutations that increase hydrophobicity result in hypersensitivity to Ca2+ . The Ca2+ hypersensitivity of the V407F mutant relies on the interaction of the cognate phenylalanine with the S4-S5 linker in the SK2 channel. Replacing the S4-S5 linker of the SK2 channel with the S4-S5 linker of the SK4 channel results in loss of the hypersensitivity caused by V407F. This difference between the S4-S5 linkers of SK2 and SK4 channels can be partially attributed to I295 equivalent to a valine in the SK4 channel. A N293A mutation in the S4-S5 linker also increases hydrophobicity at the HA-S4-S5 interface and elevates the channel apparent Ca2+ sensitivity. The double N293A/V407F mutations generate a highly Ca2+ sensitive channel, with an EC50 of 0.02 μmol/L. The MD simulations of this double-mutant channel revealed a larger channel cytoplasmic gate. CONCLUSION The electrophysiological data and MD simulations collectively suggest a crucial role of the interactions between the HA helix and S4-S5 linker in the apparent Ca2+ sensitivity of SK2 channels.
Collapse
Affiliation(s)
- Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Meng Cui
- Department of Pharmaceutical Sciences, Northeastern University School of Pharmacy, Boston, MA, USA
| | - Razan Orfali
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Adam Viegas
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Misa Nguyen
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Eman H M Mohammed
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Khalid A Zoghebi
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Simin Rahighi
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Keykavous Parang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| |
Collapse
|
11
|
Wang H, Li S, Wang X, He C, Wang T, Wang Y, Guo W. Vasodilation activity of dipfluzine metabolites in isolated rat basilar arteries and their underlying mechanisms. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 79:103430. [PMID: 32544426 DOI: 10.1016/j.etap.2020.103430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/25/2020] [Accepted: 05/27/2020] [Indexed: 06/11/2023]
Abstract
Identifying the metabolites of a drug has become an indispensable task in the development of new drugs. Dipfluzine (Dip) is a promising candidate for the treatment of cerebral vascular diseases and has 5 metabolites (M1∼M5) in rat urine and liver microsomes, but their biological activity is still unknown. Because selective cerebral vasodilation is a main role of Dip, we investigated the vasodilation of Dip and its 5 metabolites in isolated Sprague-Dawley (SD) male rat basilar arteries preconstricted with high-K+ or 5-HT. The results showed that only M1 possessed concentration-dependent inhibitory activity on the vasoconstriction of arteries with or without the endothelium, and M1 has a more potent vasodilatory effect than Dip on both contraction models. Like Dip, the vasodilatory mechanisms of M1 may be not only related to receptor-operated and voltage-dependent calcium ion channels of smooth muscle cells but also to the release of NO and EDHF from endothelial cells and the opening of Ca2+-activated K+ channels and ATP-sensitive potassium ion channels. Unlike Dip, the vasodilation mechanism of M1 is also related to the opening of voltage-sensitive K+ channel. Together with more selectivity to non-VDCC than Dip, this may partially explain why M1 has stronger vasodilatory effects than Dip. The mechanisms of vasodilation of Dip and M1 may result from the combined action of these or other factors, especially blocking non-endothelium dependent non-VDCC and endothelium dependent IKCa channels. These results point to the possibility that M1 provides synergism for the clinical use of Dip, which may inform the synthesis of new drugs.
Collapse
Affiliation(s)
- Huan Wang
- College of Pharmacy, Hebei Medical University, Shijiazhuang, China
| | - Shiji Li
- Department of Digestive Endoscope, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, China
| | - Xiaohui Wang
- College of Pharmacy, Hebei Medical University, Shijiazhuang, China
| | - Chaoxing He
- College of Pharmacy, Hebei Medical University, Shijiazhuang, China
| | - Tianshi Wang
- College of Pharmacy, Hebei Medical University, Shijiazhuang, China
| | - Yongli Wang
- College of Pharmacy, Hebei Medical University, Shijiazhuang, China
| | - Wei Guo
- College of Pharmacy, Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
12
|
Chen T, Zhu J, Hang CH, Wang YH. The Potassium SK Channel Activator NS309 Protects Against Experimental Traumatic Brain Injury Through Anti-Inflammatory and Immunomodulatory Mechanisms. Front Pharmacol 2019; 10:1432. [PMID: 31849677 PMCID: PMC6895208 DOI: 10.3389/fphar.2019.01432] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 11/08/2019] [Indexed: 12/22/2022] Open
Abstract
Neuroinflammation plays important roles in neuronal cell death and functional deficits after TBI. Small conductance Ca2+-activated K+ channels (SK) have been shown to be potential therapeutic targets for treatment of neurological disorders, such as stroke and Parkinson’s disease (PD). The aim of the present study was to investigate the role of SK channels in an animal model of TBI induced by controlled cortical impact (CCI). The SK channels activator NS309 at a concentration of 2 mg/kg was administered by intraperitoneal injection, and no obviously organ-related toxicity of NS309 was found in Sprague-Dawley (SD) rats. Treatment with NS309 significantly reduced brain edema after TBI, but had no effect on contusion volume. This protection can be observed even when the administration was delayed by 4 h after injury. NS309 attenuated the TBI-induced deficits in neurological function, which was accompanied by the reduced neuronal apoptosis. The results of immunohistochemistry showed that NS309 decreased the number of neutrophils, lymphocytes, and microglia cells, with no effect on astrocytes. In addition, NS309 markedly decreased the levels of pro-inflammatory cytokines (IL-1β, IL-6 and TNF-α) and chemokines (MCP-1, MIP-2, and RANTES), but increased the levels of anti-inflammatory cytokines (IL-4, IL-10, and TGF-β1) after TBI. The results of RT-PCR and western blot showed that NS309 increased TSG-6 expression and inhibited NF-κB activation. Furthermore, knockdown of TSG-6 using in vivo transfection with TSG-6 specific shRNA partially reversed the protective and anti-inflammatory effects of NS309 against TBI. In summary, our results indicate that the SK channel activator NS309 could modulate inflammation-associated immune cells and cytokines via regulating the TSG-6/NF-κB pathway after TBI. The present study offers a new sight into the mechanisms responsible for SK channels activation with implications for the treatment of TBI.
Collapse
Affiliation(s)
- Tao Chen
- Department of Neurosurgery, The 904th Hospital of PLA, School of Medicine, Anhui Medical University, Wuxi, China.,Department of Neurosurgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jie Zhu
- Department of Neurosurgery, The 904th Hospital of PLA, School of Medicine, Anhui Medical University, Wuxi, China
| | - Chun-Hua Hang
- Department of Neurosurgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yu-Hai Wang
- Department of Neurosurgery, The 904th Hospital of PLA, School of Medicine, Anhui Medical University, Wuxi, China
| |
Collapse
|
13
|
Behringer EJ, Hakim MA. Functional Interaction among K Ca and TRP Channels for Cardiovascular Physiology: Modern Perspectives on Aging and Chronic Disease. Int J Mol Sci 2019; 20:ijms20061380. [PMID: 30893836 PMCID: PMC6471369 DOI: 10.3390/ijms20061380] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/07/2019] [Accepted: 03/15/2019] [Indexed: 12/16/2022] Open
Abstract
Effective delivery of oxygen and essential nutrients to vital organs and tissues throughout the body requires adequate blood flow supplied through resistance vessels. The intimate relationship between intracellular calcium ([Ca2+]i) and regulation of membrane potential (Vm) is indispensable for maintaining blood flow regulation. In particular, Ca2+-activated K+ (KCa) channels were ascertained as transducers of elevated [Ca2+]i signals into hyperpolarization of Vm as a pathway for decreasing vascular resistance, thereby enhancing blood flow. Recent evidence also supports the reverse role for KCa channels, in which they facilitate Ca2+ influx into the cell interior through open non-selective cation (e.g., transient receptor potential; TRP) channels in accord with robust electrical (hyperpolarization) and concentration (~20,000-fold) transmembrane gradients for Ca2+. Such an arrangement supports a feed-forward activation of Vm hyperpolarization while potentially boosting production of nitric oxide. Furthermore, in vascular types expressing TRP channels but deficient in functional KCa channels (e.g., collecting lymphatic endothelium), there are profound alterations such as downstream depolarizing ionic fluxes and the absence of dynamic hyperpolarizing events. Altogether, this review is a refined set of evidence-based perspectives focused on the role of the endothelial KCa and TRP channels throughout multiple experimental animal models and vascular types. We discuss the diverse interactions among KCa and TRP channels to integrate Ca2+, oxidative, and electrical signaling in the context of cardiovascular physiology and pathology. Building from a foundation of cellular biophysical data throughout a wide and diverse compilation of significant discoveries, a translational narrative is provided for readers toward the treatment and prevention of chronic, age-related cardiovascular disease.
Collapse
Affiliation(s)
- Erik J Behringer
- Department of Basic Sciences, 11041 Campus Street, Risley Hall, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Md A Hakim
- Department of Basic Sciences, 11041 Campus Street, Risley Hall, Loma Linda University, Loma Linda, CA 92350, USA.
| |
Collapse
|
14
|
Hakim MA, Behringer EJ. Simultaneous Measurements of Intracellular Calcium and Membrane Potential in Freshly Isolated and Intact Mouse Cerebral Endothelium. J Vis Exp 2019. [PMID: 30735188 DOI: 10.3791/58832] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cerebral arteries and their respective microcirculation deliver oxygen and nutrients to the brain via blood flow regulation. Endothelial cells line the lumen of blood vessels and command changes in vascular diameter as needed to meet the metabolic demand of neurons. Primary endothelial-dependent signaling pathways of hyperpolarization of membrane potential (Vm) and nitric oxide typically operate in parallel to mediate vasodilation and thereby increase blood flow. Although integral to coordinating vasodilation over several millimeters of vascular length, components of endothelium-derived hyperpolarization (EDH) have been historically difficult to measure. These components of EDH entail intracellular Ca2+ [Ca2+]i increases and subsequent activation of small- and intermediate conductance Ca2+-activated K+ (SKCa/IKCa) channels. Here, we present a simplified illustration of the isolation of fresh endothelium from mouse cerebral arteries; simultaneous measurements of endothelial [Ca2+]i and Vm using Fura-2 photometry and intracellular sharp electrodes, respectively; and a continuous superfusion of salt solutions and pharmacological agents under physiological conditions (pH 7.4, 37 °C). Posterior cerebral arteries from the Circle of Willis are removed free of the posterior communicating and the basilar arteries. Enzymatic digestion of cleaned posterior cerebral arterial segments and subsequent trituration facilitates removal of adventitia, perivascular nerves, and smooth muscle cells. Resulting posterior cerebral arterial endothelial "tubes" are then secured under a microscope and examined using a camera, photomultiplier tube, and one to two electrometers while under continuous superfusion. Collectively, this method can simultaneously measure changes in endothelial [Ca2+]i and Vm in discrete cellular locations, in addition to the spreading of EDH through gap junctions up to millimeter distances along the intact endothelium. This method is expected to yield a high-throughput analysis of the cerebral endothelial functions underlying mechanisms of blood flow regulation in the normal and diseased brain.
