1
|
Mierke CT. Mechanosensory entities and functionality of endothelial cells. Front Cell Dev Biol 2024; 12:1446452. [PMID: 39507419 PMCID: PMC11538060 DOI: 10.3389/fcell.2024.1446452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
The endothelial cells of the blood circulation are exposed to hemodynamic forces, such as cyclic strain, hydrostatic forces, and shear stress caused by the blood fluid's frictional force. Endothelial cells perceive mechanical forces via mechanosensors and thus elicit physiological reactions such as alterations in vessel width. The mechanosensors considered comprise ion channels, structures linked to the plasma membrane, cytoskeletal spectrin scaffold, mechanoreceptors, and junctional proteins. This review focuses on endothelial mechanosensors and how they alter the vascular functions of endothelial cells. The current state of knowledge on the dysregulation of endothelial mechanosensitivity in disease is briefly presented. The interplay in mechanical perception between endothelial cells and vascular smooth muscle cells is briefly outlined. Finally, future research avenues are highlighted, which are necessary to overcome existing limitations.
Collapse
|
2
|
Osikoya O, Hula N, da Silva RDNO, Goulopoulou S. Perivascular Adipose Tissue and Uterine Artery Adaptations to Pregnancy. Microcirculation 2024; 31:e12857. [PMID: 38826057 DOI: 10.1111/micc.12857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 03/28/2024] [Accepted: 04/22/2024] [Indexed: 06/04/2024]
Abstract
Pregnancy is characterized by longitudinal maternal, physiological adaptations to support the development of a fetus. One of the cardinal maternal adaptations during a healthy pregnancy is a progressive increase in uterine artery blood flow. This facilitates sufficient blood supply for the development of the placenta and the growing fetus. Regional hemodynamic changes in the uterine circulation, such as a vast reduction in uterine artery resistance, are mainly facilitated by changes in uterine artery reactivity and myogenic tone along with remodeling of the uterine arteries. These regional changes in vascular reactivity have been attributed to pregnancy-induced adaptations of cell-to-cell communication mechanisms, with an emphasis on the interaction between endothelial and vascular smooth muscle cells. Perivascular adipose tissue (PVAT) is considered the fourth layer of the vascular wall and contributes to the regulation of vascular reactivity in most vascular beds and most species. This review focuses on mechanisms of uterine artery reactivity and the role of PVAT in pregnancy-induced maternal vascular adaptations, with an emphasis on the uterine circulation.
Collapse
Affiliation(s)
- Oluwatobiloba Osikoya
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA
| | - Nataliia Hula
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University, Loma Linda, California, USA
| | - Renée de Nazaré Oliveira da Silva
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University, Loma Linda, California, USA
| | - Styliani Goulopoulou
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University, Loma Linda, California, USA
- Department of Gynecology and Obstetrics, Loma Linda University, Loma Linda, California, USA
| |
Collapse
|
3
|
Coccarelli A, Pant S, Polydoros I, Harraz OF. A new model for evaluating pressure-induced vascular tone in small cerebral arteries. Biomech Model Mechanobiol 2024; 23:271-286. [PMID: 37925376 PMCID: PMC10901969 DOI: 10.1007/s10237-023-01774-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/17/2023] [Indexed: 11/06/2023]
Abstract
The capacity of small cerebral arteries (SCAs) to adapt to pressure fluctuations has a fundamental physiological role and appears to be relevant in different pathological conditions. Here, we present a new computational model for quantifying the link, and its contributors, between luminal pressure and vascular tone generation in SCAs. This is assembled by combining a chemical sub-model, representing pressure-induced smooth muscle cell (SMC) signalling, with a mechanical sub-model for the tone generation and its transduction at tissue level. The devised model can accurately reproduce the impact of luminal pressure on different cytoplasmic components involved in myogenic signalling, both in the control case and when combined with some specific pharmacological interventions. Furthermore, the model is also able to capture and predict experimentally recorded pressure-outer diameter relationships obtained for vessels under control conditions, both in a Ca2 + -free bath and under drug inhibition. The modularity of the proposed framework allows the integration of new components for the study of a broad range of processes involved in the vascular function.
Collapse
Affiliation(s)
- Alberto Coccarelli
- Zienkiewicz Institute for Modelling, Data and AI, Faculty of Science and Engineering, Swansea University, Swansea, UK.
| | - Sanjay Pant
- Zienkiewicz Institute for Modelling, Data and AI, Faculty of Science and Engineering, Swansea University, Swansea, UK
| | - Ioannis Polydoros
- Zienkiewicz Institute for Modelling, Data and AI, Faculty of Science and Engineering, Swansea University, Swansea, UK
| | - Osama F Harraz
- Department of Pharmacology, Larner College of Medicine, and Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, USA
| |
Collapse
|
4
|
Turner SR, Al‐Ghabkari A, Carlson DA, Chappellaz M, Sutherland C, Haystead TAJ, Cole WC, MacDonald JA. Death-associated protein kinase 3 regulates the myogenic reactivity of cerebral arteries. Exp Physiol 2023; 108:986-997. [PMID: 37084168 PMCID: PMC10988501 DOI: 10.1113/ep090631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 03/28/2023] [Indexed: 04/22/2023]
Abstract
NEW FINDINGS What is the central question of this study? DAPK3 contributes to the Ca2+ -sensitization of vascular smooth muscle contraction: does this protein kinase participate in the myogenic response of cerebral arteries? What is the main finding and its importance? Small molecule inhibitors of DAPK3 effectively block the myogenic responses of cerebral arteries. HS38-dependent changes to vessel constriction occur independent of LC20 phosphorylation, and therefore DAPK3 appears to operate via the actin cytoskeleton. A role for DAPK3 in the myogenic response was not previously reported, and the results support a potential new therapeutic target in the cerebrovascular system. ABSTRACT The vascular smooth muscle (VSM) of resistance blood vessels is a target of intrinsic autoregulatory responses to increased intraluminal pressure, the myogenic response. In the brain, the myogenic reactivity of cerebral arteries is critical to homeostatic blood flow regulation. Here we provide the first evidence to link the death-associated protein kinase 3 (DAPK3) to the myogenic response of rat and human cerebral arteries. DAPK3 is a Ser/Thr kinase involved in Ca2+ -sensitization mechanisms of smooth muscle contraction. Ex vivo administration of a specific DAPK3 inhibitor (i.e., HS38) could attenuate vessel constrictions invoked by serotonin as well as intraluminal pressure elevation. The HS38-dependent dilatation was not associated with any change in myosin light chain (LC20) phosphorylation. The results suggest that DAPK3 does not regulate Ca2+ sensitization pathways during the myogenic response of cerebral vessels but rather operates to control the actin cytoskeleton. A slow return of myogenic tone was observed during the sustained ex vivo exposure of cerebral arteries to HS38. Recovery of tone was associated with greater LC20 phosphorylation that suggests intrinsic signalling compensation in response to attenuation of DAPK3 activity. Additional experiments with VSM cells revealed HS38- and siDAPK-dependent effects on the actin cytoskeleton and focal adhesion kinase phosphorylation status. The translational importance of DAPK3 to the human cerebral vasculature was noted, with robust expression of the protein kinase and significant HS38-dependent attenuation of myogenic reactivity found for human pial vessels.
Collapse
Affiliation(s)
- Sara R. Turner
- Department of Biochemistry & Molecular Biology, Cumming School of MedicineUniversity of CalgaryCalgaryABCanada
| | - Abdulhameed Al‐Ghabkari
- Department of Biochemistry & Molecular Biology, Cumming School of MedicineUniversity of CalgaryCalgaryABCanada
| | - David A. Carlson
- Department of Pharmacology & Cancer BiologyDuke University School of MedicineDurhamNCUSA
| | - Mona Chappellaz
- Department of Biochemistry & Molecular Biology, Cumming School of MedicineUniversity of CalgaryCalgaryABCanada
| | - Cindy Sutherland
- Department of Biochemistry & Molecular Biology, Cumming School of MedicineUniversity of CalgaryCalgaryABCanada
- Department of Physiology and Pharmacology, Cumming School of MedicineUniversity of CalgaryCalgaryABCanada
| | - Timothy A. J. Haystead
- Department of Pharmacology & Cancer BiologyDuke University School of MedicineDurhamNCUSA
| | - William C. Cole
- Department of Physiology and Pharmacology, Cumming School of MedicineUniversity of CalgaryCalgaryABCanada
| | - Justin A. MacDonald
- Department of Biochemistry & Molecular Biology, Cumming School of MedicineUniversity of CalgaryCalgaryABCanada
| |
Collapse
|
5
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
6
|
Coccarelli A, Nelson MD. Modeling Reactive Hyperemia to Better Understand and Assess Microvascular Function: A Review of Techniques. Ann Biomed Eng 2023; 51:479-492. [PMID: 36709231 PMCID: PMC9928923 DOI: 10.1007/s10439-022-03134-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/25/2022] [Indexed: 01/30/2023]
Abstract
Reactive hyperemia is a well-established technique for the non-invasive evaluation of the peripheral microcirculatory function, measured as the magnitude of limb re-perfusion after a brief period of ischemia. Despite widespread adoption by researchers and clinicians alike, many uncertainties remain surrounding interpretation, compounded by patient-specific confounding factors (such as blood pressure or the metabolic rate of the ischemic limb). Mathematical modeling can accelerate our understanding of the physiology underlying the reactive hyperemia response and guide in the estimation of quantities which are difficult to measure experimentally. In this work, we aim to provide a comprehensive guide for mathematical modeling techniques that can be used for describing the key phenomena involved in the reactive hyperemia response, alongside their limitations and advantages. The reported methodologies can be used for investigating specific reactive hyperemia aspects alone, or can be combined into a computational framework to be used in (pre-)clinical settings.
Collapse
Affiliation(s)
- Alberto Coccarelli
- Zienkiewicz Centre for Computational Engineering, Faculty of Science and Engineering, Swansea University, Swansea, UK.
| | - Michael D Nelson
- Department of Kinesiology, University of Texas at Arlington, Arlington, TX, USA
| |
Collapse
|
7
|
Johnson AC. Hippocampal Vascular Supply and Its Role in Vascular Cognitive Impairment. Stroke 2023; 54:673-685. [PMID: 36848422 PMCID: PMC9991081 DOI: 10.1161/strokeaha.122.038263] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/22/2022] [Indexed: 03/01/2023]
Abstract
The incidence of age-related dementia is increasing as the world population ages and due to lack of effective treatments for dementia. Vascular contributions to cognitive impairment and dementia are increasing as the prevalence of pathologies associated with cerebrovascular disease rise, including chronic hypertension, diabetes, and ischemic stroke. The hippocampus is a bilateral deep brain structure that is central to learning, memory, and cognitive function and highly susceptible to hypoxic/ischemic injury. Compared with cortical brain regions such as the somatosensory cortex, less is known about the function of the hippocampal vasculature that is critical in maintaining neurocognitive health. This review focuses on the hippocampal vascular supply, presenting what is known about hippocampal hemodynamics and blood-brain barrier function during health and disease, and discusses evidence that supports its contribution to vascular cognitive impairment and dementia. Understanding vascular-mediated hippocampal injury that contributes to memory dysfunction during healthy aging and cerebrovascular disease is essential to develop effective treatments to slow cognitive decline. The hippocampus and its vasculature may represent one such therapeutic target to mitigate the dementia epidemic.
Collapse
Affiliation(s)
- Abbie C Johnson
- Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington
| |
Collapse
|
8
|
Mazzuca MQ, Buyukcelebi K, Lin C, Khalil RA. Increased Ca 2+-dependent intrinsic tone and arterial stiffness in mesenteric microvessels of hypertensive pregnant rats. Biochem Pharmacol 2023; 208:115353. [PMID: 36435203 PMCID: PMC9877182 DOI: 10.1016/j.bcp.2022.115353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/26/2022]
Abstract
Preeclampsia is a pregnancy-related hypertensive disorder (HTN-Preg) with unclear mechanisms. We have shown increased vascular reactivity to extrinsic vasoconstrictors in HTN-Preg rats. Here, we test whether microvascular intrinsic tone and arterial stiffness could contribute to HTN-Preg, and examined the underlying cellular mechanisms. On gestational day 19, BP was recorded in normal pregnant (Preg) rats and Preg rats with reduced uterine perfusion pressure (RUPP), and mesenteric microvessels were mounted on a pressure myograph for measurement of intrinsic tone, simultaneous changes in [Ca2+]i (fura-2 340/380 ratio), and arterial stiffness. Arteries were incubated in Ca2+-containing and 0 Ca2+ (2 mM EGTA) Krebs, pressurized at 10 to 110 mmHg in 10 mmHg increments, and the % change in vessel diameter from initial diameter at 10 mmHg was analyzed for measurement of total (active + passive) intrinsic tone and passive intrinsic response, respectively. The passive response was then subtracted from the total intrinsic tone to determine the active myogenic tone. The strain-stress relationship was also constructed as a measure of arterial stiffness. BP was higher in RUPP vs Preg rats. In Ca2+-containing Krebs, increases in intraluminal pressure caused smaller increases in diameter and greater increases in [Ca2+]i in microvessels of RUPP vs Preg rats, suggesting increased Ca2+-dependent myogenic tone. In 0 Ca2+ Krebs, increases in pressure also caused less increases in diameter in microvessels of RUPP vs Preg rats, but with no changes in [Ca2+]i, suggesting changes in the structure and mechanics of the arterial wall. The total and passive strain-stress relationship was shifted to the left in microvessels of RUPP vs Preg rats, suggesting increased arterial wall stiffness. Histology and immunohistochemistry showed greater vascular wall thickness and collagen-I staining in RUPP vs Preg rats, supporting changes in the wall architecture and structural proteins. The increased active myogenic tone and underlying increases in Ca2+ signaling as well as the increased passive intrinsic response, arterial stiffness and collagen-I in the mesenteric microvessels could play a role in the regulation of blood flow to the splanchnic region and the increased vascular resistance and BP in HTN-Preg.