Collapse
Affiliation(s)
- Md A Hakim
- Department of Basic Sciences, Loma Linda University
| | | |
Collapse
|
15
|
Guerra G, Lucariello A, Perna A, Botta L, De Luca A, Moccia F. The Role of Endothelial Ca 2+ Signaling in Neurovascular Coupling: A View from the Lumen. Int J Mol Sci 2018; 19:E938. [PMID: 29561829 PMCID: PMC5979341 DOI: 10.3390/ijms19040938] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 03/16/2018] [Accepted: 03/17/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Neurovascular coupling (NVC) is the mechanism whereby an increase in neuronal activity (NA) leads to local elevation in cerebral blood flow (CBF) to match the metabolic requirements of firing neurons. Following synaptic activity, an increase in neuronal and/or astrocyte Ca2+ concentration leads to the synthesis of multiple vasoactive messengers. Curiously, the role of endothelial Ca2+ signaling in NVC has been rather neglected, although endothelial cells are known to control the vascular tone in a Ca2+-dependent manner throughout peripheral vasculature. METHODS We analyzed the literature in search of the most recent updates on the potential role of endothelial Ca2+ signaling in NVC. RESULTS We found that several neurotransmitters (i.e., glutamate and acetylcholine) and neuromodulators (e.g., ATP) can induce dilation of cerebral vessels by inducing an increase in endothelial Ca2+ concentration. This, in turn, results in nitric oxide or prostaglandin E2 release or activate intermediate and small-conductance Ca2+-activated K⁺ channels, which are responsible for endothelial-dependent hyperpolarization (EDH). In addition, brain endothelial cells express multiple transient receptor potential (TRP) channels (i.e., TRPC3, TRPV3, TRPV4, TRPA1), which induce vasodilation by activating EDH. CONCLUSIONS It is possible to conclude that endothelial Ca2+ signaling is an emerging pathway in the control of NVC.
Collapse
Affiliation(s)
- Germano Guerra
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, via F. De Santis, 86100 Campobasso, Italy.
| | - Angela Lucariello
- Department of Mental Health and Preventive Medicine, Section of Human Anatomy, University of Campania "L. Vanvitelli", 81100 Naples, Italy.
| | - Angelica Perna
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, via F. De Santis, 86100 Campobasso, Italy.
| | - Laura Botta
- Laboratory of General Physiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, via Forlanini 6, 27100 Pavia, Italy.
| | - Antonio De Luca
- Department of Mental Health and Preventive Medicine, Section of Human Anatomy, University of Campania "L. Vanvitelli", 81100 Naples, Italy.
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, via Forlanini 6, 27100 Pavia, Italy.
| |
Collapse
|
16
|
Jackson WF. Boosting the signal: Endothelial inward rectifier K + channels. Microcirculation 2018; 24. [PMID: 27652592 DOI: 10.1111/micc.12319] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 09/12/2016] [Indexed: 12/19/2022]
Abstract
Endothelial cells express a diverse array of ion channels including members of the strong inward rectifier family composed of KIR 2 subunits. These two-membrane spanning domain channels are modulated by their lipid environment, and exist in macromolecular signaling complexes with receptors, protein kinases and other ion channels. Inward rectifier K+ channel (KIR ) currents display a region of negative slope conductance at membrane potentials positive to the K+ equilibrium potential that allows outward current through the channels to be activated by membrane hyperpolarization, permitting KIR to amplify hyperpolarization induced by other K+ channels and ion transporters. Increases in extracellular K+ concentration activate KIR allowing them to sense extracellular K+ concentration and transduce this change into membrane hyperpolarization. These properties position KIR to participate in the mechanism of action of hyperpolarizing vasodilators and contribute to cell-cell conduction of hyperpolarization along the wall of microvessels. The expression of KIR in capillaries in electrically active tissues may allow KIR to sense extracellular K+ , contributing to functional hyperemia. Understanding the regulation of expression and function of microvascular endothelial KIR will improve our understanding of the control of blood flow in the microcirculation in health and disease and may provide new targets for the development of therapeutics in the future.
Collapse
Affiliation(s)
- William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
17
|
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
|
18
|
Zhang B, Naik JS, Jernigan NL, Walker BR, Resta TC. Reduced membrane cholesterol after chronic hypoxia limits Orai1-mediated pulmonary endothelial Ca 2+ entry. Am J Physiol Heart Circ Physiol 2017; 314:H359-H369. [PMID: 29101179 DOI: 10.1152/ajpheart.00540.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Endothelial dysfunction in chronic hypoxia (CH)-induced pulmonary hypertension is characterized by reduced store-operated Ca2+ entry (SOCE) and diminished Ca2+-dependent production of endothelium-derived vasodilators. We recently reported that SOCE in pulmonary arterial endothelial cells (PAECs) is tightly regulated by membrane cholesterol and that decreased membrane cholesterol is responsible for impaired SOCE after CH. However, the ion channels involved in cholesterol-sensitive SOCE are unknown. We hypothesized that cholesterol facilitates SOCE in PAECs through the interaction of Orai1 and stromal interaction molecule 1 (STIM1). The role of cholesterol in Orai1-mediated SOCE was initially assessed using CH exposure in rats (4 wk, 380 mmHg) as a physiological stimulus to decrease PAEC cholesterol. The effects of Orai1 inhibition with AnCoA4 on SOCE were examined in isolated PAEC sheets from control and CH rats after cholesterol supplementation, substitution of endogenous cholesterol with epicholesterol (Epichol), or vehicle treatment. Whereas cholesterol restored endothelial SOCE in CH rats, both Epichol and AnCoA4 attenuated SOCE only in normoxic controls. The Orai1 inhibitor had no further effect in cells pretreated with Epichol. Using cultured pulmonary endothelial cells to allow better mechanistic analysis of the molecular components of cholesterol-regulated SOCE, we found that Epichol, AnCoA4, and Orai1 siRNA each inhibited SOCE compared with their respective controls. Epichol had no additional effect after knockdown of Orai1. Furthermore, Epichol substitution significantly reduced STIM1-Orai1 interactions as assessed by a proximity ligation assay. We conclude that membrane cholesterol is required for the STIM1-Orai1 interaction necessary to elicit endothelial SOCE. Furthermore, reduced PAEC membrane cholesterol after CH limits Orai1-mediated SOCE. NEW & NOTEWORTHY This research demonstrates a novel contribution of cholesterol to regulate the interaction of Orai1 and stromal interaction molecule 1 required for pulmonary endothelial store-operated Ca2+ entry. The results provide a mechanistic basis for impaired pulmonary endothelial Ca2+ influx after chronic hypoxia that may contribute to pulmonary hypertension.
Collapse
Affiliation(s)
- Bojun Zhang
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Jay S Naik
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| |
Collapse
|
19
|
Zaborska KE, Wareing M, Austin C. Comparisons between perivascular adipose tissue and the endothelium in their modulation of vascular tone. Br J Pharmacol 2017; 174:3388-3397. [PMID: 27747871 PMCID: PMC5610163 DOI: 10.1111/bph.13648] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/16/2016] [Accepted: 09/28/2016] [Indexed: 01/06/2023] Open
Abstract
The endothelium is an established modulator of vascular tone; however, the recent discovery of the anti-contractile nature of perivascular adipose tissue (PVAT) suggests that the fat, which surrounds many blood vessels, can also modulate vascular tone. Both the endothelium and PVAT secrete vasoactive substances, which regulate vascular function. Many of these factors are common to both the endothelium and PVAT; therefore, this review will highlight the potential shared mechanisms in the modulation of vascular tone. Endothelial dysfunction is a hallmark of many vascular diseases, including hypertension and obesity. Moreover, PVAT dysfunction is now being reported in several cardio-metabolic disorders. Thus, this review will also discuss the mechanistic insights into endothelial and PVAT dysfunction in order to evaluate whether PVAT modulation of vascular contractility is similar to that of the endothelium in health and disease. LINKED ARTICLES This article is part of a themed section on Molecular Mechanisms Regulating Perivascular Adipose Tissue - Potential Pharmacological Targets? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.20/issuetoc.
Collapse
Affiliation(s)
- K E Zaborska
- Institute of Cardiovascular SciencesUniversity of ManchesterUK
| | - M Wareing
- Maternal and Fetal Health Research Centre, Institute of Human DevelopmentUniversity of ManchesterUK
| | - C Austin
- Faculty of Health and Social CareEdge Hill UniversityUK
| |
Collapse
|
20
|
Salomonsson M, Brasen JC, Sorensen CM. Role of renal vascular potassium channels in physiology and pathophysiology. Acta Physiol (Oxf) 2017; 221:14-31. [PMID: 28371470 DOI: 10.1111/apha.12882] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/10/2016] [Accepted: 03/22/2017] [Indexed: 12/31/2022]
Abstract
The control of renal vascular tone is important for the regulation of salt and water balance, blood pressure and the protection against damaging elevated glomerular pressure. The K+ conductance is a major factor in the regulation of the membrane potential (Vm ) in vascular smooth muscle (VSMC) and endothelial cells (EC). The vascular tone is controlled by Vm via its effect on the opening probability of voltage-operated Ca2+ channels (VOCC) in VSMC. When K+ conductance increases Vm becomes more negative and vasodilation follows, while deactivation of K+ channels leads to depolarization and vasoconstriction. K+ channels in EC indirectly participate in the control of vascular tone by endothelium-derived vasodilation. Therefore, by regulating the tone of renal resistance vessels, K+ channels have a potential role in the control of fluid homoeostasis and blood pressure as well as in the protection of the renal parenchyma. The main classes of K+ channels (calcium activated (KCa ), inward rectifier (Kir ), voltage activated (Kv ) and ATP sensitive (KATP )) have been found in the renal vessels. In this review, we summarize results available in the literature and our own studies in the field. We compare the ambiguous in vitro and in vivo results. We discuss the role of single types of K+ channels and the integrated function of several classes. We also deal with the possible role of renal vascular K+ channels in the pathophysiology of hypertension, diabetes mellitus and sepsis.
Collapse
Affiliation(s)
| | - J. C. Brasen
- Department of Electrical Engineering; Technical University of Denmark; Kgs. Lyngby Denmark
| | - C. M. Sorensen
- Department of Biomedical Sciences; Division of Renal and Vascular Physiology; University of Copenhagen; Copenhagen Denmark
| |
Collapse
|
21
|
Zhang B, Naik JS, Jernigan NL, Walker BR, Resta TC. Reduced membrane cholesterol limits pulmonary endothelial Ca 2+ entry after chronic hypoxia. Am J Physiol Heart Circ Physiol 2017; 312:H1176-H1184. [PMID: 28364016 DOI: 10.1152/ajpheart.00097.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/17/2017] [Accepted: 03/24/2017] [Indexed: 12/22/2022]
Abstract
Chronic hypoxia (CH)-induced pulmonary hypertension is associated with diminished production of endothelium-derived Ca2+-dependent vasodilators such as nitric oxide. Interestingly, ATP-induced endothelial Ca2+ entry as well as membrane cholesterol (Chol) are decreased in pulmonary arteries from CH rats (4 wk, barometric pressure = 380 Torr) compared with normoxic controls. Store-operated Ca2+ entry (SOCE) and depolarization-induced Ca2+ entry are major components of the response to ATP and are similarly decreased after CH. We hypothesized that membrane Chol facilitates both SOCE and depolarization-induced pulmonary endothelial Ca2+ entry and that CH attenuates these responses by decreasing membrane Chol. To test these hypotheses, we administered Chol or epicholesterol (Epichol) to acutely isolated pulmonary arterial endothelial cells (PAECs) from control and CH rats to either supplement or replace native Chol, respectively. The efficacy of membrane Chol manipulation was confirmed by filipin staining. Epichol greatly reduced ATP-induced Ca2+ influx in PAECs from control rats. Whereas Epichol similarly blunted endothelial SOCE in PAECs from both groups, Chol supplementation restored diminished SOCE in PAECs from CH rats while having no effect in controls. Similar effects of Chol manipulation on PAEC Ca2+ influx were observed in response to a depolarizing stimulus of KCl. Furthermore, KCl-induced Ca2+ entry was inhibited by the T-type Ca2+ channel antagonist mibefradil but not the L-type Ca2+ channel inhibitor diltiazem. We conclude that PAEC membrane Chol is required for ATP-induced Ca2+ entry and its two components, SOCE and depolarization-induced Ca2+ entry, and that reduced Ca2+ entry after CH may be due to loss of this key regulator.NEW & NOTEWORTHY This research is the first to examine the direct role of membrane cholesterol in regulating pulmonary endothelial agonist-induced Ca2+ entry and its components. The results provide a potential mechanism by which chronic hypoxia impairs pulmonary endothelial Ca2+ influx, which may contribute to pulmonary hypertension.
Collapse
Affiliation(s)
- Bojun Zhang
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Jay S Naik
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
22
|
Hearon CM, Kirby BS, Luckasen GJ, Larson DG, Dinenno FA. Endothelium-dependent vasodilatory signalling modulates α 1 -adrenergic vasoconstriction in contracting skeletal muscle of humans. J Physiol 2016; 594:7435-7453. [PMID: 27561916 DOI: 10.1113/jp272829] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/17/2016] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS 'Functional sympatholysis' describes the ability of contracting skeletal muscle to attenuate sympathetic vasoconstriction, and is critical to ensure proper blood flow and oxygen delivery to metabolically active skeletal muscle. The signalling mechanism responsible for sympatholysis in healthy humans is unknown. Evidence from animal models has identified endothelium-derived hyperpolarization (EDH) as a potential mechanism capable of attenuating sympathetic vasoconstriction. In this study, increasing endothelium-dependent signalling during exercise significantly enhanced the ability of contracting skeletal muscle to attenuate sympathetic vasoconstriction in humans. This is the first study in humans to identify endothelium-dependent regulation of sympathetic vasoconstriction in contracting skeletal muscle, and specifically supports a role for EDH-like vasodilatory signalling. Impaired functional sympatholysis is a common feature of cardiovascular ageing, hypertension and heart failure, and thus identifying fundamental mechanisms responsible for sympatholysis is clinically relevant. ABSTRACT Stimulation of α-adrenoceptors elicits vasoconstriction in resting skeletal muscle that is blunted during exercise in an intensity-dependent manner. In humans, the underlying mechanisms remain unclear. We tested the hypothesis that stimulating endothelium-dependent vasodilatory signalling will enhance the ability of contracting skeletal muscle to blunt α1 -adrenergic vasoconstriction. Changes in forearm vascular conductance (FVC; Doppler ultrasound, brachial intra-arterial pressure via catheter) to local intra-arterial infusion of phenylephrine (PE; α1 -adrenoceptor agonist) were calculated during (1) infusion of the endothelium-dependent vasodilators acetylcholine (ACh) and adenosine triphosphate (ATP), the endothelium-independent vasodilator (sodium nitroprusside, SNP), or potassium chloride (KCl) at rest; (2) mild or moderate intensity handgrip exercise; and (3) combined mild exercise + ACh, ATP, SNP, or KCl infusions in healthy adults. Robust vasoconstriction to PE was observed during vasodilator infusion alone and mild exercise, and this was blunted during moderate intensity exercise (ΔFVC: -34 ± 4 and -34 ± 3 vs. -13 ± 2%, respectively, P < 0.05). Infusion of ACh or ATP during mild exercise significantly attenuated PE vasoconstriction similar to levels observed during moderate exercise (ACh: -3 ± 4; ATP: -18 ± 4%). In contrast, infusion of SNP or KCl during mild exercise did not attenuate PE-mediated vasoconstriction (-32 ± 5 and -46 ± 3%). To further study the role of endothelium-dependent hyperpolarization (EDH), ACh trials were repeated with combined nitric oxide synthase and cyclooxygenase inhibition. Here, PE-mediated vasoconstriction was blunted at rest (blockade: -20 ± 5 vs. CONTROL -31 ± 3% vs.; P < 0.05) and remained blunted during exercise (blockade: -15 ± 5 vs. CONTROL -14 ± 5%). We conclude that stimulation of EDH-like vasodilatation can blunt α1 -adrenergic vasoconstriction in contracting skeletal muscle of humans.
Collapse
Affiliation(s)
- Christopher M Hearon
- Human Cardiovascular Physiology Laboratory, Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, 80523, USA
| | - Brett S Kirby
- Human Cardiovascular Physiology Laboratory, Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, 80523, USA
| | - Gary J Luckasen
- Medical Center of the Rockies Foundation, University of Colorado Health System, Loveland, CO, 80538, USA
| | - Dennis G Larson
- Medical Center of the Rockies Foundation, University of Colorado Health System, Loveland, CO, 80538, USA
| | - Frank A Dinenno
- Human Cardiovascular Physiology Laboratory, Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, 80523, USA.,Center for Cardiovascular Research, Colorado State University, Fort Collins, CO, 80523, USA
| |
Collapse
|
23
|
Linfante I, Cipolla MJ. Improving Reperfusion Therapies in the Era of Mechanical Thrombectomy. Transl Stroke Res 2016; 7:294-302. [PMID: 27221511 PMCID: PMC4929023 DOI: 10.1007/s12975-016-0469-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 05/08/2016] [Accepted: 05/10/2016] [Indexed: 12/27/2022]
Abstract
Recent positive clinical trials using mechanical thrombectomy proved that endovascular recanalization is an effective treatment for patients with acute stroke secondary to large vessel occlusions. The trials offer definite evidence that in acute ischemia recanalization is a powerful predictor of good outcome. However, even in the era of rapid and effective recanalization using endovascular approaches, the percentage of patients with good outcomes varies between 33 and 71 %. In addition, the number of patients who are eligible for endovascular thrombectomy is small and usually based on having salvageable tissue on imaging. There is therefore room for improvement to both enhance the effectiveness of current practice and expand treatment to a larger subset of stroke patients. In this review, we highlight some of the most promising approaches to improve endovascular therapy by combining with strategies to enhance collateral perfusion and vascular protection.