Collapse
Affiliation(s)
- Marc Q Mazzuca
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, United States
| | - Kadir Buyukcelebi
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, United States
| | - Chen Lin
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, United States
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
9
|
Wang S, Tang C, Liu Y, Border JJ, Roman RJ, Fan F. Impact of impaired cerebral blood flow autoregulation on cognitive impairment. FRONTIERS IN AGING 2022; 3:1077302. [PMID: 36531742 PMCID: PMC9755178 DOI: 10.3389/fragi.2022.1077302] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 11/23/2022] [Indexed: 11/16/2023]
Abstract
Although the causes of cognitive impairment are multifactorial, emerging evidence indicates that cerebrovascular dysfunction plays an essential role in dementia. One of the most critical aspects of cerebrovascular dysfunction is autoregulation of cerebral blood flow (CBF), mainly mediated by the myogenic response, which is often impaired in dementia individuals with comorbidities, such as diabetes and hypertension. However, many unsolved questions remain. How do cerebrovascular networks coordinately modulate CBF autoregulation in health and disease? Does poor CBF autoregulation have an impact on cognitive impairment, and what are the underlying mechanisms? This review summarizes the cerebral vascular structure and myogenic (a three-phase model), metabolic (O2, CO2, adenosine, and H+), and endothelial (shear stress) factors in the regulation of CBF; and the consequences of CBF dysautoregulation. Other factors contributing to cerebrovascular dysfunction, such as impaired functional hyperemia and capillary abnormalities, are included as well. Moreover, this review highlights recent studies from our lab in terms of novel mechanisms involved in CBF autoregulation and addresses a hypothesis that there is a three-line of defense for CBF autoregulation in the cerebral vasculature.
Collapse
Affiliation(s)
- Shaoxun Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Chengyun Tang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Yedan Liu
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Jane J Border
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
10
|
Morse CJ, Boerman EM, McDonald MW, Padilla J, Olver TD. The role of nitric oxide in flow-induced and myogenic responses in 1A, 2A, and 3A branches of the porcine middle cerebral artery. J Appl Physiol (1985) 2022; 133:1228-1236. [PMID: 36227166 PMCID: PMC9715271 DOI: 10.1152/japplphysiol.00209.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 09/13/2022] [Accepted: 10/09/2022] [Indexed: 12/15/2022] Open
Abstract
Myogenic and flow-induced reactivity contribute to cerebral autoregulation, with potentially divergent roles for smaller versus larger arteries. The present study tested the hypotheses that compared with first-order (1A) branches of the middle cerebral artery, second- and third-order branches (2A and 3A, respectively) exhibit greater myogenic reactivity but reduced flow-induced constriction. Furthermore, nitric oxide synthase (NOS) inhibition may amplify myogenic reactivity and abolish instances of flow-induced dilation. Isolated porcine cerebral arteries mounted in a pressure myograph were exposed to incremental increases in intraluminal pressure (40-120 mmHg; n = 41) or flow (1-1,170 µL/min; n = 31). Intraluminal flows were adjusted to achieve 5, 10, 20, and 40 dyn/cm2 of wall shear stress at 60 mmHg. Myogenic tone was greater in 3A versus 1A arteries (P < 0.05). There was an inverse relationship between myogenic reactivity and passive arterial diameter (P < 0.01). NOS inhibition increased basal tone to a lesser extent in 3A versus 1A arteries (P < 0.01) but did not influence myogenic reactivity (P = 0.49). Increasing flow decreased luminal diameter (P ≤ 0.01), with increased vasoconstriction at 10-40 dyn/cm2 of shear stress (P < 0.01). However, relative responses were similar between 1A, 2A, and 3A arteries (P = 0.40) with and without NOS inhibition conditions (P ≥ 0.29). Whereas NOS inhibition increases basal myogenic tone, and myogenic reactivity was less in smaller versus larger arteries (range = ∼100-550 µM), neither NOS inhibition nor luminal diameter influences flow-induced constriction in porcine cerebral arteries.NEW & NOTEWORTHY This study demonstrated size-dependent heterogeneity in myogenic reactivity in porcine cerebral arteries. Smaller branches of the middle cerebral artery exhibited increased myogenic reactivity, but attenuated NOS-dependent increases in myogenic tone compared with larger branches. Flow-dependent regulation does not exhibit the same variation; diameter-independent flow-induced vasoconstrictions occur across all branch orders and are not affected by NOS inhibition. Conceptually, flow-induced vasoconstriction contributes to cerebral autoregulation, particularly in larger arteries with low myogenic tone.
Collapse
Affiliation(s)
- Cameron J Morse
- Department Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Erika M Boerman
- Department Medical Physiology and Pharmacology, University of Missouri, Columbia, Missouri
| | - Matthew W McDonald
- Department Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jaume Padilla
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - T Dylan Olver
- Department Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
11
|
Advanced Maternal Age Impairs Uterine Artery Adaptations to Pregnancy in Rats. Int J Mol Sci 2022; 23:ijms23169191. [PMID: 36012456 PMCID: PMC9409016 DOI: 10.3390/ijms23169191] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 11/17/2022] Open
Abstract
Advanced maternal age (≥35 years) is associated with pregnancy complications. Aging impairs vascular reactivity and increases vascular stiffness. We hypothesized that uterine artery adaptations to pregnancy are impaired with advanced age. Uterine arteries of nonpregnant and pregnant (gestational day 20) young (4 months) and aged (9 months; ~35 years in humans) Sprague-Dawley rats were isolated. Functional (myogenic tone, n = 6−10/group) and mechanical (circumferential stress-strain, n = 10−24/group) properties were assessed using pressure myography and further assessment of elastin and collagen (histology, n = 4−6/group), and matrix metalloproteinase-2 (MMP-2, zymography, n = 6/group). Aged dams had worse pregnancy outcomes, including smaller litters and fetal weights (both p < 0.0001). Only in arteries of pregnant young dams did higher pressures (>100 mmHg) cause forced vasodilation. Across the whole pressure range (4−160 mmHg), myogenic behavior was enhanced in aged vs. young pregnant dams (p = 0.0010). Circumferential stress and strain increased with pregnancy in young and aged dams (p < 0.0001), but strain remained lower in aged vs. young dams (p < 0.05). Arteries from young nonpregnant rats had greater collagen:elastin ratios than the other groups (p < 0.05). In aged rats only, pregnancy increased MMP-2 active capacity. Altered functional and structural vascular adaptations to pregnancy may impair fetal growth and development with advanced maternal age.
Collapse
|
12
|
Lansdell TA, Chambers LC, Dorrance AM. Endothelial Cells and the Cerebral Circulation. Compr Physiol 2022; 12:3449-3508. [PMID: 35766836 DOI: 10.1002/cphy.c210015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endothelial cells form the innermost layer of all blood vessels and are the only vascular component that remains throughout all vascular segments. The cerebral vasculature has several unique properties not found in the peripheral circulation; this requires that the cerebral endothelium be considered as a unique entity. Cerebral endothelial cells perform several functions vital for brain health. The cerebral vasculature is responsible for protecting the brain from external threats carried in the blood. The endothelial cells are central to this requirement as they form the basis of the blood-brain barrier. The endothelium also regulates fibrinolysis, thrombosis, platelet activation, vascular permeability, metabolism, catabolism, inflammation, and white cell trafficking. Endothelial cells regulate the changes in vascular structure caused by angiogenesis and artery remodeling. Further, the endothelium contributes to vascular tone, allowing proper perfusion of the brain which has high energy demands and no energy stores. In this article, we discuss the basic anatomy and physiology of the cerebral endothelium. Where appropriate, we discuss the detrimental effects of high blood pressure on the cerebral endothelium and the contribution of cerebrovascular disease endothelial dysfunction and dementia. © 2022 American Physiological Society. Compr Physiol 12:3449-3508, 2022.
Collapse
Affiliation(s)
- Theresa A Lansdell
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Laura C Chambers
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| |
Collapse
|
13
|
Functionally linked potassium channel activity in cerebral endothelial and smooth muscle cells is compromised in Alzheimer's disease. Proc Natl Acad Sci U S A 2022; 119:e2204581119. [PMID: 35727988 PMCID: PMC9245656 DOI: 10.1073/pnas.2204581119] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Patients with Alzheimer’s disease show hypoperfusion of the brain and this may contribute to disease progression. To elucidate underlying mechanisms, we studied pial arteries from 18-mo-old mice with Alzheimer’s disease due to overexpression of amyloid precursor protein. We found enhanced pressure-induced constriction of arteries because of reduction in ryanodine receptor-mediated, local calcium-release events (“Ca2+ sparks”) in arterial smooth muscle cells and a consequent decrease in the activity of large-conductance Ca2+-activated K+ (BK) channels. This phenotype was partially recapitulated by application of an amyloid-β peptide to healthy arteries. Our results will direct further research to restore cerebrovascular function, which is damaged in Alzheimer’s disease, leading to potentially new treatment options. The brain microcirculation is increasingly viewed as a potential target for disease-modifying drugs in the treatment of Alzheimer’s disease patients, reflecting a growing appreciation of evidence that cerebral blood flow is compromised in such patients. However, the pathogenic mechanisms in brain resistance arteries underlying blood flow defects have not yet been elucidated. Here we probed the roles of principal vasodilatory pathways in cerebral arteries using the APP23 mouse model of Alzheimer’s disease, in which amyloid precursor protein is increased approximately sevenfold, leading to neuritic plaques and cerebrovascular accumulation of amyloid-β similar to those in patients with Alzheimer’s disease. Pial arteries from APP23 mice (18 mo old) exhibited enhanced pressure-induced (myogenic) constriction because of a profound reduction in ryanodine receptor-mediated, local calcium-release events (“Ca2+ sparks”) in arterial smooth muscle cells and a consequent decrease in the activity of large-conductance Ca2+-activated K+ (BK) channels. The ability of the endothelial cell inward rectifier K+ (Kir2.1) channel to cause dilation was also compromised. Acute application of amyloid-β 1-40 peptide to cerebral arteries from wild-type mice partially recapitulated the BK dysfunction seen in APP23 mice but had no effect on Kir2.1 function. If mirrored in human Alzheimer’s disease, these tandem defects in K+ channel-mediated vasodilation could account for the clinical cerebrovascular presentation seen in patients: reduced blood flow and crippled functional hyperemia. These data direct future research toward approaches that reverse this dual vascular channel dysfunction, with the ultimate aim of restoring healthy cerebral blood flow and improving clinical outcomes.
Collapse
|
14
|
Eiken O, Keramidas ME, Sköldefors H, Kölegård R. Human cardiovascular adaptation to hypergravity. Am J Physiol Regul Integr Comp Physiol 2022; 322:R597-R608. [PMID: 35470711 DOI: 10.1152/ajpregu.00043.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Despite decades of experience from high-G exposures in aircraft and centrifuges, information is scarce regarding primary cardiovascular adaptations to +Gz loads in relaxed humans. Thus, effects of G-training are typically evaluated after regimens that are confounded by concomitant use of anti-G straining maneuvers, anti-G suits and pressure breathing. Accordingly, the aim was to evaluate cardiovascular adaptations to repeated +Gz exposures in the relaxed state. Eleven men underwent 5 weeks of centrifuge G training, consisting of 15 × 40 min +Gz exposures at G levels close to their individual relaxed G-level tolerance. Before and after the training regimen, relaxed G-level tolerance was investigated during rapid (ROR) and gradual (GOR) onset-rate G exposures, and cardiovascular responses were investigated during orthostatic provocation and vascular pressure-distension tests. The G training resulted in: (i) a 13% increase in relaxed ROR G tolerance (P < 0.001), but no change in GOR G tolerance, (ii) increased pressure resistance in the arteries and arterioles of the legs (P < 0.001), but not the arms, (iii) a reduced initial drop in arterial pressure upon ROR high G, but no change in arterial pressure under basal resting conditions or during GOR G loading, or orthostatic provocation. The results suggest +Gz adaptation via enhanced pressure resistance in dependent arteries/arterioles. Presumably this reflects local adaptations to high transmural pressures, resulting from the +Gz-induced exaggeration of the intravascular hydrostatic pressure gradients.
Collapse
Affiliation(s)
- Ola Eiken
- Division of Environmental Physiology, Swedish Aerospace Physiology Centre, School of Chemistry, Bioengineering and Health, Royal Institute of Technology, KTH, Stockholm, Sweden
| | - Michail E Keramidas
- Division of Environmental Physiology, Swedish Aerospace Physiology Centre, School of Chemistry, Bioengineering and Health, Royal Institute of Technology, KTH, Stockholm, Sweden
| | | | - Roger Kölegård
- Division of Environmental Physiology, Swedish Aerospace Physiology Centre, School of Chemistry, Bioengineering and Health, Royal Institute of Technology, KTH, Stockholm, Sweden
| |
Collapse
|
15
|
Sancho M, Fletcher J, Welsh DG. Inward Rectifier Potassium Channels: Membrane Lipid-Dependent Mechanosensitive Gates in Brain Vascular Cells. Front Cardiovasc Med 2022; 9:869481. [PMID: 35419431 PMCID: PMC8995785 DOI: 10.3389/fcvm.2022.869481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Cerebral arteries contain two primary and interacting cell types, smooth muscle (SMCs) and endothelial cells (ECs), which are each capable of sensing particular hemodynamic forces to set basal tone and brain perfusion. These biomechanical stimuli help confer tone within arterial networks upon which local neurovascular stimuli function. Tone development is intimately tied to arterial membrane potential (VM) and changes in intracellular [Ca2+] driven by voltage-gated Ca2+ channels (VGCCs). Arterial VM is in turn set by the dynamic interplay among ion channel species, the strongly inward rectifying K+ (Kir) channel being of special interest. Kir2 channels possess a unique biophysical signature in that they strongly rectify, display negative slope conductance, respond to elevated extracellular K+ and are blocked by micromolar Ba2+. While functional Kir2 channels are expressed in both smooth muscle and endothelium, they lack classic regulatory control, thus are often viewed as a simple background conductance. Recent literature has provided new insight, with two membrane lipids, phosphatidylinositol 4,5-bisphosphate (PIP2) and cholesterol, noted to (1) stabilize Kir2 channels in a preferred open or closed state, respectively, and (2) confer, in association with the cytoskeleton, caveolin-1 (Cav1) and syntrophin, hemodynamic sensitivity. It is these aspects of vascular Kir2 channels that will be the primary focus of this review.
Collapse
Affiliation(s)
- Maria Sancho
- Department of Pharmacology, University of Vermont, Burlington, VT, United States
- Department of Physiology, Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- *Correspondence: Maria Sancho,
| | - Jacob Fletcher
- Department of Physiology and Pharmacology, Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Donald G. Welsh
- Department of Physiology and Pharmacology, Robarts Research Institute, University of Western Ontario, London, ON, Canada
- Donald G. Welsh,
| |
Collapse
|
16
|
Kuszynski DS, Christian BD, Dorrance AM, Lauver DA. Clopidogrel treatment inhibits P2Y 2-Mediated constriction in the rabbit middle cerebral artery. Eur J Pharmacol 2021; 911:174545. [PMID: 34606835 PMCID: PMC8577565 DOI: 10.1016/j.ejphar.2021.174545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 11/28/2022]
Abstract
Clopidogrel is an effective purinergic 2Y12 receptor (P2Y12) antagonist used to prevent arterial thrombosis, but its use is associated with adverse bleeding. Clinical studies have demonstrated that clopidogrel users have an increased risk of cerebral microbleeds and intracerebral hemorrhage. Our previous studies suggest that non-platelet mechanisms mediate these adverse bleeding events; we hypothesize that clopidogrel or one of its metabolites interacts with blood vessels directly to cause bleeding. New Zealand white rabbits (1.9-2.7 kg) were treated orally with vehicle or clopidogrel (3 or 10 mg/kg) for three days. On the fourth day, the rabbits were anesthetized for blood collection and then euthanized. The brain was collected, and the middle cerebral arteries were isolated. We used light transmission aggregometry and pressure myography to elucidate the mechanisms of the off-target effects associated with clopidogrel treatment. We confirmed that inhibition of P2Y12 activation by clopidogrel inhibited ADP-induced platelet aggregation but had no impact on P2Y12-independent arachidonic acid- or collagen-induced platelet aggregation. Analysis of middle cerebral arteries from clopidogrel treated rabbits showed that clopidogrel did not affect P2Y4, P2Y6, and P2Y14 receptor-mediated contraction but attenuated the contractile response after P2Y2 receptor activation. Further analysis determined P2Y2-mediated constriction was endothelium-dependent. Vasoconstriction is a primary component of hemostasis, and impaired vasoconstriction can prolong bleeding. These results suggest clopidogrel inhibits the endothelial P2Y2 receptor in the middle cerebral artery, which provides a mechanistic explanation for the adverse cerebral bleeding associated with the drug.
Collapse
Affiliation(s)
- Dawn S Kuszynski
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA; Institute of Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - Barbara D Christian
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - D Adam Lauver
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
17
|
Sicotte B, Brochu M. Fetal Sex and Fetal Environment Interact to Alter Diameter, Myogenic Tone, and Contractile Response to Thromboxane Analog in Rat Umbilical Cord Vessels. Front Physiol 2021; 12:620058. [PMID: 34603067 PMCID: PMC8481594 DOI: 10.3389/fphys.2021.620058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 08/25/2021] [Indexed: 11/29/2022] Open
Abstract
Fetal growth needs adequate blood perfusion from both sides of the placenta, on the maternal side through the uterine vessels and on the fetal side through the umbilical cord. In a model of intrauterine growth restriction (IUGR) induced by reduced blood volume expansion, uterine artery remodeling was blunted. The aim of this study is to determine if IUGR and fetus sex alter the functional and mechanical parameters of umbilical cord blood vessels. Pregnant rats were given a low sodium (IUGR) or a control diet for the last 7 days of pregnancy. Umbilical arteries and veins from term (22 day) fetal rats were isolated and set-up in wire myographs. Myogenic tone, diameter, length tension curve and contractile response to thromboxane analog U46619 and serotonin (5-HT) were measured. In arteries from IUGR fetuses, myogenic tone was increased in both sexes while diameter was significantly greater only in male fetuses. In umbilical arteries collected from the control group, the maximal contraction to U46619 was lower in females than males. Compared to the control groups, the maximal response decreased in IUGR male arteries and increased in female ones, thus abolishing the sexual dimorphism observed in the control groups. Reduced contractile response to U46619 was observed in the IUGR vein of both sexes. No difference between groups was observed in response to 5HT in arteries. In conclusion, the change in parameters of the umbilical cord blood vessels in response to a mild insult seems to show adaptation that favors better exchange of deoxygenated and wasted blood from the fetus to the placenta with increased myogenic tone.
Collapse
Affiliation(s)
- Benoit Sicotte
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Michèle Brochu
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
18
|
Carnevale D, Carnevale L, Perrotta S, Pallante F, Migliaccio A, Iodice D, Perrotta M, Lembo G. Chronic 3D Vascular-Immune Interface Established by Coculturing Pressurized Resistance Arteries and Immune Cells. Hypertension 2021; 78:1648-1661. [PMID: 34565186 PMCID: PMC8516815 DOI: 10.1161/hypertensionaha.121.17447] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Daniela Carnevale
- Department of Molecular Medicine, "Sapienza University" of Rome, Italy (D.C., S.P., M.P., G.L.).,Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli (IS), Italy (D.C., L.C., F.P., A.M., D.I., G.L.)
| | - Lorenzo Carnevale
- Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli (IS), Italy (D.C., L.C., F.P., A.M., D.I., G.L.)
| | - Sara Perrotta
- Department of Molecular Medicine, "Sapienza University" of Rome, Italy (D.C., S.P., M.P., G.L.)
| | - Fabio Pallante
- Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli (IS), Italy (D.C., L.C., F.P., A.M., D.I., G.L.)
| | - Agnese Migliaccio
- Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli (IS), Italy (D.C., L.C., F.P., A.M., D.I., G.L.)
| | - Daniele Iodice
- Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli (IS), Italy (D.C., L.C., F.P., A.M., D.I., G.L.)
| | - Marialuisa Perrotta
- Department of Molecular Medicine, "Sapienza University" of Rome, Italy (D.C., S.P., M.P., G.L.)
| | - Giuseppe Lembo
- Department of Molecular Medicine, "Sapienza University" of Rome, Italy (D.C., S.P., M.P., G.L.).,Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli (IS), Italy (D.C., L.C., F.P., A.M., D.I., G.L.)
| |
Collapse
|
19
|
Cadmium-induced hypertension is associated with renal myosin light chain phosphatase inhibition via increased T697 phosphorylation and p44 mitogen-activated protein kinase levels. Hypertens Res 2021; 44:941-954. [PMID: 33972751 DOI: 10.1038/s41440-021-00662-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 12/29/2020] [Accepted: 02/17/2021] [Indexed: 02/03/2023]
Abstract
Dietary intake of the heavy metal cadmium (Cd2+) is implicated in hypertension, but potassium supplementation reportedly mitigates hypertension. This study aims to elucidate the hypertensive mechanism of Cd2+. Vascular reactivity and protein expression were assessed in Cd2+-exposed rats for 8 weeks to determine the calcium-handling effect of Cd2+ and the possible signaling pathways and mechanisms involved. Cd2+ induced hypertension in vivo by significantly (p < 0.001) elevating systolic blood pressure (160 ± 2 and 155 ± 1 vs 120 ± 1 mm Hg), diastolic blood pressure (119 ± 2 and 110 ± 1 vs 81 ± 1 mm Hg), and mean arterial pressure (133 ± 2 and 125 ± 1 vs 94 ± 1 mm Hg) (SBP, DBP, and MAP, respectively), while potassium supplementation protected against elevation of these parameters. The mechanism involved augmentation of the phosphorylation of renal myosin light chain phosphatase targeting subunit 1 (MYPT1) at threonine 697 (T697) (2.58 ± 0.36 vs 1 ± 0) and the expression of p44 mitogen-activated protein kinase (MAPK) (1.78 ± 0.20 vs 1 ± 0). While acetylcholine (ACh)-induced relaxation was unaffected, 5 mg/kg b.w. Cd2+ significantly (p < 0.001) attenuated phenylephrine (Phe)-induced contraction of the aorta, and 2.5 mg/kg b.w. Cd2+ significantly (p < 0.05) augmented sodium nitroprusside (SNP)-induced relaxation of the aorta. These results support the vital role of the kidney in regulating blood pressure changes after Cd2+ exposure, which may be a key drug target for hypertension management. Given the differential response to Cd2+, it is apparent that its hypertensive effects could be mediated by myosin light chain phosphatase (MLCP) inhibition via phosphorylation of renal MYPT1-T697 and p44 MAPK. Further investigation of small arteries and the Rho-kinase/MYPT1 interaction is recommended.
Collapse
|
20
|
Robles FAB, Panerai RB, Katsogridakis E, Chacon M. Superior fitting of arterial resistance and compliance parameters with genetic algorithms in models of dynamic cerebral autoregulation. IEEE Trans Biomed Eng 2021; 69:503-512. [PMID: 34314353 DOI: 10.1109/tbme.2021.3100288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE The capacity of discriminating between normal and impaired dynamic cerebral autoregulation (dCA), based on spontaneous fluctuations in arterial blood pressure (ABP) and cerebral blood flow (CBF), has considerable clinical relevance. This study aimed to quantify the separate contributions of vascular resistance and compliance as parameters that could reflect myogenic and metabolic mechanisms to dCA. METHODS Forty-five subjects were studied under normo and hypercapnic conditions induced by breathing a mixture of 5% carbon dioxide in air. Dynamic cerebrovascular resistance and compliance models with ABP as input and CBFV as output were fitted using Genetic Algorithms to identify parameter values for each subject, and respiratory condition. RESULTS The efficiency of dCA was assessed from the models generated CBFV response to an ABP step change, corresponding to an autoregulation index of 5.561.57 in normocapnia and 2.381.73 in hypercapnia, with an area under the ROC curve (AUC) of 0.9 between both conditions. Vascular compliance increased from 0.750.7 ml/mmHg in normocapnia to 5.8212.0 ml/mmHg during hypercapnia, with an AUC of 0.88. CONCLUSION we demonstrated that Genetic Algorithms are a powerful tool to provide accurate identification of model parameters expressing the performance of human CA Significance: Further work is needed to validate this approach in clinical applications where individualised model parameters could provide relevant diagnostic and prognostic information about dCA impairment Index Terms arterial compliance, autoregulation impairment, cerebral blood flow, Genetic Algorithms, hypercapnia.