Collapse
Affiliation(s)
- Italo Linfante
- Miami Cardiac and Vascular Institute and Neuroscience Center, Baptist Hospital, Miami, FL, USA
| | - Marilyn J Cipolla
- Department of Neurological Sciences and Pharmacology, University of Vermont College of Medicine, 149 Beaumont Ave.; HSRF 416A, Burlington, VT, 05405, USA.
| |
Collapse
|
24
|
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
|
25
|
Behringer EJ, Segal SS. Membrane potential governs calcium influx into microvascular endothelium: integral role for muscarinic receptor activation. J Physiol 2015; 593:4531-48. [PMID: 26260126 DOI: 10.1113/jp271102] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 08/03/2015] [Indexed: 01/12/2023] Open
Abstract
In resistance arteries, coupling a rise of intracellular calcium concentration ([Ca(2+)]i) to endothelial cell hyperpolarization underlies smooth muscle cell relaxation and vasodilatation, thereby increasing tissue blood flow and oxygen delivery. A controversy persists as to whether changes in membrane potential (V(m)) alter endothelial cell [Ca(2+)]i. We tested the hypothesis that V(m) governs [Ca(2+)]i in endothelium of resistance arteries by performing Fura-2 photometry while recording and controlling V(m) of intact endothelial tubes freshly isolated from superior epigastric arteries of C57BL/6 mice. Under resting conditions, [Ca(2+)]i did not change when V(m) shifted from baseline (∼-40 mV) via exposure to 10 μM NS309 (hyperpolarization to ∼-80 mV), via equilibration with 145 mm [K(+)]o (depolarization to ∼-5 mV), or during intracellular current injection (±0.5 to 5 nA, 20 s pulses) while V(m) changed linearly between ∼-80 mV and +10 mV. In contrast, during the plateau (i.e. Ca(2+) influx) phase of the [Ca(2+)]i response to approximately half-maximal stimulation with 100 nm ACh (∼EC50), [Ca(2+)]i increased as V(m) hyperpolarized below -40 mV and decreased as V(m) depolarized above -40 mV. The magnitude of [Ca(2+)]i reduction during depolarizing current injections correlated with the amplitude of the plateau [Ca(2+)]i response to ACh. The effect of hyperpolarization on [Ca(2+)]i was abolished following removal of extracellular Ca(2+), was enhanced subtly by raising extracellular [Ca(2+)] from 2 mm to 10 mm and was reduced by half in endothelium of TRPV4(-/-) mice. Thus, during submaximal activation of muscarinic receptors, V(m) can modulate Ca(2+) entry through the plasma membrane in accord with the electrochemical driving force.
Collapse
Affiliation(s)
- Erik J Behringer
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, 65212, USA
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, 65212, USA.,Dalton Cardiovascular Research Center, Columbia, MO, 65211, USA
| |
Collapse
|
26
|
Tajbakhsh N, Sokoya EM. Compromised endothelium-dependent hyperpolarization-mediated dilations can be rescued by NS309 in obese Zucker rats. Microcirculation 2015; 21:747-53. [PMID: 25047389 DOI: 10.1111/micc.12157] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 07/15/2014] [Accepted: 07/17/2014] [Indexed: 11/30/2022]
Abstract
OBJECTIVE NO and a non-NO/prostacyclin EDH mechanism are major contributors of vascular tone and cerebral blood flow. However, the effect of metabolic syndrome on EDH-mediated responses in cerebral vessels remains unknown and may offer another avenue for therapeutic targeting. The purpose of this study was to investigate EDH-dependent responses in cerebral arteries during metabolic syndrome. METHODS EDH-dependent dilations were assessed in MCAs isolated from nondiabetic obese and lean Zucker rats in the presence and absence of NS309, an activator of SKCa and IKCa channels. IKCa channel expression and activity were assessed by western blotting and pressure myography, respectively. RESULTS EDH-mediated dilations were significantly attenuated in the obese compared to the lean Zucker rat MCA. Luminal delivery of 1 μM NS309 enhanced EDH-mediated responses in lean and obese Zucker cerebral vessels. Both dose-dependent dilations to luminal NS309 and IKCa protein expression in pooled cerebral arteries were comparable between the two groups. CONCLUSIONS Our results suggest that pharmacological targeting of IKCa channels can rescue EDH-mediated dilations in obese Zucker rat MCAs. Compromised EDH-mediated dilations in obesity are not due to impaired IKCa channel expression or activity.
Collapse
Affiliation(s)
- Negara Tajbakhsh
- Discipline of Medical Biotechnology, School of Medicine, Flinders University, Bedford Park, South Australia, Australia
| | | |
Collapse
|
27
|
Gokina NI, Bonev AD, Phillips J, Gokin AP, Veilleux K, Oppenheimer K, Goloman G. Impairment of IKCa channels contributes to uteroplacental endothelial dysfunction in rat diabetic pregnancy. Am J Physiol Heart Circ Physiol 2015; 309:H592-604. [PMID: 26092991 DOI: 10.1152/ajpheart.00901.2014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 06/19/2015] [Indexed: 12/18/2022]
Abstract
Diabetes in rat pregnancy is associated with impaired vasodilation of the maternal uteroplacental vasculature. In the present study, we explored the role of endothelial cell (EC) Ca(2+)-activated K(+) channels of small conductance (SKCa channels) and intermediate conductance (IKCa channels) in diabetes-induced uterine vascular dysfunction. Diabetes was induced by injection of streptozotocin to second-day pregnant rats and confirmed by the development of maternal hyperglycemia. Control rats were injected with citrate buffer. Changes in smooth muscle cell intracellular Ca(2+) concentration, membrane potential, and vasodilation induced by SKCa/IKCa channel activators were studied in uteroplacental arteries of control and diabetic rats. The impact of diabetes on SKCa- and IKCa-mediated currents was explored in freshly dissociated ECs. NS309 evoked a potent vasodilation that was effectively inhibited by TRAM-34 but not by apamin. NS309-induced smooth muscle cell intracellular Ca(2+) concentration, membrane potential, and dilator responses were significantly diminished by diabetes; N-cyclohexyl-N-2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-4-pyrimidinamine (CyPPA)-evoked responses were not affected. Ca(2+)-activated ion currents in ECs were insensitive to paxilline, markedly inhibited by charybdotoxin (ChTX), and diminished by apamin. NS309-induced EC currents were generated mostly due to activation of ChTX-sensitive channels. Maternal diabetes resulted in a significant reduction in ChTX-sensitive currents with no effect on apamin-sensitive or CyPPA-induced currents. We concluded that IKCa channels play a prevalent role over SKCa channels in the generation of endothelial K(+) currents and vasodilation of uteroplacental arteries. Impaired function of IKCa channels importantly contributes to diabetes-induced uterine endothelial dysfunction. Therapeutic restoration of IKCa channel function may be a novel strategy for improvement of maternal uteroplacental blood flow in pregnancies complicated by diabetes.
Collapse
Affiliation(s)
- Natalia I Gokina
- Department of Obstetrics, Gynecology and Reproductive Sciences, College of Medicine, University of Vermont, Burlington, Vermont; and
| | - Adrian D Bonev
- Department of Pharmacology, College of Medicine, University of Vermont, Burlington, Vermont
| | - Julie Phillips
- Department of Obstetrics, Gynecology and Reproductive Sciences, College of Medicine, University of Vermont, Burlington, Vermont; and
| | - Alexander P Gokin
- Department of Obstetrics, Gynecology and Reproductive Sciences, College of Medicine, University of Vermont, Burlington, Vermont; and
| | - Kelsey Veilleux
- Department of Obstetrics, Gynecology and Reproductive Sciences, College of Medicine, University of Vermont, Burlington, Vermont; and
| | - Karen Oppenheimer
- Department of Obstetrics, Gynecology and Reproductive Sciences, College of Medicine, University of Vermont, Burlington, Vermont; and
| | - Gabriela Goloman
- Department of Obstetrics, Gynecology and Reproductive Sciences, College of Medicine, University of Vermont, Burlington, Vermont; and
| |
Collapse
|
28
|
AlSuleimani YM, Hiley CR. The GPR55 agonist lysophosphatidylinositol relaxes rat mesenteric resistance artery and induces Ca(2+) release in rat mesenteric artery endothelial cells. Br J Pharmacol 2015; 172:3043-57. [PMID: 25652040 DOI: 10.1111/bph.13107] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 01/11/2015] [Accepted: 02/02/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Lysophosphatidylinositol (LPI), a lipid signalling molecule, activates GPR55 and elevates intracellular Ca(2+). Here, we examine the actions of LPI in the rat resistance mesenteric artery and Ca(2+) responses in endothelial cells isolated from the artery. EXPERIMENTAL APPROACH Vascular responses were studied using wire myographs. Single-cell fluorescence imaging was performed using a MetaFluor system. Hypotensive effects of LPI were assessed using a Biopac system. KEY RESULTS In isolated arteries, LPI-induced vasorelaxation was concentration- and endothelium-dependent and inhibited by CID 16020046, a GPR55 antagonist. The CB1 receptor antagonist AM 251 had no effect, whereas rimonabant and O-1918 significantly potentiated LPI responses. Vasorelaxation was reduced by charybdotoxin and iberiotoxin, alone or combined. LPI decreased systemic arterial pressure. GPR55 is expressed in rat mesenteric artery. LPI caused biphasic elevations of endothelial cell intracellular Ca(2+). Pretreatment with thapsigargin or 2-aminoethoxydiphenyl borate abolished both phases. The PLC inhibitor U73122 attenuated the initial phase and enhanced the second phase, whereas the Rho-associated kinase inhibitor Y-27632 abolished the late phase but not the early phase. CONCLUSIONS AND IMPLICATIONS LPI is an endothelium-dependent vasodilator in the rat small mesenteric artery and a hypotensive agent. The vascular response involves activation of Ca(2+)-sensitive K(+) channels and is not mediated by CB1 receptors, but unexpectedly enhanced by antagonists of the 'endothelial anandamide' receptor. In endothelial cells, LPI utilizes PLC-IP3 and perhaps ROCK-RhoA pathways to elevate intracellular Ca(2+). Overall, these findings support an endothelial site of action for LPI and suggest a possible role for GPR55 in vasculature.
Collapse
Affiliation(s)
- Y M AlSuleimani
- Department of Pharmacology, University of Cambridge, Cambridge, UK.,Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Alkoudh, Sultanate of Oman
| | - C R Hiley
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| |
Collapse
|
29
|
Abstract
Functional magnetic resonance imaging (fMRI) provides a unique view of the working human mind. The blood-oxygen-level-dependent (BOLD) signal, detected in fMRI, reflects changes in deoxyhemoglobin driven by localized changes in brain blood flow and blood oxygenation, which are coupled to underlying neuronal activity by a process termed neurovascular coupling. Over the past 10 years, a range of cellular mechanisms, including astrocytes, pericytes, and interneurons, have been proposed to play a role in functional neurovascular coupling. However, the field remains conflicted over the relative importance of each process, while key spatiotemporal features of BOLD response remain unexplained. Here, we review current candidate neurovascular coupling mechanisms and propose that previously overlooked involvement of the vascular endothelium may provide a more complete picture of how blood flow is controlled in the brain. We also explore the possibility and consequences of conditions in which neurovascular coupling may be altered, including during postnatal development, pathological states, and aging, noting relevance to both stimulus-evoked and resting-state fMRI studies.