Collapse
|
21
|
Lidington D, Wan H, Bolz SS. Cerebral Autoregulation in Subarachnoid Hemorrhage. Front Neurol 2021; 12:688362. [PMID: 34367053 PMCID: PMC8342764 DOI: 10.3389/fneur.2021.688362] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/25/2021] [Indexed: 12/28/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is a devastating stroke subtype with a high rate of mortality and morbidity. The poor clinical outcome can be attributed to the biphasic course of the disease: even if the patient survives the initial bleeding emergency, delayed cerebral ischemia (DCI) frequently follows within 2 weeks time and levies additional serious brain injury. Current therapeutic interventions do not specifically target the microvascular dysfunction underlying the ischemic event and as a consequence, provide only modest improvement in clinical outcome. SAH perturbs an extensive number of microvascular processes, including the “automated” control of cerebral perfusion, termed “cerebral autoregulation.” Recent evidence suggests that disrupted cerebral autoregulation is an important aspect of SAH-induced brain injury. This review presents the key clinical aspects of cerebral autoregulation and its disruption in SAH: it provides a mechanistic overview of cerebral autoregulation, describes current clinical methods for measuring autoregulation in SAH patients and reviews current and emerging therapeutic options for SAH patients. Recent advancements should fuel optimism that microvascular dysfunction and cerebral autoregulation can be rectified in SAH patients.
Collapse
Affiliation(s)
- Darcy Lidington
- Department of Physiology, University of Toronto, Toronto, ON, Canada.,Toronto Centre for Microvascular Medicine at the Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Toronto, ON, Canada
| | - Hoyee Wan
- Department of Physiology, University of Toronto, Toronto, ON, Canada.,Toronto Centre for Microvascular Medicine at the Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Toronto, ON, Canada
| | - Steffen-Sebastian Bolz
- Department of Physiology, University of Toronto, Toronto, ON, Canada.,Toronto Centre for Microvascular Medicine at the Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Toronto, ON, Canada.,Heart & Stroke/Richard Lewar Centre of Excellence for Cardiovascular Research, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
22
|
Szenasi A, Amrein K, Czeiter E, Szarka N, Toth P, Koller A. Molecular Pathomechanisms of Impaired Flow-Induced Constriction of Cerebral Arteries Following Traumatic Brain Injury: A Potential Impact on Cerebral Autoregulation. Int J Mol Sci 2021; 22:6624. [PMID: 34205652 PMCID: PMC8234931 DOI: 10.3390/ijms22126624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 01/21/2023] Open
Abstract
(1) Background: Traumatic brain injury (TBI) frequently occurs worldwide, resulting in high morbidity and mortality. Here, we hypothesized that TBI impairs an autoregulatory mechanism, namely the flow-induced constriction of isolated rat middle cerebral arteries (MCAs). (2) Methods: TBI was induced in anaesthetized rats by weight drop model, and then MCAs were isolated and transferred into a pressure-flow chamber. The internal diameter was measured by a video-microscopy. (3) Results: In MCAs from intact rats, increases in flow and pressure + flow elicited constrictions (-26 ± 1.9 µm and -52 ± 2.8 µm, p < 0.05), which were significantly reduced after TBI or in the presence of thromboxane-prostanoid (TP receptor) antagonist SQ 29,548. Flow-induced constrictions were significantly reduced by HET0016, inhibitor of cytochrome P450 4A (CYP450 4A). Arachidonic acid, (AA, 10-7 M), and CYP-450 4A metabolite 20-hydroxyeicosatetraenoic acid (20-HETE) elicited constrictions of intact MCA (-26 ± 2.3% and -31 ± 3.6%), which were significantly reduced after TBI (to 11 ± 1.3% and -16 ±2.5%). The TP receptor agonist U46619 (10-7 M) elicited substantial constrictions of MCA from intact rats (-21 ± 3.3%), which were also significantly reduced, after TBI (to -16 ± 2.4%). (4) Conclusions: Flow-induced constrictor response of MCA is impaired by traumatic brain injury, likely due to the reduced ability of cytochrome P450 4A to convert arachidonic acid to constrictor prostaglandins and the mitigated sensitivity of thromboxane-prostanoid receptors.
Collapse
Affiliation(s)
- Annamaria Szenasi
- Department of Translational Medicine, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary;
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 1085 Budapest, Hungary
| | - Krisztina Amrein
- Department of Neurosurgery and Szentágothai Research Center, University of Pecs, Medical School, 7623 Pecs, Hungary; (K.A.); (E.C.); (P.T.)
| | - Endre Czeiter
- Department of Neurosurgery and Szentágothai Research Center, University of Pecs, Medical School, 7623 Pecs, Hungary; (K.A.); (E.C.); (P.T.)
| | - Nikolett Szarka
- MTA-PTE Clinical Neuroscience MR Research Group, 7623 Pecs, Hungary;
| | - Peter Toth
- Department of Neurosurgery and Szentágothai Research Center, University of Pecs, Medical School, 7623 Pecs, Hungary; (K.A.); (E.C.); (P.T.)
- MTA-PTE Clinical Neuroscience MR Research Group, 7623 Pecs, Hungary;
| | - Akos Koller
- Department of Translational Medicine, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary;
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 1085 Budapest, Hungary
- Research Center for Sports Physiology, University of Physical Education, 1123 Budapest, Hungary
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|
23
|
Koide M, Ferris HR, Nelson MT, Wellman GC. Impaired Cerebral Autoregulation After Subarachnoid Hemorrhage: A Quantitative Assessment Using a Mouse Model. Front Physiol 2021; 12:688468. [PMID: 34168571 PMCID: PMC8218876 DOI: 10.3389/fphys.2021.688468] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/26/2021] [Indexed: 01/01/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) is a common form of hemorrhagic stroke associated with high rates of mortality and severe disability. SAH patients often develop severe neurological deficits days after ictus, events attributed to a phenomenon referred to as delayed cerebral ischemia (DCI). Recent studies indicate that SAH-induced DCI results from a multitude of cerebral circulatory disturbances including cerebral autoregulation malfunction. Cerebral autoregulation incorporates the influence of blood pressure (BP) on arterial diameter in the homeostatic regulation of cerebral blood flow (CBF), which is necessary for maintaining constant brain perfusion during physiological swings in systemic BP. In this study, we quantitatively examined the impact of SAH on cerebral autoregulation using a mouse endovascular perforation model and a newly developed approach combining absolute and relative CBF measurements. This method enables a direct quantitative comparison of cerebral autoregulation between individual animals (e.g., SAH vs. control or sham-operated mice), which cannot be done solely using relative CBF changes by laser Doppler flowmetry. Here, absolute CBF was measured via injection of fluorescent microspheres at a baseline BP. In separate groups of animals, in vivo laser Doppler flowmetry was used to measure relative CBF changes over a range of BP using phlebotomy and the pressor phenylephrine to lower and raise BP, respectively. Absolute CBF measurements from microspheres were then used to calibrate laser Doppler measurements to calculate the relationship between CBF and BP, i.e., “cerebral autoregulation curves.” Un-operated and sham-operated groups exhibited similar cerebral autoregulatory curves, showing comparable levels of relatively constant CBF over a range of BP from ~80 mmHg to ~130 mmHg. In contrast, SAH animals exhibited a narrower autoregulatory range of BP, which was primarily due to a decrease in the upper limit of BP whereby cerebral autoregulation was maintained. Importantly, SAH animals also exhibited a marked decrease in CBF throughout the entire range of BP. In sum, this study provides evidence of the dramatic reduction in cortical CBF and the diminished range of autoregulation after SAH. Furthermore, this novel methodology should pave the way for future studies examining pathological mechanisms and/or therapeutic strategies targeting impaired cerebral autoregulation, a pathology common to many cardiovascular and cerebrovascular disorders.
Collapse
Affiliation(s)
- Masayo Koide
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, United States.,Vermont Center for Cardiovascular and Brain Health, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Hannah R Ferris
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Mark T Nelson
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, United States.,Vermont Center for Cardiovascular and Brain Health, Larner College of Medicine, University of Vermont, Burlington, VT, United States.,Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - George C Wellman
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| |
Collapse
|
24
|
Wenceslau CF, McCarthy CG, Earley S, England SK, Filosa JA, Goulopoulou S, Gutterman DD, Isakson BE, Kanagy NL, Martinez-Lemus LA, Sonkusare SK, Thakore P, Trask AJ, Watts SW, Webb RC. Guidelines for the measurement of vascular function and structure in isolated arteries and veins. Am J Physiol Heart Circ Physiol 2021; 321:H77-H111. [PMID: 33989082 DOI: 10.1152/ajpheart.01021.2020] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The measurement of vascular function in isolated vessels has revealed important insights into the structural, functional, and biomechanical features of the normal and diseased cardiovascular system and has provided a molecular understanding of the cells that constitutes arteries and veins and their interaction. Further, this approach has allowed the discovery of vital pharmacological treatments for cardiovascular diseases. However, the expansion of the vascular physiology field has also brought new concerns over scientific rigor and reproducibility. Therefore, it is appropriate to set guidelines for the best practices of evaluating vascular function in isolated vessels. These guidelines are a comprehensive document detailing the best practices and pitfalls for the assessment of function in large and small arteries and veins. Herein, we bring together experts in the field of vascular physiology with the purpose of developing guidelines for evaluating ex vivo vascular function. By using this document, vascular physiologists will have consistency among methodological approaches, producing more reliable and reproducible results.
Collapse
Affiliation(s)
- Camilla F Wenceslau
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Cameron G McCarthy
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Scott Earley
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, Reno School of Medicine, University of Nevada, Reno, Nevada
| | - Sarah K England
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri
| | - Jessica A Filosa
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Styliani Goulopoulou
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - David D Gutterman
- Department of Medicine, Medical College of Wisconsin Cardiovascular Center, Milwaukee, Wisconsin
| | - Brant E Isakson
- Department of Molecular Physiology and Biophysics, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Nancy L Kanagy
- Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico
| | - Luis A Martinez-Lemus
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Swapnil K Sonkusare
- Department of Molecular Physiology and Biophysics, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Pratish Thakore
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, Reno School of Medicine, University of Nevada, Reno, Nevada
| | - Aaron J Trask
- Center for Cardiovascular Research, The Heart Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - R Clinton Webb
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
25
|
Eiken O, Elia A, Sköldefors H, Sundblad P, Keramidas ME, Kölegård R. Adaptation to 5 weeks of intermittent local vascular pressure increments; mechanisms to be considered in the development of primary hypertension? Am J Physiol Heart Circ Physiol 2021; 320:H1303-H1312. [PMID: 33481701 DOI: 10.1152/ajpheart.00763.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The aims were to study effects of iterative exposures to moderate elevations of local intravascular pressure on arterial/arteriolar stiffness and plasma levels of vasoactive substances. Pressures in the vasculature of an arm were increased by 150 mmHg in healthy men (n = 11) before and after a 5-wk regimen, during which the vasculature in one arm was exposed to fifteen 40-min sessions of moderately increased transmural pressure (+65 to +105 mmHg). This vascular pressure training and the pressure-distension determinations were conducted by exposing the subjects' arm versus remaining part of the body to differential ambient pressure. During the pressure-distension determinations, venous samples were simultaneously obtained from pressurized and unpressurized vessels. Pressure training reduced arterial pressure distension by 40 ± 23% and pressure-induced flow by 33 ± 30% (P < 0.01), but only in the pressure-trained arm, suggesting local adaptive mechanisms. The distending pressure-diameter and distending pressure-flow curves, with training-induced increments in pressure thresholds and reductions in response gains, suggest that the increased precapillary stiffness was attributable to increased contractility and structural remodeling of the walls. Acute vascular pressure provocation induced local release of angiotensin-II (ANG II) and endothelin-1 (ET-1) (P < 0.05), suggesting that these vasoconstrictors limited the pressure distension. Pressure training increased basal levels of ET-1 and induced local pressure release of matrix metalloproteinase 7 (P < 0.05), suggesting involvement of these substances in vascular remodeling. The findings are compatible with the notion that local intravascular pressure load acts as a prime mover in the development of primary hypertension.NEW & NOTEWORTHY Adaptive responses to arterial/arteriolar pressure elevation have typically been investigated in cross-sectional studies in hypertensive patients or in longitudinal studies in experimental animals. The present investigation shows that in healthy individuals, fifteen 40-min, carefully controlled, moderate transmural pressure elevations markedly increase in vivo stiffness (i.e. reduce pressure distension) in arteries and arterioles. The response is mediated via local mechanisms, and it appears that endothelin-1, angiotensin-II, and matrix metalloproteinase 7 may have key roles.
Collapse
Affiliation(s)
- O Eiken
- Division of Environmental Physiology, Swedish Aerospace Physiology Center, KTH Royal Institute of Technology, Stockholm, Sweden
| | - A Elia
- Division of Environmental Physiology, Swedish Aerospace Physiology Center, KTH Royal Institute of Technology, Stockholm, Sweden
| | - H Sköldefors
- Division of Environmental Physiology, Swedish Aerospace Physiology Center, KTH Royal Institute of Technology, Stockholm, Sweden
| | - P Sundblad
- Division of Environmental Physiology, Swedish Aerospace Physiology Center, KTH Royal Institute of Technology, Stockholm, Sweden
| | - M E Keramidas
- Division of Environmental Physiology, Swedish Aerospace Physiology Center, KTH Royal Institute of Technology, Stockholm, Sweden
| | - R Kölegård
- Division of Environmental Physiology, Swedish Aerospace Physiology Center, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
26
|
Shadrina NK. A Mathematical Model of the Wall of a Rat Cerebral Resistance Vessel. Biophysics (Nagoya-shi) 2021. [DOI: 10.1134/s0006350921010188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
27
|
Hansen AB, Moralez G, Romero SA, Gasho C, Tymko MM, Ainslie PN, Hofstätter F, Rainer SL, Lawley JS, Hearon CM. Mechanisms of sympathetic restraint in human skeletal muscle during exercise: role of α-adrenergic and nonadrenergic mechanisms. Am J Physiol Heart Circ Physiol 2020; 319:H192-H202. [PMID: 32502375 DOI: 10.1152/ajpheart.00208.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sympathetic vasoconstriction is mediated by α-adrenergic receptors under resting conditions. During exercise, increased sympathetic nerve activity (SNA) is directed to inactive and active skeletal muscle; however, it is unclear what mechanism(s) are responsible for vasoconstriction during large muscle mass exercise in humans. The aim of this study was to determine the contribution of α-adrenergic receptors to sympathetic restraint of inactive skeletal muscle and active skeletal muscle during cycle exercise in healthy humans. In ten male participants (18-35 yr), mean arterial pressure (intra-arterial catheter) and forearm vascular resistance (FVR) and conductance (FVC) were assessed during cycle exercise (60% total peak workload) alone and during combined cycle exercise + handgrip exercise (HGE) before and after intra-arterial blockade of α- and β-adrenoreceptors via phentolamine and propranolol, respectively. Cycle exercise caused vasoconstriction in the inactive forearm that was attenuated ~80% with adrenoreceptor blockade (%ΔFVR, +81.7 ± 84.6 vs. +9.7 ± 30.7%; P = 0.05). When HGE was performed during cycle exercise, the vasodilatory response to HGE was restrained by ~40% (ΔFVC HGE, +139.3 ± 67.0 vs. cycle exercise: +81.9 ± 66.3 ml·min-1·100 mmHg-1; P = 0.03); however, the restraint of active skeletal muscle blood flow was not due to α-adrenergic signaling. These findings highlight that α-adrenergic receptors are the primary, but not the exclusive mechanism by which sympathetic vasoconstriction occurs in inactive and active skeletal muscle during exercise. Metabolic activity or higher sympathetic firing frequencies may alter the contribution of α-adrenergic receptors to sympathetic vasoconstriction. Finally, nonadrenergic vasoconstrictor mechanisms may be important for understanding the regulation of blood flow during exercise.NEW & NOTEWORTHY Sympathetic restraint of vascular conductance to inactive skeletal muscle is critical to maintain blood pressure during moderate- to high-intensity whole body exercise. This investigation shows that cycle exercise-induced restraint of inactive skeletal muscle vascular conductance occurs primarily because of activation of α-adrenergic receptors. Furthermore, exercise-induced vasoconstriction restrains the subsequent vasodilatory response to hand-grip exercise; however, the restraint of active skeletal muscle vasodilation was in part due to nonadrenergic mechanisms. We conclude that α-adrenergic receptors are the primary but not exclusive mechanism by which sympathetic vasoconstriction restrains blood flow in humans during whole body exercise and that metabolic activity modulates the contribution of α-adrenergic receptors.
Collapse
Affiliation(s)
- Alexander B Hansen
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Gilbert Moralez
- Department of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Steven A Romero
- University of North Texas Health Science Center, Fort Worth, Texas
| | - Christopher Gasho
- Division of Pulmonary and Critical Care, Department of Medicine, University of Loma Lida, Loma Lida, California
| | - Michael M Tymko
- Centre of Heart, Lung, and Vascular Health, School of Health and Exercise Sciences, University of British Columbia-Okanagan, Kelowna, British Columbia, Canada.,Physical Activity and Diabetes Laboratory, Faculty of Kinesiology, Sport and Recreation, University of Alberta, Edmonton, Alberta, Canada
| | - Philip N Ainslie
- Centre of Heart, Lung, and Vascular Health, School of Health and Exercise Sciences, University of British Columbia-Okanagan, Kelowna, British Columbia, Canada
| | - Florian Hofstätter
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Simon L Rainer
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Justin S Lawley
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Christopher M Hearon
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Dallas, Dallas, Texas.,University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
28
|
Kim JT, Won SY, Kang K, Kim SH, Park MS, Choi KH, Nam TS, Denis SW, Ferdinandusse S, Lee JE, Choi SY, Kim MK. ACOX3 Dysfunction as a Potential Cause of Recurrent Spontaneous Vasospasm of Internal Carotid Artery. Transl Stroke Res 2020; 11:1041-1051. [PMID: 31975215 DOI: 10.1007/s12975-020-00779-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/30/2019] [Accepted: 01/02/2020] [Indexed: 11/24/2022]
Abstract
Recurrent spontaneous vasospasm of the extracranial internal carotid artery (RSV-eICA) is a rarely recognized cause of ischemic stroke in young adults. However, its pathophysiology remains largely unknown. Through whole-exome sequencing of the ACOX3 gene of two dizygotic Korean twin brothers affected by RSV-eICA, we identified two compound heterozygous missense variants c.235 T > G (p.F79 V) and c.665G > A (p.G222E). In silico analysis indicated that both variants were classified as pathogenic. In vitro ACOX3 enzyme assay indicated practically no enzyme activity in both F79 V and G222E mutants. To determine the effect of the mutants on vasospasm, we used a collagen contraction assay on human aortic smooth muscle cells (HASMC). Carbachol, a cholinergic agonist, induces contraction of HASMC. Knockdown of ACOX3 in HASMC, using siRNA, significantly repressed HASMC contraction triggered by carbachol. The carbachol-induced HASMC contraction was restored by transfection with plasmids encoding siRNA-resistant wild-type ACOX3, but not by transfection with ACOX3 G222E or by co-transfection with ACOX3 F79 V and ACOX3 G222E, indicating that the two ACOX3 mutants suppress carbachol-induced HASMC contraction. We propose that an ACOX3 dysfunction elicits a prolonged loss of the basal aortic myogenic tone. As a result, smooth muscles of the ICA's intermediate segment, in which the sympathetic innervation is especially rich, becomes hypersensitive to sympathomimetic stimuli (e.g., heavy exercise) leading to a recurrent vasospasm. Therefore, ACOX3 dysfunction would be a causal mechanism of RSV-eICA. For the first time, we report the possible involvement of ACOX3 in maintaining the basal myogenic tone of human arterial smooth muscle.
Collapse
Affiliation(s)
- Joon-Tae Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - So Yeon Won
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, South Korea
| | - KyungWook Kang
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - Sang-Hoon Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - Man-Seok Park
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - Kang-Ho Choi
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - Tai-Seung Nam
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - Simone W Denis
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| | - Sacha Ferdinandusse
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| | - Ji Eun Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, South Korea.
| | - Seok-Yong Choi
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju, 61469, South Korea.
| | - Myeong-Kyu Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea.
| |
Collapse
|
29
|
Venkatesh I, Shubha HV, Karthik N, RaviShankar S. A Study of reversal of diastolic blood flow in the middle cerebral artery using doppler ultrasound in the prognostication in sick neonates. J Clin Neonatol 2020. [DOI: 10.4103/jcn.jcn_83_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
30
|
Lyubashina OA, Mamontov OV, Volynsky MA, Zaytsev VV, Kamshilin AA. Contactless Assessment of Cerebral Autoregulation by Photoplethysmographic Imaging at Green Illumination. Front Neurosci 2019; 13:1235. [PMID: 31798408 PMCID: PMC6863769 DOI: 10.3389/fnins.2019.01235] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 10/31/2019] [Indexed: 12/15/2022] Open
Abstract
Accurate and practical assessment of the brain circulation is needed to adequately estimate the viability of cerebral blood flow regulatory mechanisms in various physiological conditions. The objective of our study was to examine feasibility of the contactless green-light imaging photoplethysmography (PPG) for assessing cerebral autoregulation by revealing the dynamic relationships between cortical microcirculation assessed by PPG and changes in systemic blood pressure caused by visceral and somatic peripheral stimuli. In anesthetized male Wistar rats, the PPG video images of the open parietal cortex (either with unimpaired or dissected dura mater), electrocardiogram, and systemic arterial blood pressure (ABP) in the femoral artery were continuously recorded before, during and after visceral (colorectal distension) or somatic (tail squeezing) stimulation. In the vast majority of experiments with intact and removed dura mater, both spontaneous and peripheral stimulation-evoked changes in ABP negatively correlated with the accompanying alterations in the amplitude of pulsatile PPG component (APC), i.e., an increase of ABP resulted in a decrease of APC and vice versa. The most pronounced ABP and APC alterations were induced by noxious stimuli. Visceral painful stimulation in all cases caused short-term hypotension with simultaneous increase in cortical APC, whereas somatic noxious stimuli in 8 of 21 trials produced hypertensive effect with decreased APC. Animals with pressure 50-70 mmHg possessed higher negative cerebrovascular response rate of ABP-APC gradients than rats with either lower or higher pressure. Severe hypotension reversed the negative ratio to positive one, which was especially evident under visceral pain stimulation. Amplitude of the pulsatile PPG component probably reflects the regulation of vascular tone of cerebral cortex in response to systemic blood pressure fluctuations. When combined with different kinds of peripheral stimuli, the technique is capable for evaluation of normal and elucidation of impaired cerebrovascular system reactivity to particular physiological events, for example pain. The reported contactless PPG monitoring of cortical circulatory dynamics during neurosurgical interventions in combination with recordings of changes in other physiological parameters, such as systemic blood pressure and ECG, has the appealing potential to monitor viability of the cortex vessels and determine the state of patient’s cerebrovascular autoregulation.
Collapse
Affiliation(s)
- Olga A Lyubashina
- Laboratory of Cortico-Visceral Physiology, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, Russia.,Valdman Institute of Pharmacology, Pavlov First Saint Petersburg State Medical University, Saint Petersburg, Russia
| | - Oleg V Mamontov
- Department of Circulation Physiology, Almazov National Medical Research Centre, Saint Petersburg, Russia.,Faculty of Applied Optics, ITMO University, Saint Petersburg, Russia
| | - Maxim A Volynsky
- Faculty of Applied Optics, ITMO University, Saint Petersburg, Russia
| | - Valeriy V Zaytsev
- Faculty of Applied Optics, ITMO University, Saint Petersburg, Russia
| | | |
Collapse
|
31
|
Cerebral Blood Flow Regulation in Pregnancy, Hypertension, and Hypertensive Disorders of Pregnancy. Brain Sci 2019; 9:brainsci9090224. [PMID: 31487961 PMCID: PMC6769869 DOI: 10.3390/brainsci9090224] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/26/2019] [Accepted: 09/03/2019] [Indexed: 01/12/2023] Open
Abstract
The regulation of cerebral blood flow (CBF) allows for the metabolic demands of the brain to be met and for normal brain function including cognition (learning and memory). Regulation of CBF ensures relatively constant blood flow to the brain despite changes in systemic blood pressure, protecting the fragile micro-vessels from damage. CBF regulation is altered in pregnancy and is further altered by hypertension and hypertensive disorders of pregnancy including preeclampsia. The mechanisms contributing to changes in CBF in normal pregnancy, hypertension, and preeclampsia have not been fully elucidated. This review summarizes what is known about changes in CBF regulation during pregnancy, hypertension, and preeclampsia.
Collapse
|
32
|
Haines L, Villalba N, Sackheim AM, Collier DM, Freeman K. Myogenic tone contributes to the regulation of permeability in mesenteric microvessels. Microvasc Res 2019; 125:103873. [PMID: 30974113 DOI: 10.1016/j.mvr.2019.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 04/03/2019] [Accepted: 04/07/2019] [Indexed: 11/30/2022]
Abstract
The microvascular endothelium plays a key role in regulating solute permeability in the gut, but the contribution of vascular smooth muscle to barrier function is unknown. We sought to determine the role of vascular smooth muscle and its myogenic tone in the vascular barrier to solutes in mesenteric microvessels. We determined vascular permeability to 4.4 kDa and 70 kDa dextrans in isolated mouse mesenteric arteries at increasing pressure increments. The myogenic response was simultaneously monitored using video edge-detection of vessel diameter and wall thickness. We expressed permeability as the apparent permeability coefficient, or the solute flux per second normalized to surface area and concentration gradient. We compared the effects of myogenic tone, L-type calcium channel blockade, calcium elimination, and endothelial removal on the permeability of each dextran. We found arteries resisted changes in 4.4 kDa and 70 kDa dextran permeability coefficients at intravascular pressures associated with myogenic tone. Manipulations that reduced or eliminated myogenic tone (L-type calcium channel blockade or calcium elimination) caused vasodilation and increased permeability coefficients. Thus, the maintenance of a reactive mesenteric vascular smooth muscle layer and its myogenic tone prevents increases in vascular permeability that would otherwise occur with increasing pressure. Conditions that impact vascular tone, such as trauma, stroke, or major surgery could diminish the gut-vascular barrier against dissemination of the microbiome.