Collapse
Affiliation(s)
- Elizabeth M C Hillman
- Departments of Biomedical Engineering and Radiology and the Kavli Institute for Brain Science, Columbia University, New York, NY 10027;
| |
Collapse
|
30
|
Chen YJ, Wallace BK, Yuen N, Jenkins DP, Wulff H, O'Donnell ME. Blood-brain barrier KCa3.1 channels: evidence for a role in brain Na uptake and edema in ischemic stroke. Stroke 2014; 46:237-44. [PMID: 25477223 DOI: 10.1161/strokeaha.114.007445] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND PURPOSE KCa3.1, a calcium-activated potassium channel, regulates ion and fluid secretion in the lung and gastrointestinal tract. It is also expressed on vascular endothelium where it participates in blood pressure regulation. However, the expression and physiological role of KCa3.1 in blood-brain barrier (BBB) endothelium has not been investigated. BBB endothelial cells transport Na(+) and Cl(-) from the blood into the brain transcellularly through the co-operation of multiple cotransporters, exchangers, pumps, and channels. In the early stages of cerebral ischemia, when the BBB is intact, edema formation occurs by processes involving increased BBB transcellular Na(+) transport. This study evaluated whether KCa3.1 is expressed on and participates in BBB ion transport. METHODS The expression of KCa3.1 on cultured cerebral microvascular endothelial cells, isolated microvessels, and brain sections was evaluated by Western blot and immunohistochemistry. Activity of KCa3.1 on cerebral microvascular endothelial cells was examined by K(+) flux assays and patch-clamp. Magnetic resonance spectroscopy and MRI were used to measure brain Na(+) uptake and edema formation in rats with focal ischemic stroke after TRAM-34 treatment. RESULTS KCa3.1 current and channel protein were identified on bovine cerebral microvascular endothelial cells and freshly isolated rat microvessels. In situ KCa3.1 expression on BBB endothelium was confirmed in rat and human brain sections. TRAM-34 treatment significantly reduced Na(+) uptake, and cytotoxic edema in the ischemic brain. CONCLUSIONS BBB endothelial cells exhibit KCa3.1 protein and activity and pharmacological blockade of KCa3.1 seems to provide an effective therapeutic approach for reducing cerebral edema formation in the first 3 hours of ischemic stroke.
Collapse
Affiliation(s)
- Yi-Je Chen
- From the Department of Pharmacology (Y.-J.C., D.P.J., H.W.) and Department of Physiology and Membrane Biology (B.K.W., N.Y., M.E.O.), University of California, Davis.
| | - Breanna K Wallace
- From the Department of Pharmacology (Y.-J.C., D.P.J., H.W.) and Department of Physiology and Membrane Biology (B.K.W., N.Y., M.E.O.), University of California, Davis
| | - Natalie Yuen
- From the Department of Pharmacology (Y.-J.C., D.P.J., H.W.) and Department of Physiology and Membrane Biology (B.K.W., N.Y., M.E.O.), University of California, Davis
| | - David P Jenkins
- From the Department of Pharmacology (Y.-J.C., D.P.J., H.W.) and Department of Physiology and Membrane Biology (B.K.W., N.Y., M.E.O.), University of California, Davis
| | - Heike Wulff
- From the Department of Pharmacology (Y.-J.C., D.P.J., H.W.) and Department of Physiology and Membrane Biology (B.K.W., N.Y., M.E.O.), University of California, Davis
| | - Martha E O'Donnell
- From the Department of Pharmacology (Y.-J.C., D.P.J., H.W.) and Department of Physiology and Membrane Biology (B.K.W., N.Y., M.E.O.), University of California, Davis
| |
Collapse
|
31
|
Cui M, Qin G, Yu K, Bowers MS, Zhang M. Targeting the Small- and Intermediate-Conductance Ca-Activated Potassium Channels: The Drug-Binding Pocket at the Channel/Calmodulin Interface. Neurosignals 2014; 22:65-78. [PMID: 25300231 DOI: 10.1159/000367896] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 08/26/2014] [Indexed: 11/19/2022] Open
Abstract
The small- and intermediate-conductance Ca(2+)-activated potassium (SK/IK) channels play important roles in the regulation of excitable cells in both the central nervous and cardiovascular systems. Evidence from animal models has implicated SK/IK channels in neurological conditions such as ataxia and alcohol use disorders. Further, genome-wide association studies have suggested that cardiovascular abnormalities such as arrhythmias and hypertension are associated with single nucleotide polymorphisms that occur within the genes encoding the SK/IK channels. The Ca(2+) sensitivity of the SK/IK channels stems from a constitutively bound Ca(2+)-binding protein: calmodulin. Small-molecule positive modulators of SK/IK channels have been developed over the past decade, and recent structural studies have revealed that the binding pocket of these positive modulators is located at the interface between the channel and calmodulin. SK/IK channel positive modulators can potentiate channel activity by enhancing the coupling between Ca(2+) sensing via calmodulin and mechanical opening of the channel. Here, we review binding pocket studies that have provided structural insight into the mechanism of action for SK/IK channel positive modulators. These studies lay the foundation for structure-based drug discovery efforts that can identify novel SK/IK channel positive modulators.
Collapse
Affiliation(s)
- Meng Cui
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Va., USA
| | | | | | | | | |
Collapse
|
32
|
Kochukov MY, Balasubramanian A, Abramowitz J, Birnbaumer L, Marrelli SP. Activation of endothelial transient receptor potential C3 channel is required for small conductance calcium-activated potassium channel activation and sustained endothelial hyperpolarization and vasodilation of cerebral artery. J Am Heart Assoc 2014; 3:jah3649. [PMID: 25142058 PMCID: PMC4310376 DOI: 10.1161/jaha.114.000913] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Transient receptor potential C3 (TRPC3) has been demonstrated to be involved in the regulation of vascular tone through endothelial cell (EC) hyperpolarization and endothelium‐dependent hyperpolarization–mediated vasodilation. However, the mechanism by which TRPC3 regulates these processes remains unresolved. We tested the hypothesis that endothelial receptor stimulation triggers rapid TRPC3 trafficking to the plasma membrane, where it provides the source of Ca2+ influx for small conductance calcium‐activated K+ (SKCa) channel activation and sustained EC hyperpolarization. Methods and Results Pressurized artery studies were performed with isolated mouse posterior cerebral artery. Treatment with a selective TRPC3 blocker (Pyr3) produced significant attenuation of endothelium‐dependent hyperpolarization–mediated vasodilation and endothelial Ca2+ response (EC‐specific Ca2+ biosensor) to intraluminal ATP. Pyr3 treatment also resulted in a reduced ATP‐stimulated global Ca2+ and Ca2+ influx in primary cultures of cerebral endothelial cells. Patch‐clamp studies with freshly isolated cerebral ECs demonstrated 2 components of EC hyperpolarization and K+ current activation in response to ATP. The early phase was dependent on intermediate conductance calcium‐activated K+ channel activation, whereas the later sustained phase relied on SKCa channel activation. The SKCa channel–dependent phase was completely blocked with TRPC3 channel inhibition or in ECs of TRPC3 knockout mice and correlated with increased trafficking of TRPC3 (but not SKCa channel) to the plasma membrane. Conclusions We propose that TRPC3 dynamically regulates SKCa channel activation through receptor‐dependent trafficking to the plasma membrane, where it provides the source of Ca2+ influx for sustained SKCa channel activation, EC hyperpolarization, and endothelium‐dependent hyperpolarization–mediated vasodilation.
Collapse
Affiliation(s)
- Mikhail Y Kochukov
- Department of Anesthesiology, Baylor College of Medicine, Houston, TX (M.Y.K., A.B., S.P.M.)
| | - Adithya Balasubramanian
- Department of Anesthesiology, Baylor College of Medicine, Houston, TX (M.Y.K., A.B., S.P.M.)
| | - Joel Abramowitz
- Division of Intramural Research, National Institute of Environmental Health Sciences Research, Triangle Park, NC (J.A., L.B.)
| | - Lutz Birnbaumer
- Division of Intramural Research, National Institute of Environmental Health Sciences Research, Triangle Park, NC (J.A., L.B.)
| | - Sean P Marrelli
- Department of Anesthesiology, Baylor College of Medicine, Houston, TX (M.Y.K., A.B., S.P.M.) Department of Physiology and Biophysics and Graduate Program in Physiology, Cardiovascular Sciences Track, Baylor College of Medicine, Houston, TX (S.P.M.)
| |
Collapse
|
33
|
Chen BR, Kozberg MG, Bouchard MB, Shaik MA, Hillman EMC. A critical role for the vascular endothelium in functional neurovascular coupling in the brain. J Am Heart Assoc 2014; 3:e000787. [PMID: 24926076 PMCID: PMC4309064 DOI: 10.1161/jaha.114.000787] [Citation(s) in RCA: 244] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background The functional modulation of blood flow in the brain is critical for brain health and is the basis of contrast in functional magnetic resonance imaging. There is evident coupling between increases in neuronal activity and increases in local blood flow; however, many aspects of this neurovascular coupling remain unexplained by current models. Based on the rapid dilation of distant pial arteries during cortical functional hyperemia, we hypothesized that endothelial signaling may play a key role in the long‐range propagation of vasodilation during functional hyperemia in the brain. Although well characterized in the peripheral vasculature, endothelial involvement in functional neurovascular coupling has not been demonstrated. Methods and Results We combined in vivo exposed‐cortex multispectral optical intrinsic signal imaging (MS‐OISI) with a novel in vivo implementation of the light‐dye technique to record the cortical hemodynamic response to somatosensory stimulus in rats before and after spatially selective endothelial disruption. We demonstrate that discrete interruption of endothelial signaling halts propagation of stimulus‐evoked vasodilation in pial arteries, and that wide‐field endothelial disruption in pial arteries significantly attenuates the hemodynamic response to stimulus, particularly the early, rapid increase and peak in hyperemia. Conclusions Involvement of endothelial pathways in functional neurovascular coupling provides new explanations for the spatial and temporal features of the hemodynamic response to stimulus and could explain previous results that were interpreted as evidence for astrocyte‐mediated control of functional hyperemia. Our results unify many aspects of blood flow regulation in the brain and body and prompt new investigation of direct links between systemic cardiovascular disease and neural deficits.