Collapse
Affiliation(s)
- Laurel Haines
- Department of Surgery, University of Vermont, Given Medical Building E301, 89 Beaumont Ave, Burlington, VT 05405, United States
| | - Nuria Villalba
- Department of Surgery, University of Vermont, Given Medical Building E301, 89 Beaumont Ave, Burlington, VT 05405, United States
| | - Adrian M Sackheim
- Department of Surgery, University of Vermont, Given Medical Building E301, 89 Beaumont Ave, Burlington, VT 05405, United States
| | - Daniel M Collier
- Department of Pharmacology, University of Vermont, Given Medical Building E301, 89 Beaumont Ave, Burlington, VT 05405, United States
| | - Kalev Freeman
- Department of Surgery, University of Vermont, Given Medical Building E301, 89 Beaumont Ave, Burlington, VT 05405, United States; Department of Pharmacology, University of Vermont, Given Medical Building E301, 89 Beaumont Ave, Burlington, VT 05405, United States.
| |
Collapse
|
33
|
Chan SL, Nelson MT, Cipolla MJ. Transient receptor potential vanilloid-4 channels are involved in diminished myogenic tone in brain parenchymal arterioles in response to chronic hypoperfusion in mice. Acta Physiol (Oxf) 2019; 225:e13181. [PMID: 30153398 DOI: 10.1111/apha.13181] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 08/22/2018] [Accepted: 08/24/2018] [Indexed: 12/11/2022]
Abstract
AIM Adaptive responses of brain parenchymal arterioles (PAs), a target for cerebral small vessel disease, to chronic cerebral hypoperfusion are largely unknown. Previous evidence suggested that transient receptor potential vanilloid 4 channels may be involved in the regulation of cerebrovascular tone. Therefore, we investigated the role of TRPV4 in adaptations of PAs in a mouse model of chronic hypoperfusion. METHODS TRPV4 knockout (-/- ) and wild-type (WT) mice were subjected to unilateral common carotid artery occlusion (UCCAo) for 28 days. Function and structure of PAs ipsilateral to UCCAo were studied isolated and pressurized in an arteriograph. RESULTS Basal tone of PAs was similar between WT and TRPV4-/- mice (22 ± 3 vs 23 ± 5%). After UCCAo, active inner diameters of PAs from WT mice were larger than control (41 ± 2 vs 26 ± 5 μm, P < 0.05) that was due to decreased tone (8 ± 2 vs 23 ± 5%, P < 0.05), increased passive inner diameters (46 ± 3 vs 34 ± 2 μm, P < 0.05), and decreased wall-to-lumen ratio (0.104 ± 0.01 vs 0.137 ± 0.01, P < 0.05). However, UCCAo did not affect vasodilation to a small- and intermediate-conductance calcium-activated potassium channel agonist NS309, the nitric oxide (NO) donor sodium nitroprusside, or constriction to a NO synthase inhibitor L-NNA. Wall thickness and distensibility in PAs from WT mice were unaffected. In TRPV4-/- mice, UCCAo had no effect on active inner diameters or tone and only increased passive inner diameters (53 ± 2 vs 43 ± 3 μm, P < 0.05). CONCLUSION Adaptive response of PAs to chronic cerebral hypoperfusion includes myogenic tone reduction and outward remodelling. TRPV4 channels were involved in tone reduction but not outward remodelling in response to UCCAo.
Collapse
Affiliation(s)
- Siu-Lung Chan
- Department of Neurological Sciences; University of Vermont College of Medicine; Burlington Vermont
| | - Mark T. Nelson
- Department of Pharmacology; University of Vermont College of Medicine; Burlington Vermont
| | - Marilyn J. Cipolla
- Department of Neurological Sciences; University of Vermont College of Medicine; Burlington Vermont
- Department of Pharmacology; University of Vermont College of Medicine; Burlington Vermont
- Department of Obstetrics, Gynecology & Reproductive Sciences; University of Vermont College of Medicine; Burlington Vermont
| |
Collapse
|
34
|
Migrino RQ, Truran S, Karamanova N, Serrano GE, Madrigal C, Davies HA, Madine J, Reaven P, Beach TG. Human cerebral collateral arteriole function in subjects with normal cognition, mild cognitive impairment, and dementia. Am J Physiol Heart Circ Physiol 2018; 315:H284-H290. [PMID: 29775413 PMCID: PMC6139628 DOI: 10.1152/ajpheart.00206.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/03/2018] [Accepted: 05/14/2018] [Indexed: 01/06/2023]
Abstract
Clinical and preclinical studies have suggested a link between cardiovascular disease and dementia disorders, but the role of the collateral brain circulation in cognitive dysfunction remains unknown. We aimed to test the hypothesis that leptomeningeal arteriole (LMA) function and response to metabolic stressors differ among subjects with dementia, mild cognitive impairment (MCI), and normal cognition (CN). After rapid autopsy, LMAs were isolated from subjects with CN ( n = 10), MCI ( n = 12), or dementia [ n = 42, Alzheimer's disease (AD), vascular dementia (VaD), or other dementia], and endothelial and smooth muscle-dependent function were measured at baseline and after exposure to β-amyloid (2 μM), palmitic acid (150 μM), or medin (5 μM) and compared. There were no differences among the groups in baseline endothelial function (maximum dilation to acetylcholine, CN: 74.1 ± 9.7%, MCI: 67.1 ± 4.8%, AD: 74.7 ± 2.8%, VaD: 72.0 ± 5.3%, and other dementia: 68.0 ± 8.0%) and smooth muscle-dependent function (CN: 93.4 ± 3.0%, MCI: 83.3 ± 4.1%, AD: 91.8 ± 1.7%, VaD: 91.7 ± 2.4%, and other dementia: 87.9 ± 4.9%). There was no correlation between last cognitive function score and baseline endothelial or smooth muscle-dependent function. LMA endothelial function and, to a lesser extent, smooth muscle-dependent function were impaired posttreatment with β-amyloid, palmitic acid, and medin. Posttreatment LMA responses were not different between subjects with CN/MCI vs. dementia. Baseline responses and impaired vasoreactivity after treatment with metabolic stressors did not differ among subjects with CN, MCI, and dementia. The results suggest that the cognitive dysfunction in dementia disorders is not attributable to differences in baseline brain collateral circulation function but may be influenced by exposure of the vasculature to metabolic stressors.
Collapse
Affiliation(s)
- Raymond Q Migrino
- Office of Research, Phoenix Veterans Affairs Health Care System, Phoenix, Arizona
- Department of Medicine, University of Arizona College of Medicine-Phoenix , Phoenix, Arizona
| | - Seth Truran
- Office of Research, Phoenix Veterans Affairs Health Care System, Phoenix, Arizona
| | - Nina Karamanova
- Office of Research, Phoenix Veterans Affairs Health Care System, Phoenix, Arizona
| | - Geidy E Serrano
- Department of Neuropathology, Banner-Sun Health Research Institute , Sun City, Arizona
| | - Calvin Madrigal
- Office of Research, Phoenix Veterans Affairs Health Care System, Phoenix, Arizona
| | - Hannah A Davies
- Department of Biochemistry, University of Liverpool , Liverpool , United Kingdom
| | - Jillian Madine
- Department of Biochemistry, University of Liverpool , Liverpool , United Kingdom
| | - Peter Reaven
- Office of Research, Phoenix Veterans Affairs Health Care System, Phoenix, Arizona
- Department of Medicine, University of Arizona College of Medicine-Phoenix , Phoenix, Arizona
| | - Thomas G Beach
- Department of Neuropathology, Banner-Sun Health Research Institute , Sun City, Arizona
| |
Collapse
|
35
|
Eid AH, El-Yazbi AF, Zouein F, Arredouani A, Ouhtit A, Rahman MM, Zayed H, Pintus G, Abou-Saleh H. Inositol 1,4,5-Trisphosphate Receptors in Hypertension. Front Physiol 2018; 9:1018. [PMID: 30093868 PMCID: PMC6071574 DOI: 10.3389/fphys.2018.01018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 07/09/2018] [Indexed: 12/21/2022] Open
Abstract
Chronic hypertension remains a major cause of global mortality and morbidity. It is a complex disease that is the clinical manifestation of multiple genetic, environmental, nutritional, hormonal, and aging-related disorders. Evidence supports a role for vascular aging in the development of hypertension involving an impairment in endothelial function together with an alteration in vascular smooth muscle cells (VSMCs) calcium homeostasis leading to increased myogenic tone. Changes in free intracellular calcium levels ([Ca2+] i ) are mediated either by the influx of Ca2+ from the extracellular space or release of Ca2+ from intracellular stores, mainly the sarcoplasmic reticulum (SR). The influx of extracellular Ca2+ occurs primarily through voltage-gated Ca2+ channels (VGCCs), store-operated Ca2+ channels (SOC), and Ca2+ release-activated channels (CRAC), whereas SR-Ca2+ release occurs through inositol trisphosphate receptor (IP3R) and ryanodine receptors (RyRs). IP3R-mediated SR-Ca2+ release, in the form of Ca2+ waves, not only contributes to VSMC contraction and regulates VGCC function but is also intimately involved in structural remodeling of resistance arteries in hypertension. This involves a phenotypic switch of VSMCs as well as an alteration of cytoplasmic Ca2+ signaling machinery, a phenomena tightly related to the aging process. Several lines of evidence implicate changes in expression/function levels of IP3R isoforms in the development of hypertension, VSMC phenotypic switch, and vascular aging. The present review discusses the current knowledge of these mechanisms in an integrative approach and further suggests potential new targets for hypertension management and treatment.
Collapse
Affiliation(s)
- Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Fouad Zouein
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Abdelilah Arredouani
- Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Allal Ouhtit
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Md M Rahman
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| | - Gianfranco Pintus
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| | - Haissam Abou-Saleh
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| |
Collapse
|
36
|
Klein A, Joseph PD, Christensen VG, Jensen LJ, Jacobsen JCB. Lack of tone in mouse small mesenteric arteries leads to outward remodeling, which can be prevented by prolonged agonist-induced vasoconstriction. Am J Physiol Heart Circ Physiol 2018; 315:H644-H657. [PMID: 29775408 DOI: 10.1152/ajpheart.00111.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inward remodeling of resistance vessels is an independent risk factor for cardiovascular events. Thus far, the remodeling process remains incompletely elucidated, but the activation level of the vascular smooth muscle cell appears to play a central role. Accordingly, previous data have suggested that an antagonistic and supposedly beneficial response, outward remodeling, may follow prolonged vasodilatation. The present study aimed to determine whether 1) outward remodeling follows 3 days of vessel culture without tone, 2) a similar response can be elicited in a much shorter 4-h timeframe, and, finally, 3) whether a 4-h response can be prevented or reversed by the presence of vasoconstrictors in the medium. Cannulated mouse small mesenteric arteries were organocultured for 3 days in the absence of tone, leading to outward remodeling that continued throughout the culture period. In more acute experiments in which cannulated small mesenteric arteries were maintained in physiological saline without tone for 4 h, we detected a similar outward remodeling that proceeded at a rate several times faster. In the 4-h experimental setting, continuous vasoconstriction to ~50% tone by abluminal application of UTP or norepinephrine + neuropeptide Y prevented outward remodeling but did not cause inward remodeling. Computational modeling was used to simulate and interpret these findings and to derive time constants of the remodeling processes. It is suggested that depriving resistance arteries of activation will lead to eutrophic outward remodeling, which can be prevented by vascular smooth muscle cell activation induced by prolonged vasoconstrictor exposure. NEW & NOTEWORTHY We have established an effective 4-h method for studying outward remodeling in pressurized mouse resistance vessels ex vivo and have determined conditions that block the remodeling response. This allows for investigating the subtle but clinically highly relevant phenomenon of outward remodeling while avoiding both laborious 3-day organoid culture of cannulated vessels and in vivo experiments lasting several weeks.