Collapse
Affiliation(s)
- Brenda R Chen
- Laboratory for Functional Optical Imaging, Departments of Biomedical Engineering and Radiology, Columbia University, New York, 10027, NY (B.R.C., M.G.K., M.B.B., M.A.S., E.C.H.)
| | - Mariel G Kozberg
- Laboratory for Functional Optical Imaging, Departments of Biomedical Engineering and Radiology, Columbia University, New York, 10027, NY (B.R.C., M.G.K., M.B.B., M.A.S., E.C.H.)
| | - Matthew B Bouchard
- Laboratory for Functional Optical Imaging, Departments of Biomedical Engineering and Radiology, Columbia University, New York, 10027, NY (B.R.C., M.G.K., M.B.B., M.A.S., E.C.H.)
| | - Mohammed A Shaik
- Laboratory for Functional Optical Imaging, Departments of Biomedical Engineering and Radiology, Columbia University, New York, 10027, NY (B.R.C., M.G.K., M.B.B., M.A.S., E.C.H.)
| | - Elizabeth M C Hillman
- Laboratory for Functional Optical Imaging, Departments of Biomedical Engineering and Radiology, Columbia University, New York, 10027, NY (B.R.C., M.G.K., M.B.B., M.A.S., E.C.H.)
| |
Collapse
|
34
|
Gauthier KM, Campbell WB, McNeish AJ. Regulation of KCa2.3 and endothelium-dependent hyperpolarization (EDH) in the rat middle cerebral artery: the role of lipoxygenase metabolites and isoprostanes. PeerJ 2014; 2:e414. [PMID: 24949235 PMCID: PMC4060036 DOI: 10.7717/peerj.414] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/15/2014] [Indexed: 01/21/2023] Open
Abstract
Background and Purpose. In rat middle cerebral arteries, endothelium-dependent hyperpolarization (EDH) is mediated by activation of calcium-activated potassium (KCa) channels specifically KCa2.3 and KCa3.1. Lipoxygenase (LOX) products function as endothelium-derived hyperpolarizing factors (EDHFs) in rabbit arteries by stimulating KCa2.3. We investigated if LOX products contribute to EDH in rat cerebral arteries. Methods. Arachidonic acid (AA) metabolites produced in middle cerebral arteries were measured using HPLC and LC/MS. Vascular tension and membrane potential responses to SLIGRL were simultaneously recorded using wire myography and intracellular microelectrodes. Results. SLIGRL, an agonist at PAR2 receptors, caused EDH that was inhibited by a combination of KCa2.3 and KCa3.1 blockade. Non-selective LOX-inhibition reduced EDH, whereas inhibition of 12-LOX had no effect. Soluble epoxide hydrolase (sEH) inhibition enhanced the KCa2.3 component of EDH. Following NO synthase (NOS) inhibition, the KCa2.3 component of EDH was absent. Using HPLC, middle cerebral arteries metabolized 14C-AA to 15- and 12-LOX products under control conditions. With NOS inhibition, there was little change in LOX metabolites, but increased F-type isoprostanes. 8-iso-PGF2α inhibited the KCa2.3 component of EDH. Conclusions. LOX metabolites mediate EDH in rat middle cerebral arteries. Inhibition of sEH increases the KCa2.3 component of EDH. Following NOS inhibition, loss of KCa2.3 function is independent of changes in LOX production or sEH inhibition but due to increased isoprostane production and subsequent stimulation of TP receptors. These findings have important implications in diseases associated with loss of NO signaling such as stroke; where inhibition of sEH and/or isoprostane formation may of benefit.
Collapse
Affiliation(s)
- Kathryn M Gauthier
- Department of Pharmacology and Toxicology, Medical College of Wisconsin , Milwaukee, WI , USA
| | - William B Campbell
- Department of Pharmacology and Toxicology, Medical College of Wisconsin , Milwaukee, WI , USA
| | - Alister J McNeish
- Reading School of Pharmacy, University of Reading , Reading, Berkshire , UK
| |
Collapse
|
35
|
Dora KA, Garland CJ. Linking hyperpolarization to endothelial cell calcium events in arterioles. Microcirculation 2013; 20:248-56. [PMID: 23311991 DOI: 10.1111/micc.12041] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 01/08/2013] [Indexed: 01/14/2023]
Abstract
Our understanding of the relationship between EC membrane potential and Ca(2+) entry has been shaped historically by data from cells in culture. Membrane hyperpolarization was associated with raised cytoplasmic [Ca(2+) ] ascribed to the increase in the inward electrochemical gradient for Ca(2+) , as ECs are generally thought to lack VGCC. Ca(2+) influx was assumed to reflect the presence of an undefined Ca(2+) "leak" channel, although the original research articles with isolated ECs did not elucidate which Ca(2+) influx channel was involved or indeed if a transporter might contribute. Overall, these early studies left many unanswered questions, not least whether a similar mechanism operates in native ECs that are coupled to each other and, in many smaller arteries and arterioles, to the adjacent vascular SMCs via gap junctions. This review discusses whether Ca(2+) leak through constitutively active EC Ca(2+) channels or a more defined, gated pathway might underlie the reported link between enhanced Ca(2+) entry and hyperpolarization. Electrophysiological evidence from ECs in isolation is compared with those in intact arteries and arterioles and the possible physiological relevance of EC Ca(2+) entry driven by hyperpolarization discussed.
Collapse
Affiliation(s)
- Kim A Dora
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK.
| | | |
Collapse
|
36
|
Cipolla MJ, Sweet JG, Gokina NI, White SL, Nelson MT. Mechanisms of enhanced basal tone of brain parenchymal arterioles during early postischemic reperfusion: role of ET-1-induced peroxynitrite generation. J Cereb Blood Flow Metab 2013; 33:1486-92. [PMID: 23778163 PMCID: PMC3790940 DOI: 10.1038/jcbfm.2013.99] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 04/30/2013] [Accepted: 05/10/2013] [Indexed: 12/14/2022]
Abstract
The contributions of vasoconstrictors (endothelin-1 (ET-1), peroxynitrite) and endothelium-dependent vasodilatory mechanisms to basal tone were investigated in parenchymal arterioles (PAs) after early postischemic reperfusion. Transient middle cerebral artery occlusion (tMCAO) was induced for 2 hours with 30 minutes reperfusion in male Wistar rats and compared with ischemia alone (permanent MCAO (pMCAO); 2.5 hours) or sham controls. Changes in lumen diameter of isolated and pressurized PAs were compared. Quantitative PCR was used to measure endothelin type B (ETB) receptors. Constriction to intravascular pressure ('basal tone') was not affected by tMCAO or pMCAO. However, constriction to inhibitors of endothelial cell, small- (SK) and intermediate- (IK) conductance, Ca(2+)-sensitive K(+) channels (apamin and TRAM-34, respectively) were significantly enhanced in PAs from tMCAO compared with pMCAO or sham. Addition of the ETB agonist sarafotoxin caused constriction in PAs from tMCAO but not from sham animals (21 ± 4% versus 3 ± 3% at 1 nmol/L; P<0.01) that was inhibited by the peroxynitrite scavenger FeTMPyP (5,10,15,20-tetrakis (N-methyl-4'-pyridyl) porphinato iron (III) chloride) (100 μmol/L). Expression of ETB receptors was not found on PA smooth muscle, suggesting that constriction to sarafotoxin after tMCAO was due to peroxynitrite and not ETB receptor expression. The maintenance of basal tone in PAs after tMCAO may restrict flow to the ischemic region and contribute to infarct expansion.
Collapse
Affiliation(s)
- Marilyn J Cipolla
- 1] Department of Neurological Sciences, University of Vermont College of Medicine, Burlington, Vermont, USA [2] Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont College of Medicine, Burlington, Vermont, USA [3] Department of Pharmacology, University of Vermont College of Medicine, Burlington, Vermont, USA
| | | | | | | | | |
Collapse
|
37
|
D'Alessandro G, Catalano M, Sciaccaluga M, Chece G, Cipriani R, Rosito M, Grimaldi A, Lauro C, Cantore G, Santoro A, Fioretti B, Franciolini F, Wulff H, Limatola C. KCa3.1 channels are involved in the infiltrative behavior of glioblastoma in vivo. Cell Death Dis 2013; 4:e773. [PMID: 23949222 PMCID: PMC3763441 DOI: 10.1038/cddis.2013.279] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 06/25/2013] [Accepted: 07/02/2013] [Indexed: 01/03/2023]
Abstract
Glioblastoma multiforme (GBM) is a diffuse brain tumor characterized by high infiltration in the brain parenchyma rendering the tumor difficult to eradicate by neurosurgery. Efforts to identify molecular targets involved in the invasive behavior of GBM suggested ion channel inhibition as a promising therapeutic approach. To determine if the Ca(2+)-dependent K(+) channel KCa3.1 could represent a key element for GBM brain infiltration, human GL-15 cells were xenografted into the brain of SCID mice that were then treated with the specific KCa3.1 blocker TRAM-34 (1-((2-chlorophenyl) (diphenyl)methyl)-1H-pyrazole). After 5 weeks of treatment, immunofluorescence analyses of cerebral slices revealed reduced tumor infiltration and astrogliosis surrounding the tumor, compared with untreated mice. Significant reduction of tumor infiltration was also observed in the brain of mice transplanted with KCa3.1-silenced GL-15 cells, indicating a direct effect of TRAM-34 on GBM-expressed KCa3.1 channels. As KCa3.1 channels are also expressed on microglia, we investigated the effects of TRAM-34 on microglia activation in GL-15 transplanted mice and found a reduction of CD68 staining in treated mice. Similar results were observed in vitro where TRAM-34 reduced both phagocytosis and chemotactic activity of primary microglia exposed to GBM-conditioned medium. Taken together, these results indicate that KCa3.1 activity has an important role in GBM invasiveness in vivo and that its inhibition directly affects glioma cell migration and reduces astrocytosis and microglia activation in response to tumor-released factors. KCa3.1 channel inhibition therefore constitutes a potential novel therapeutic approach to reduce GBM spreading into the surrounding tissue.