Collapse
Affiliation(s)
- Anika Klein
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Philomeena Daphne Joseph
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Vibeke Grøsfjeld Christensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Lars Jørn Jensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Jens Christian Brings Jacobsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
37
|
Matin N, Pires PW, Garver H, Jackson WF, Dorrance AM. DOCA-salt hypertension impairs artery function in rat middle cerebral artery and parenchymal arterioles. Microcirculation 2018; 23:571-579. [PMID: 27588564 DOI: 10.1111/micc.12308] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 08/30/2016] [Indexed: 01/25/2023]
Abstract
OBJECTIVE Chronic hypertension induces detrimental changes in the structure and function of surface cerebral arteries. Very little is known about PAs, which perfuse distinct neuronal populations in the cortex and may play a role in cerebrovascular disorders. We investigated the effect of DOCA-salt induced hypertension on endothelial function and artery structure in PAs and MCAs. METHODS Uninephrectomized male Sprague-Dawley rats were implanted with a subcutaneous pellet containing DOCA (150 mg/kg b.w.) and drank salt water (1% NaCl and 0.2% KCl) for 4 weeks. Sham rats were uninephrectomized and drank tap water. Vasoreactivity and passive structure in the MCAs and the PAs were assessed by pressure myography. RESULTS Both MCAs and PAs from DOCA-salt rats exhibited impaired endothelium-dependent dilation (P<.05). In the PAs, addition of NO and COX inhibitors enhanced dilation in DOCA-salt rats (P<.05), suggesting that dysfunctional NO and COX-dependent signaling could contribute to impaired endothelium-mediated dilation. MCAs from DOCA-salt rats exhibited inward remodeling (P<.05). CONCLUSIONS Hypertension-induced MCA remodeling coupled with impaired endothelium-dependent dilation in both the MCAs and PAs may exacerbate the risk of cerebrovascular accidents and the associated morbidity and mortality.
Collapse
Affiliation(s)
- Nusrat Matin
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA.
| | - Paulo W Pires
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA.,Department of Pharmacology, Center for Cardiovascular Research, University of Nevada School of Medicine, Reno, NV, USA
| | - Hannah Garver
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
38
|
Nakada T, Kwee IL, Igarashi H, Suzuki Y. Aquaporin-4 Functionality and Virchow-Robin Space Water Dynamics: Physiological Model for Neurovascular Coupling and Glymphatic Flow. Int J Mol Sci 2017; 18:E1798. [PMID: 28820467 PMCID: PMC5578185 DOI: 10.3390/ijms18081798] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 08/15/2017] [Accepted: 08/16/2017] [Indexed: 11/16/2022] Open
Abstract
The unique properties of brain capillary endothelium, critical in maintaining the blood-brain barrier (BBB) and restricting water permeability across the BBB, have important consequences on fluid hydrodynamics inside the BBB hereto inadequately recognized. Recent studies indicate that the mechanisms underlying brain water dynamics are distinct from systemic tissue water dynamics. Hydrostatic pressure created by the systolic force of the heart, essential for interstitial circulation and lymphatic flow in systemic circulation, is effectively impeded from propagating into the interstitial fluid inside the BBB by the tightly sealed endothelium of brain capillaries. Instead, fluid dynamics inside the BBB is realized by aquaporin-4 (AQP-4), the water channel that connects astrocyte cytoplasm and extracellular (interstitial) fluid. Brain interstitial fluid dynamics, and therefore AQP-4, are now recognized as essential for two unique functions, namely, neurovascular coupling and glymphatic flow, the brain equivalent of systemic lymphatics.
Collapse
Affiliation(s)
- Tsutomu Nakada
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, Niigata 951-8585, Japan.
- Department of Neurology, University of California, Davis, VANCHCS, Martinez, CA 94553, USA.
| | - Ingrid L Kwee
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, Niigata 951-8585, Japan.
- Department of Neurology, University of California, Davis, VANCHCS, Martinez, CA 94553, USA.
| | - Hironaka Igarashi
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, Niigata 951-8585, Japan.
| | - Yuji Suzuki
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, Niigata 951-8585, Japan.
| |
Collapse
|
39
|
Duffin J, Sobczyk O, Crawley A, Poublanc J, Venkatraghavan L, Sam K, Mutch A, Mikulis D, Fisher J. The role of vascular resistance in BOLD responses to progressive hypercapnia. Hum Brain Mapp 2017; 38:5590-5602. [PMID: 28782872 DOI: 10.1002/hbm.23751] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 07/20/2017] [Accepted: 07/20/2017] [Indexed: 12/22/2022] Open
Abstract
The ability of the cerebral vasculature to regulate vascular diameter, hence resistance and cerebral blood flow (CBF), in response to metabolic demands (neurovascular coupling), and perfusion pressure changes (autoregulation) may be assessed by measuring the CBF response to carbon dioxide (CO2 ). In healthy individuals, the CBF response to a ramp CO2 stimulus from hypocapnia to hypercapnia is assumed sigmoidal or linear. However, other response patterns commonly occur, especially in individuals with cerebrovascular disease, and these remain unexplained. CBF responses to CO2 in a vascular region are determined by the combined effects of the innate vascular responses to CO2 and the local perfusion pressure; the latter ensuing from pressure-flow interactions within the cerebral vascular network. We modeled this situation as two vascular beds perfused in parallel from a fixed resistance source. Our premise is that all vascular beds have a sigmoidal reduction of resistance in response to a progressive rise in CO2 . Surrogate CBF data to test the model was provided by magnetic resonance imaging of blood oxygen level-dependent (BOLD) signals. The model successfully generated all the various BOLD-CO2 response patterns, providing a physiological explanation of CBF distribution as relative differences in the network of vascular bed resistance responses to CO2 . Hum Brain Mapp 38:5590-5602, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- James Duffin
- Department of Physiology, University Health Network, Toronto, Canada.,Department of Anaesthesia and Pain Management, University Health Network, University of Toronto, Toronto, Canada
| | - Olivia Sobczyk
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Adrian Crawley
- Joint Department of Medical Imaging and the Functional Neuroimaging Lab, University Health Network, Toronto, Canada
| | - Julien Poublanc
- Joint Department of Medical Imaging and the Functional Neuroimaging Lab, University Health Network, Toronto, Canada
| | - Lashmi Venkatraghavan
- Department of Anaesthesia and Pain Management, University Health Network, University of Toronto, Toronto, Canada
| | - Kevin Sam
- Joint Department of Medical Imaging and the Functional Neuroimaging Lab, University Health Network, Toronto, Canada
| | - Alan Mutch
- Department of Anesthesia and Perioperative Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David Mikulis
- Institute of Medical Science, University of Toronto, Toronto, Canada.,Joint Department of Medical Imaging and the Functional Neuroimaging Lab, University Health Network, Toronto, Canada
| | - Joseph Fisher
- Department of Physiology, University Health Network, Toronto, Canada.,Department of Anaesthesia and Pain Management, University Health Network, University of Toronto, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| |
Collapse
|
40
|
Pires PW, Ko E, Pritchard HA, Rudokas M, Yamasaki E, Earley S. The angiotensin II receptor type 1b is the primary sensor of intraluminal pressure in cerebral artery smooth muscle cells. J Physiol 2017; 595:4735-4753. [PMID: 28475214 PMCID: PMC5509855 DOI: 10.1113/jp274310] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 05/03/2017] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS The angiotensin II receptor type 1b (AT1 Rb ) is the primary sensor of intraluminal pressure in cerebral arteries. Pressure or membrane-stretch induced stimulation of AT1 Rb activates the TRPM4 channel and results in inward transient cation currents that depolarize smooth muscle cells, leading to vasoconstriction. Activation of either AT1 Ra or AT1 Rb with angiotensin II stimulates TRPM4 currents in cerebral artery myocytes and vasoconstriction of cerebral arteries. The expression of AT1 Rb mRNA is ∼30-fold higher than AT1 Ra in whole cerebral arteries and ∼45-fold higher in isolated cerebral artery smooth muscle cells. Higher levels of expression are likely to account for the obligatory role of AT1 Rb for pressure-induced vasoconstriction. ABSTRACT: Myogenic vasoconstriction, which reflects the intrinsic ability of smooth muscle cells to contract in response to increases in intraluminal pressure, is critically important for the autoregulation of blood flow. In smooth muscle cells from cerebral arteries, increasing intraluminal pressure engages a signalling cascade that stimulates cation influx through transient receptor potential (TRP) melastatin 4 (TRPM4) channels to cause membrane depolarization and vasoconstriction. Substantial evidence indicates that the angiotensin II receptor type 1 (AT1 R) is inherently mechanosensitive and initiates this signalling pathway. Rodents express two types of AT1 R - AT1 Ra and AT1 Rb - and conflicting studies provide support for either isoform as the primary sensor of intraluminal pressure in peripheral arteries. We hypothesized that mechanical activation of AT1 Ra increases TRPM4 currents to induce myogenic constriction of cerebral arteries. However, we found that development of myogenic tone was greater in arteries from AT1 Ra knockout animals compared with controls. In patch-clamp experiments using native cerebral arterial myocytes, membrane stretch-induced cation currents were blocked by the TRPM4 inhibitor 9-phenanthrol in both groups. Further, the AT1 R blocker losartan (1 μm) diminished myogenic tone and blocked stretch-induced cation currents in cerebral arteries from both groups. Activation of AT1 R with angiotensin II (30 nm) also increased TRPM4 currents in smooth muscle cells and constricted cerebral arteries from both groups. Expression of AT1 Rb mRNA was ∼30-fold greater than AT1 Ra in cerebral arteries, and knockdown of AT1 Rb selectively diminished myogenic constriction. We conclude that AT1 Rb , acting upstream of TRPM4 channels, is the primary sensor of intraluminal pressure in cerebral artery smooth muscle cells.
Collapse
Affiliation(s)
- Paulo W. Pires
- Department of Pharmacology, Center for Cardiovascular Research, University of NevadaReno School of MedicineRenoNV89557‐0318USA
| | - Eun‐A. Ko
- Department of Physiology and Cell Biology, University of NevadaReno School of MedicineRenoNV89557‐0318USA
| | - Harry A.T. Pritchard
- Department of Pharmacology, Center for Cardiovascular Research, University of NevadaReno School of MedicineRenoNV89557‐0318USA
| | - Michael Rudokas
- Department of Pharmacology, Center for Cardiovascular Research, University of NevadaReno School of MedicineRenoNV89557‐0318USA
| | - Evan Yamasaki
- Department of Pharmacology, Center for Cardiovascular Research, University of NevadaReno School of MedicineRenoNV89557‐0318USA
| | - Scott Earley
- Department of Pharmacology, Center for Cardiovascular Research, University of NevadaReno School of MedicineRenoNV89557‐0318USA
| |
Collapse
|
41
|
El-Yazbi AF, Abd-Elrahman KS. ROK and Arteriolar Myogenic Tone Generation: Molecular Evidence in Health and Disease. Front Pharmacol 2017; 8:87. [PMID: 28280468 PMCID: PMC5322222 DOI: 10.3389/fphar.2017.00087] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/10/2017] [Indexed: 12/13/2022] Open
Abstract
The myogenic response is an inherent property of resistance arteries that warrants a relatively constant blood flow in response to changes in perfusion pressure and protect delicate organs from vascular insufficiencies and excessive blood flow. This fundamental phenomenon has been extensively studied aiming to elucidate the underlying mechanisms triggering smooth muscle contraction in response to intraluminal pressure elevation, particularly, Rho-associated kinase (ROK)-mediated Ca2+-independent mechanisms. The size of the resistance arteries limits the capacity to examine changes in protein phosphorylation/expression levels associated with ROK signaling. A highly sensitive biochemical detection approach was beneficial in examining the role of ROK in different force generation mechanisms along the course of myogenic constriction. In this mini review, we summarize recent results showing direct evidence for the contribution of ROK in development of myogenic response at the level of mechanotransduction, myosin light chain phosphatase inhibition and dynamic actin cytoskeleton reorganization. We will also present evidence that alterations in ROK signaling could underlie the progressive loss in myogenic response in a rat model of type 2 diabetes.