Collapse
Affiliation(s)
- G D'Alessandro
- Institute Pasteur, Cenci Bolognetti Foundation, Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Qian X, Francis M, Solodushko V, Earley S, Taylor MS. Recruitment of dynamic endothelial Ca2+ signals by the TRPA1 channel activator AITC in rat cerebral arteries. Microcirculation 2013; 20:138-48. [PMID: 22928941 DOI: 10.1111/micc.12004] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 08/24/2012] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Stimulation of endothelial TRP channels, specifically TRPA1, promotes vasodilation of cerebral arteries through activation of Ca2+ -dependent effectors along the myoendothelial interface. However, presumed TRPA1-triggered endothelial Ca2+ signals have not been described. We investigated whether TRPA1 activation induces specific spatial and temporal changes in Ca2+ signals along the intima that correlates with incremental vasodilation. METHODS Confocal imaging, immunofluorescence staining, and custom image analysis were employed. RESULTS We found that endothelial cells of rat cerebral arteries exhibit widespread basal Ca2+ dynamics (44 ± 6 events/minute from 26 ± 3 distinct sites in a 3.6 × 10(4) μm2 field). The TRPA1 activator AITC increased Ca2+ signals in a concentration-dependent manner, soliciting new events at distinct sites. Origination of these new events corresponded spatially with TRPA1 densities in IEL holes, and the events were prevented by the TRPA1 inhibitor HC-030031. Concentration-dependent expansion of Ca2+ events in response to AITC correlated precisely with dilation of pressurized cerebral arteries (p = 0.93 by F-test). Correspondingly, AITC caused rapid endothelium-dependent suppression of asynchronous Ca2+ waves in subintimal smooth muscle. CONCLUSIONS Our findings indicate that factors that stimulate TRPA1 channels expand Ca2+ signal-effector coupling at discrete sites along the endothelium to evoke graded cerebral artery vasodilation.
Collapse
Affiliation(s)
- Xun Qian
- Department of Physiology, University of South Alabama College of Medicine, Mobile, Alabama, USA
| | | | | | | | | |
Collapse
|
39
|
Pires PW, Dams Ramos CM, Matin N, Dorrance AM. The effects of hypertension on the cerebral circulation. Am J Physiol Heart Circ Physiol 2013; 304:H1598-614. [PMID: 23585139 DOI: 10.1152/ajpheart.00490.2012] [Citation(s) in RCA: 265] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Maintenance of brain function depends on a constant blood supply. Deficits in cerebral blood flow are linked to cognitive decline, and they have detrimental effects on the outcome of ischemia. Hypertension causes alterations in cerebral artery structure and function that can impair blood flow, particularly during an ischemic insult or during periods of low arterial pressure. This review will focus on the historical discoveries, novel developments, and knowledge gaps in 1) hypertensive cerebral artery remodeling, 2) vascular function with emphasis on myogenic reactivity and endothelium-dependent dilation, and 3) blood-brain barrier function. Hypertensive artery remodeling results in reduction in the lumen diameter and an increase in the wall-to-lumen ratio in most cerebral arteries; this is linked to reduced blood flow postischemia and increased ischemic damage. Many factors that are increased in hypertension stimulate remodeling; these include the renin-angiotensin-aldosterone system and reactive oxygen species levels. Endothelial function, vital for endothelium-mediated dilation and regulation of myogenic reactivity, is impaired in hypertension. This is a consequence of alterations in vasodilator mechanisms involving nitric oxide, epoxyeicosatrienoic acids, and ion channels, including calcium-activated potassium channels and transient receptor potential vanilloid channel 4. Hypertension causes blood-brain barrier breakdown by mechanisms involving inflammation, oxidative stress, and vasoactive circulating molecules. This exposes neurons to cytotoxic molecules, leading to neuronal loss, cognitive decline, and impaired recovery from ischemia. As the population ages and the incidence of hypertension, stroke, and dementia increases, it is imperative that we gain a better understanding of the control of cerebral artery function in health and disease.
Collapse
Affiliation(s)
- Paulo W Pires
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | | | | | | |
Collapse
|
40
|
Unstructured to structured transition of an intrinsically disordered protein peptide in coupling Ca²⁺-sensing and SK channel activation. Proc Natl Acad Sci U S A 2013; 110:4828-33. [PMID: 23487779 DOI: 10.1073/pnas.1220253110] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Most proteins, such as ion channels, form well-organized 3D structures to carry out their specific functions. A typical voltage-gated potassium channel subunit has six transmembrane segments (S1-S6) to form the voltage-sensing domain and the pore domain. Conformational changes of these domains result in opening of the channel pore. Intrinsically disordered (ID) proteins/peptides are considered equally important for the protein functions. However, it is difficult to explore the structural features underlying the functions of ID proteins/peptides by conventional methods, such as X-ray crystallography, because of the flexibility of their secondary structures. Unlike voltage-gated potassium channels, families of small- and intermediate-conductance Ca(2+)-activated potassium (SK/IK) channels with important roles in regulating membrane excitability are activated exclusively by Ca(2+)-bound calmodulin (CaM). Upon binding of Ca(2+) to CaM, a 2 × 2 structure forms between CaM and the CaM-binding domain. A channel fragment that connects S6 and the CaM-binding domain is not visible in the protein crystal structure, suggesting that this fragment is an ID fragment. Here we show that the conformation of the ID fragment in SK channels becomes readily identifiable in the presence of NS309, the most potent compound that potentiates the channel activities. This well-defined conformation of the ID fragment, stabilized by NS309, increases the channel open probability at a given Ca(2+) concentration. Our results demonstrate that the ID fragment, itself a target for drugs modulating SK channel activities, plays a unique role in coupling Ca(2+) sensing by CaM and mechanical opening of SK channels.
Collapse
|
41
|
Zhang M, Pascal JM, Schumann M, Armen RS, Zhang JF. Identification of the functional binding pocket for compounds targeting small-conductance Ca²⁺-activated potassium channels. Nat Commun 2013; 3:1021. [PMID: 22929778 PMCID: PMC3563359 DOI: 10.1038/ncomms2017] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 07/20/2012] [Indexed: 12/14/2022] Open
Abstract
Small- and intermediate-conductance Ca2+-activated potassium channels, activated by Ca2+-bound calmodulin, play an important role in regulating membrane excitability. These channels are also linked to clinical abnormalities. A tremendous amount of effort has been devoted to developing small molecule compounds targeting these channels. However, these compounds often suffer from low potency and lack of selectivity, hindering their potentials for clinical use. A key contributing factor is the lack of knowledge of the binding site(s) for these compounds. Here we demonstrate by X-ray crystallography that the binding pocket for the compounds of the 1-EBIO class is located at the calmodulin-channel interface. We show that, based on structure data and molecular docking, mutations of the channel can effectively change the potency of these compounds. Our results provide insight into the molecular nature of the binding pocket and its contribution to the potency and selectivity of the compounds of the 1-EBIO class.
Collapse
Affiliation(s)
- Miao Zhang
- Department of Molecular Physiology and Biophysics, Thomas Jefferson University, 1020 Locust Street, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | |
Collapse
|
42
|
Kochukov MY, Balasubramanian A, Noel RC, Marrelli SP. Role of TRPC1 and TRPC3 channels in contraction and relaxation of mouse thoracic aorta. J Vasc Res 2012; 50:11-20. [PMID: 23095462 DOI: 10.1159/000342461] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 07/04/2012] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND/AIMS Canonical transient receptor potential (TRPC) channels modulate membrane potential and intracellular Ca(2+). We examined the role of TRPC1 and TRPC3 channels in vasocontraction and relaxation in mouse aorta. METHODS Vasocontraction and relaxation of aorta from wild-type (WT), TRPC1 KO and TRPC3 knockout (KO) mice were measured for phenylephrine (Phe) and carbachol (CCh). Intracellular Ca(2+) was measured in primary aorta endothelial cells (EC) and whole cell K(+) current in freshly isolated smooth muscle cells (SMC). RESULTS AND CONCLUSION TRPC1 KO aorta showed increased vasocontraction to Phe compared to WT and TRPC3 KO aorta due to diminished role of BK(Ca) channels. BK(Ca) mRNA (aorta) and whole cell current (SMC) were reduced versus WT. Contraction in WT aorta was increased to TRPC1 KO level by BK(Ca) channel inhibition. Relaxation to CCh was reduced in TRPC1 KO and TRPC3 KO aortas with concomitant reduction in EC Ca(2+) response. Pyr3 (TRPC3 blocker) reduced the Ca(2+) response to CCh in EC from WT, but not TRPC3 KO mice. In summary, TRPC1 attenuates receptor-mediated contraction through activation and/or expression of SMC BK(Ca) channels while TRPC3 does not contribute to receptor-mediated constriction. Both TRPC1 and TRPC3 participate in EC Ca(2+) influx and vasorelaxation of aorta.
Collapse
Affiliation(s)
- M Y Kochukov
- Department of Anesthesiology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
43
|
McNeish AJ, Jimenez-Altayo F, Cottrell GS, Garland CJ. Statins and selective inhibition of Rho kinase protect small conductance calcium-activated potassium channel function (K(Ca)2.3) in cerebral arteries. PLoS One 2012; 7:e46735. [PMID: 23056429 PMCID: PMC3466297 DOI: 10.1371/journal.pone.0046735] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 09/01/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND In rat middle cerebral and mesenteric arteries the K(Ca)2.3 component of endothelium-dependent hyperpolarization (EDH) is lost following stimulation of thromboxane (TP) receptors, an effect that may contribute to the endothelial dysfunction associated with cardiovascular disease. In cerebral arteries, K(Ca)2.3 loss is associated with NO synthase inhibition, but is restored if TP receptors are blocked. The Rho/Rho kinase pathway is central for TP signalling and statins indirectly inhibit this pathway. The possibility that Rho kinase inhibition and statins sustain K(Ca)2.3 hyperpolarization was investigated in rat middle cerebral arteries (MCA). METHODS MCAs were mounted in a wire myograph. The PAR2 agonist, SLIGRL was used to stimulate EDH responses, assessed by simultaneous measurement of smooth muscle membrane potential and tension. TP expression was assessed with rt-PCR and immunofluorescence. RESULTS Immunofluorescence detected TP in the endothelial cell layer of MCA. Vasoconstriction to the TP agonist, U46619 was reduced by Rho kinase inhibition. TP receptor stimulation lead to loss of K(Ca)2.3 mediated hyperpolarization, an effect that was reversed by Rho kinase inhibitors or simvastatin. K(Ca)2.3 activity was lost in L-NAME-treated arteries, but was restored by Rho kinase inhibition or statin treatment. The restorative effect of simvastatin was blocked after incubation with geranylgeranyl-pyrophosphate to circumvent loss of isoprenylation. CONCLUSIONS Rho/Rho kinase signalling following TP stimulation and L-NAME regulates endothelial cell K(Ca)2.3 function. The ability of statins to prevent isoprenylation and perhaps inhibit of Rho restores/protects the input of K(Ca)2.3 to EDH in the MCA, and represents a beneficial pleiotropic effect of statin treatment.
Collapse
Affiliation(s)
- Alister J McNeish
- Reading School of Pharmacy, University of Reading, Reading, Berkshire, United Kingdom.
| | | | | | | |
Collapse
|
44
|
Kuiper EFE, Nelemans A, Luiten P, Nijholt I, Dolga A, Eisel U. K(Ca)2 and k(ca)3 channels in learning and memory processes, and neurodegeneration. Front Pharmacol 2012; 3:107. [PMID: 22701424 PMCID: PMC3372087 DOI: 10.3389/fphar.2012.00107] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 05/19/2012] [Indexed: 11/13/2022] Open
Abstract
Calcium-activated potassium (KCa) channels are present throughout the central nervous system as well as many peripheral tissues. Activation of KCa channels contribute to maintenance of the neuronal membrane potential and was shown to underlie the afterhyperpolarization (AHP) that regulates action potential firing and limits the firing frequency of repetitive action potentials. Different subtypes of KCa channels were anticipated on the basis of their physiological and pharmacological profiles, and cloning revealed two well defined but phylogenetic distantly related groups of channels. The group subject of this review includes both the small conductance KCa2 channels (KCa2.1, KCa2.2, and KCa2.3) and the intermediate-conductance (KCa3.1) channel. These channels are activated by submicromolar intracellular Ca2+ concentrations and are voltage independent. Of all KCa channels only the KCa2 channels can be potently but differentially blocked by the bee-venom apamin. In the past few years modulation of KCa channel activation revealed new roles for KCa2 channels in controlling dendritic excitability, synaptic functioning, and synaptic plasticity. Furthermore, KCa2 channels appeared to be involved in neurodegeneration, and learning and memory processes. In this review, we focus on the role of KCa2 and KCa3 channels in these latter mechanisms with emphasis on learning and memory, Alzheimer’s disease and on the interplay between neuroinflammation and different neurotransmitters/neuromodulators, their signaling components and KCa channel activation.
Collapse
Affiliation(s)
- Els F E Kuiper
- Molecular Neurobiology, University of Groningen Groningen, Netherlands
| | | | | | | | | | | |
Collapse
|
45
|
Kerr PM, Tam R, Narang D, Potts K, McMillan D, McMillan K, Plane F. Endothelial calcium-activated potassium channels as therapeutic targets to enhance availability of nitric oxide. Can J Physiol Pharmacol 2012; 90:739-52. [PMID: 22626011 DOI: 10.1139/y2012-075] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The vascular endothelium plays a critical role in vascular health by controlling arterial diameter, regulating local cell growth, and protecting blood vessels from the deleterious consequences of platelet aggregation and activation of inflammatory responses. Circulating chemical mediators and physical forces act directly on the endothelium to release diffusible relaxing factors, such as nitric oxide (NO), and to elicit hyperpolarization of the endothelial cell membrane potential, which can spread to the surrounding smooth muscle cells via gap junctions. Endothelial hyperpolarization, mediated by activation of calcium-activated potassium (K(Ca)) channels, has generally been regarded as a distinct pathway for smooth muscle relaxation. However, recent evidence supports a role for endothelial K(Ca) channels in production of endothelium-derived NO, and indicates that pharmacological activation of these channels can enhance NO-mediated responses. In this review we summarize the current data on the functional role of endothelial K(Ca) channels in regulating NO-mediated changes in arterial diameter and NO production, and explore the tempting possibility that these channels may represent a novel avenue for therapeutic intervention in conditions associated with reduced NO availability such as hypertension, hypercholesterolemia, smoking, and diabetes mellitus.
Collapse
Affiliation(s)
- Paul M Kerr
- Department of Pharmacology, 9-62 Medical Sciences Building, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | | | | | | | | | | | | |
Collapse
|
46
|
Sorensen CM, Braunstein TH, Holstein-Rathlou NH, Salomonsson M. Role of vascular potassium channels in the regulation of renal hemodynamics. Am J Physiol Renal Physiol 2012; 302:F505-18. [DOI: 10.1152/ajprenal.00052.2011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
K+ conductance is a major determinant of membrane potential ( Vm) in vascular smooth muscle (VSMC) and endothelial cells (EC). The vascular tone is controlled by Vm through the action of voltage-operated Ca2+ channels (VOCC) in VSMC. Increased K+ conductance leads to hyperpolarization and vasodilation, while inactivation of K+ channels causes depolarization and vasoconstriction. K+ channels in EC indirectly participate in the control of vascular tone by several mechanisms, e.g., release of nitric oxide and endothelium-derived hyperpolarizing factor. In the kidney, a change in the activity of one or more classes of K+ channels will lead to a change in hemodynamic resistance and therefore of renal blood flow and glomerular filtration pressure. Through these effects, the activity of renal vascular K+ channels influences renal salt and water excretion, fluid homeostasis, and ultimately blood pressure. Four main classes of K+ channels [calcium activated (KCa), inward rectifier (Kir), voltage activated (KV), and ATP sensitive (KATP)] are found in the renal vasculature. Several in vitro experiments have suggested a role for individual classes of K+ channels in the regulation of renal vascular function. Results from in vivo experiments are sparse. We discuss the role of the different classes of renal vascular K+ channels and their possible role in the integrated function of the renal microvasculature. Since several pathological conditions, among them hypertension, are associated with alterations in K+ channel function, the role of renal vascular K+ channels in the control of salt and water excretion deserves attention.
Collapse
Affiliation(s)
- Charlotte Mehlin Sorensen
- Institute of Biomedical Sciences, Division of Renal and Vascuar Physiology, The Panum Institute, and
| | - Thomas Hartig Braunstein
- Danish National Research Foundation Center for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark
| | | | - Max Salomonsson
- Institute of Biomedical Sciences, Division of Renal and Vascuar Physiology, The Panum Institute, and
| |
Collapse
|
47
|
Vascular Targets for Ischemic Stroke Treatment. Transl Stroke Res 2012. [DOI: 10.1007/978-1-4419-9530-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
48
|
Abstract
It has been known for more than 60 years, and suspected for over 100, that alveolar hypoxia causes pulmonary vasoconstriction by means of mechanisms local to the lung. For the last 20 years, it has been clear that the essential sensor, transduction, and effector mechanisms responsible for hypoxic pulmonary vasoconstriction (HPV) reside in the pulmonary arterial smooth muscle cell. The main focus of this review is the cellular and molecular work performed to clarify these intrinsic mechanisms and to determine how they are facilitated and inhibited by the extrinsic influences of other cells. Because the interaction of intrinsic and extrinsic mechanisms is likely to shape expression of HPV in vivo, we relate results obtained in cells to HPV in more intact preparations, such as intact and isolated lungs and isolated pulmonary vessels. Finally, we evaluate evidence regarding the contribution of HPV to the physiological and pathophysiological processes involved in the transition from fetal to neonatal life, pulmonary gas exchange, high-altitude pulmonary edema, and pulmonary hypertension. Although understanding of HPV has advanced significantly, major areas of ignorance and uncertainty await resolution.
Collapse
Affiliation(s)
- J T Sylvester
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, The Johns Hopkins University School ofMedicine, Baltimore, Maryland, USA.
| | | | | | | |
Collapse
|
49
|
Davis CM, Siler DA, Alkayed NJ. Endothelium-derived hyperpolarizing factor in the brain: influence of sex, vessel size and disease state. ACTA ACUST UNITED AC 2011; 7:293-303. [PMID: 21612351 DOI: 10.2217/whe.11.26] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The endothelial layer of cells lining the intimal surface of blood vessels is essential for vascular function. The endothelium releases multiple vasodilator and protective factors, including nitric oxide, prostacyclin and endothelium-derived hyperpolarizing factor; an imbalance in these factors predisposes individuals to vascular diseases such as stroke. These factors are differentially regulated by vessel size, sex hormones and disease state, therefore playing differential roles in different tissues following vascular injury. In particular, the endothelium-derived hyperpolarizing factor candidate termed epoxyeicosatrienoic acid, plays a prominent role in microvessel function, especially after ischemia, thereby making this signaling pathway an attractive target for therapy in vascular disease, including stroke.
Collapse
Affiliation(s)
- Catherine M Davis
- Cerebrovascular Research Division, Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR 97239-3098, USA
| | | | | |
Collapse
|
50
|
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
|