Collapse
Affiliation(s)
- Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of BeirutBeirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria UniversityAlexandria, Egypt
| | - Khaled S Abd-Elrahman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria UniversityAlexandria, Egypt; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of OttawaOttawa, ON, Canada
| |
Collapse
|
42
|
Abd-Elrahman KS, Colinas O, Walsh EJ, Zhu HL, Campbell CM, Walsh MP, Cole WC. Abnormal myosin phosphatase targeting subunit 1 phosphorylation and actin polymerization contribute to impaired myogenic regulation of cerebral arterial diameter in the type 2 diabetic Goto-Kakizaki rat. J Cereb Blood Flow Metab 2017; 37:227-240. [PMID: 26721393 PMCID: PMC5363741 DOI: 10.1177/0271678x15622463] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 10/26/2015] [Accepted: 11/17/2015] [Indexed: 12/11/2022]
Abstract
The myogenic response of cerebral resistance arterial smooth muscle to intraluminal pressure elevation is a key physiological mechanism regulating blood flow to the brain. Rho-associated kinase plays a critical role in the myogenic response by activating Ca2+ sensitization mechanisms: (i) Rho-associated kinase inhibits myosin light chain phosphatase by phosphorylating its targeting subunit myosin phosphatase targeting subunit 1 (at T855), augmenting 20 kDa myosin regulatory light chain (LC20) phosphorylation and force generation; and (ii) Rho-associated kinase stimulates cytoskeletal actin polymerization, enhancing force transmission to the cell membrane. Here, we tested the hypothesis that abnormal Rho-associated kinase-mediated myosin light chain phosphatase regulation underlies the dysfunctional cerebral myogenic response of the Goto-Kakizaki rat model of type 2 diabetes. Basal levels of myogenic tone, LC20, and MYPT1-T855 phosphorylation were elevated and G-actin content was reduced in arteries of pre-diabetic 8-10 weeks Goto-Kakizaki rats with normal serum insulin and glucose levels. Pressure-dependent myogenic constriction, LC20, and myosin phosphatase targeting subunit 1 phosphorylation and actin polymerization were suppressed in both pre-diabetic Goto-Kakizaki and diabetic (18-20 weeks) Goto-Kakizaki rats, whereas RhoA, ROK2, and MYPT1 expression were unaffected. We conclude that abnormal Rho-associated kinase-mediated Ca2+ sensitization contributes to the dysfunctional cerebral myogenic response in the Goto-Kakizaki model of type 2 diabetes.
Collapse
Affiliation(s)
- Khaled S Abd-Elrahman
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Olaia Colinas
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Emma J Walsh
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Hai-Lei Zhu
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Christine M Campbell
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Michael P Walsh
- The Smooth Muscle Research Group, Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - William C Cole
- The Smooth Muscle Research Group, Departments of Physiology & Pharmacology, Libin Cardiovascular Institute & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| |
Collapse
|
43
|
Mohammad K, Hicks M, Buchhalter J, Esser MJ, Irvine L, Thomas S, Scott J, Javadyan J, Kamaluddeen M. Hemodynamic instability associated with increased risk of death or brain injury in neonates with hypoxic ischemic encephalopathy. J Neonatal Perinatal Med 2017; 10:363-370. [PMID: 29843258 DOI: 10.3233/npm-1816162] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
OBJECTIVE To determine the association between hemodynamic instability requiring inotropes and brain injury or death in neonates with hypoxic ischemic encephalopathy (HIE). METHODS Retrospective cohort study of 221 neonates with HIE. Brain injury was defined using four HIE patterns based on MRI diffusion or T1 changes. The primary outcome was death or brain injury. Secondary outcomes were abnormal MRI, death, and abnormal EEG. Logistic regression was used to examine the risk of death or brain injury with the use of inotropes while adjusting for confounding factors. RESULTS Brain injury or death occurred more often in neonates who received inotropes (71.1%, 69/97) compared to those who did not (44.3%, 55/124). The use of inotropes was associated with increased risk of death or brain injury (OR 3.11; 95% CI 1.39-7.004) and abnormal MRI (OR 2.78; 95% CI 1.22-6.34) after adjusting for confounding factors. Mortality was significantly higher in neonates exposed to inotropes (21.6%, 21/97) compared with those who did not receive inotropes (4%, 5/124), P < 0.001. CONCLUSION In infants with HIE, hemodynamic instability requiring inotropes in the first 72 hours of life was associated with increased risk of death or brain injury detected by MRI.
Collapse
Affiliation(s)
- K Mohammad
- Department of Pediatrics, University of Calgary, Calgary, Canada
| | - M Hicks
- Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - J Buchhalter
- Department of Pediatrics, University of Calgary, Calgary, Canada
| | - M J Esser
- Department of Pediatrics, University of Calgary, Calgary, Canada
| | - L Irvine
- Department of Pediatrics, University of Calgary, Calgary, Canada
| | - S Thomas
- Department of Pediatrics, University of Calgary, Calgary, Canada
| | - J Scott
- Department Diagnostic Imaging, University of Calgary, Calgary, Canada
| | - J Javadyan
- Department of Pediatrics, University of Calgary, Calgary, Canada
| | - M Kamaluddeen
- Department of Pediatrics, University of Calgary, Calgary, Canada
| |
Collapse
|
44
|
The Multifaceted Roles of PI3Kγ in Hypertension, Vascular Biology, and Inflammation. Int J Mol Sci 2016; 17:ijms17111858. [PMID: 27834808 PMCID: PMC5133858 DOI: 10.3390/ijms17111858] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/22/2016] [Accepted: 11/03/2016] [Indexed: 12/12/2022] Open
Abstract
PI3Kγ is a multifaceted protein, crucially involved in cardiovascular and immune systems. Several studies described the biological and physiological functions of this enzyme in the regulation of cardiovascular system, while others stressed its role in the modulation of immunity. Although PI3Kγ has been historically investigated for its role in leukocytes, the last decade of research also dedicated efforts to explore its functions in the cardiovascular system. In this review, we report an overview recapitulating how PI3Kγ signaling participates in the regulation of vascular functions involved in blood pressure regulation. Moreover, we also summarize the main functions of PI3Kγ in immune responses that could be potentially important in the interaction with the cardiovascular system. Considering that vascular and immune mechanisms are increasingly emerging as intertwining players in hypertension, PI3Kγ could be an intriguing pathway acting on both sides. The availability of specific inhibitors introduces a perspective of further translational research and clinical approaches that could be exploited in hypertension.
Collapse
|
45
|
Bell ED, Donato AJ, Monson KL. Cerebrovascular dysfunction following subfailure axial stretch. J Mech Behav Biomed Mater 2016; 65:627-633. [PMID: 27736719 DOI: 10.1016/j.jmbbm.2016.09.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 09/17/2016] [Accepted: 09/21/2016] [Indexed: 11/28/2022]
Abstract
Cerebral blood vessels are vital to maintaining the health of the brain. Traumatic brain injury (TBI) commonly results in autoregulatory dysfunction and associated failure of cerebral vessels to maintain homeostasis in the brain. While post-injury changes to brain biochemistry are known to contribute to this dysfunction, tissue deformation may also directly alter vascular smooth muscle cell (SMC) function. As a first step toward understanding stretch-induced dysfunction, this study investigates the effect of overstretch on the contractile behavior of SMCs in middle cerebral arteries (MCAs). We hypothesized that vessel function is altered above a threshold of stretch and strain rate. Twenty-four MCAs from Sprague Dawley rats were tested. Following development of basal SMC tone, vessels were subjected to increasing levels of isosmotic extracellular potassium (K+). Samples were then subjected to an axial overstretch of either 1.2*λIV or 1.3*λIV at strain rates of 0.2 or 20s-1. Following overstretch, SMC contractile behavior was measured again, both immediately and 60min after overstretch. Control vessels were subjected to the same protocol but without overstretch. SMC contractile behavior was characterized using both percent contraction (%C) relative to the fully dilated inner diameter and the K+ dose required to evoke the half maximal contractile response (EC50). Control vessels exhibited increased sensitivity to K+ in successive characterization tests, so all effects were quantified relative to the time-matched control response. Samples exhibited the typical biphasic response to extracellular K+, dilating and contracting in response to small and large K+ concentrations, respectively. As hypothesized, axial overstretch altered SMC contractile behavior, as seen in a decrease in %C for sub-maximal contractile K+ doses (p<0.05) and an increase in EC50 (p<0.01), but only for the test group stretched rapidly to 1.3*λIV. While the change in %C was only significantly different immediately after overstretch, the change to EC50 persisted for 60min. These results indicate that deformation can alter SMC contractile behavior and thus potentially play a role in cerebrovascular autoregulatory dysfunction independent of the pathological chemical environment in the brain post-TBI.
Collapse
Affiliation(s)
- E David Bell
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Laboratory of Head Injury and Vessel Biomechanics, Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Anthony J Donato
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA
| | - Kenneth L Monson
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Laboratory of Head Injury and Vessel Biomechanics, Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
46
|
Tsai HI, Chung PCH, Lee CW, Yu HP. Cerebral perfusion monitoring in acute care surgery: current and perspective use. Expert Rev Med Devices 2016; 13:865-75. [DOI: 10.1080/17434440.2016.1219655] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
47
|
Warnert EAH, Hart EC, Hall JE, Murphy K, Wise RG. The major cerebral arteries proximal to the Circle of Willis contribute to cerebrovascular resistance in humans. J Cereb Blood Flow Metab 2016; 36:1384-95. [PMID: 26661241 PMCID: PMC4976750 DOI: 10.1177/0271678x15617952] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 10/19/2015] [Indexed: 01/22/2023]
Abstract
Cerebral autoregulation ensures constant cerebral blood flow during periods of increased blood pressure by increasing cerebrovascular resistance. However, whether this increase in resistance occurs at the level of major cerebral arteries as well as at the level of smaller pial arterioles is still unknown in humans. Here, we measure cerebral arterial compliance, a measure that is inversely related to cerebrovascular resistance, with our novel non-invasive magnetic resonance imaging-based measurement, which employs short inversion time pulsed arterial spin labelling to map arterial blood volume at different phases of the cardiac cycle. We investigate the differential response of the cerebrovasculature during post exercise ischemia (a stimulus which leads to increased cerebrovascular resistance because of increases in blood pressure and sympathetic outflow). During post exercise ischemia in eight normotensive men (30.4 ± 6.4 years), cerebral arterial compliance decreased in the major cerebral arteries at the level of and below the Circle of Willis, while no changes were measured in arteries above the Circle of Willis. The reduction in arterial compliance manifested as a reduction in the arterial blood volume during systole. This study provides the first evidence that in humans the major cerebral arteries may play an important role in increasing cerebrovascular resistance.
Collapse
Affiliation(s)
- Esther A H Warnert
- Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff, UK
| | - Emma C Hart
- BHI CardioNomics Research Group, School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | - Judith E Hall
- Department of Anaesthetics and Intensive Care Medicine, School of Medicine, Cardiff University, Cardiff, UK
| | - Kevin Murphy
- Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff, UK
| | - Richard G Wise
- Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff, UK
| |
Collapse
|
48
|
|
49
|
Krishnamoorthy-Natarajan G, Koide M. BK Channels in the Vascular System. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 128:401-38. [PMID: 27238270 DOI: 10.1016/bs.irn.2016.03.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Autoregulation of blood flow is essential for the preservation of organ function to ensure continuous supply of oxygen and essential nutrients and removal of metabolic waste. This is achieved by controlling the diameter of muscular arteries and arterioles that exhibit a myogenic response to changes in arterial blood pressure, nerve activity and tissue metabolism. Large-conductance voltage and Ca(2+)-dependent K(+) channels (BK channels), expressed exclusively in smooth muscle cells (SMCs) in the vascular wall of healthy arteries, play a critical role in regulating the myogenic response. Activation of BK channels by intracellular, local, and transient ryanodine receptor-mediated "Ca(2+) sparks," provides a hyperpolarizing influence on the SMC membrane potential thereby decreasing the activity of voltage-dependent Ca(2+) channels and limiting Ca(2+) influx to promote SMC relaxation and vasodilation. The BK channel α subunit, a large tetrameric protein with each monomer consisting of seven-transmembrane domains, a long intracellular C-terminal tail and an extracellular N-terminus, associates with the β1 and γ subunits in vascular SMCs. The BK channel is regulated by factors originating within the SMC or from the endothelium, perivascular nerves and circulating blood, that significantly alter channel gating properties, Ca(2+) sensitivity and expression of the α and/or β1 subunit. The BK channel thus serves as a central receiving dock that relays the effects of the changes in several such concomitant autocrine and paracrine factors and influences cardiovascular health. This chapter describes the primary mechanism of regulation of myogenic response by BK channels and the alterations to this mechanism wrought by different vasoactive mediators.
Collapse
Affiliation(s)
| | - M Koide
- University of Vermont, Burlington, VT, United States
| |
Collapse
|
50
|
Brozovich FV, Nicholson CJ, Degen CV, Gao YZ, Aggarwal M, Morgan KG. Mechanisms of Vascular Smooth Muscle Contraction and the Basis for Pharmacologic Treatment of Smooth Muscle Disorders. Pharmacol Rev 2016; 68:476-532. [PMID: 27037223 PMCID: PMC4819215 DOI: 10.1124/pr.115.010652] [Citation(s) in RCA: 321] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The smooth muscle cell directly drives the contraction of the vascular wall and hence regulates the size of the blood vessel lumen. We review here the current understanding of the molecular mechanisms by which agonists, therapeutics, and diseases regulate contractility of the vascular smooth muscle cell and we place this within the context of whole body function. We also discuss the implications for personalized medicine and highlight specific potential target molecules that may provide opportunities for the future development of new therapeutics to regulate vascular function.
Collapse
Affiliation(s)
- F V Brozovich
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - C J Nicholson
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - C V Degen
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - Yuan Z Gao
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - M Aggarwal
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - K G Morgan
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| |
Collapse
|