1
|
Silva JF, Polk FD, Martin PE, Thai SH, Savu A, Gonzales M, Kath AM, Gee MT, Pires PW. Sex-specific mechanisms of cerebral microvascular BK Ca dysfunction in a mouse model of Alzheimer's disease. Alzheimers Dement 2024. [PMID: 39698895 DOI: 10.1002/alz.14438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/18/2024] [Accepted: 11/06/2024] [Indexed: 12/20/2024]
Abstract
INTRODUCTION Cerebrovascular dysfunction occurs in Alzheimer's disease (AD), impairing hemodynamic regulation. Large conductance Ca2+-activated K+ channels (BKCa) regulate cerebrovascular reactivity and are impaired in AD. BKCa activity depends on intracellular Ca2+ (Ca2+ sparks) and nitro-oxidative post-translational modifications. However, whether these mechanisms underlie BKCa impairment in AD remains unknown. METHODS Cerebral arteries from 5x-FAD and wild-type (WT) littermates were used for molecular biology, electrophysiology, ex vivo, and in vivo experiments. RESULTS Arterial BKCa activity is reduced in 5x-FAD via sex-dependent mechanisms: in males, there is lower BKα subunit expression and less Ca2+ sparks. In females, we observed reversible nitro-oxidative modification of BKCa. Further, BKCa is involved in hemodynamic regulation in WT mice, and its dysfunction is associated with vascular deficits in 5x-FAD. DISCUSSION Our data highlight the central role played by BKCa in cerebral hemodynamic regulation and that molecular mechanisms of its impairment diverge based on sex in 5x-FAD. HIGHLIGHTS Cerebral microvascular BKCa dysfunction occurs in both female and male 5x-FAD. Reduction in BKα subunit protein and Ca2+ sparks drive the dysfunction in males. Nitro-oxidative stress is present in females, but not males, 5x-FAD. Reversible nitro-oxidation of BKα underlies BKCa dysfunction in female 5x-FAD.
Collapse
Affiliation(s)
- Josiane F Silva
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Felipe D Polk
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Paige E Martin
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Stephenie H Thai
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Andrea Savu
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Matthew Gonzales
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Allison M Kath
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Michael T Gee
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Paulo W Pires
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
- Sarver Heart Center, University of Arizona College of Medicine, Tucson, Arizona, USA
- Bio5 Institute, University of Arizona College of Medicine, Tucson, Arizona, USA
| |
Collapse
|
2
|
Dustin CM, Shiva SS, Vazquez A, Saeed A, Pascoal T, Cifuentes-Pagano E, Pagano PJ. NOX2 in Alzheimer's and Parkinson's disease. Redox Biol 2024; 78:103433. [PMID: 39616884 DOI: 10.1016/j.redox.2024.103433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 12/11/2024] Open
Abstract
Alzheimer's Disease (AD), and related dementias, represent a growing concern for the worldwide population given the increased numbers of people of advanced age. Marked by significant degradation of neurological tissues and critical processes, in addition to more specific factors such as the presence of amyloid plaques and neurofibrillary tangles in AD, robust discussion is ongoing regarding the precise mechanisms by which these diseases arise. One of the major interests in recent years has been the contribution of reactive oxygen species (ROS) and, particularly, the contribution of the ROS-generating NADPH Oxidase proteins. NADPH Oxidase 2 (NOX2), the prototypical member of the family, represents a particularly interesting target for study given its close association with vascular and inflammatory processes in all tissues, including the brain, and the association of these processes with AD development and progression. In this review, we discuss the most relevant and recent work regarding the contribution of NOX2 to AD progression in neuronal, microglial, and cerebrovascular signaling. Furthermore, we will discuss the most promising NOX2-targeted therapeutics for potential AD management and treatment.
Collapse
Affiliation(s)
- Christopher M Dustin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Sruti S Shiva
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Alberto Vazquez
- Department of Radiology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 1526, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 1526, USA
| | - Anum Saeed
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Tharick Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Eugenia Cifuentes-Pagano
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Patrick J Pagano
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
3
|
Pacella J, Lembo G, Carnevale L. A Translational Perspective on the Interplay Between Hypertension, Inflammation and Cognitive Impairment. Can J Cardiol 2024; 40:2368-2377. [PMID: 39455022 DOI: 10.1016/j.cjca.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/26/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Hypertension represents the major risk factor in the onset of cardiovascular disease worldwide. Preclinically, several mouse models of hypertension have been developed to investigate the pathophysiological link between hypertension and vascular impairment. Specifically, angiotensin-II infusion, transverse aortic constriction, deoxycorticosterone acetate salt, and N(ω)-nitro-L-arginine methyl ester (L-NAME) administration as hypertensive stimuli at the preclinical level permit the unveiling of a proinflammatory response driven by the innate and adaptive immune system and leads to vascular injury in terms of structural and functional alterations. Vascular impairment seems to be particularly critical at the cerebral level wherein arterioles, venules, and capillaries finely tune blood supply across the whole brain leading to the onset of a well known clinical condition named cerebral small vessel disease (cSVD) characterized by extensive brain injury, which culminates in the decline of cognitive functions. Advances in magnetic resonance imaging permit identification and accurate diagnosis of specific cSVD biomarkers including white matter hyperintensities, lacunar strokes, cerebral microbleeds, and enlarged perivascular spaces, each of which proved to be associated with a specific cognitive domain impairment. Such an approach in combination with pharmacological interventions targeted to the lowering of blood pressure and the prevention of vascular thrombosis formation represents a solid strategy in the prevention and the management of cSVD cognitive decay.
Collapse
Affiliation(s)
- Jacopo Pacella
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy; Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
| | - Lorenzo Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy. https://twitter.com/LorCarnevale
| |
Collapse
|
4
|
Lochhead JJ, Ronaldson PT, Davis TP. The role of oxidative stress in blood-brain barrier disruption during ischemic stroke: Antioxidants in clinical trials. Biochem Pharmacol 2024; 228:116186. [PMID: 38561092 PMCID: PMC11410550 DOI: 10.1016/j.bcp.2024.116186] [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: 01/22/2024] [Revised: 03/19/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Ischemic stroke is one of the leading causes of death and disability. Occlusion and reperfusion of cerebral blood vessels (i.e., ischemia/reperfusion (I/R) injury) generates reactive oxygen species (ROS) that contribute to brain cell death and dysfunction of the blood-brain barrier (BBB) via oxidative stress. BBB disruption influences the pathogenesis of ischemic stroke by contributing to cerebral edema, hemorrhagic transformation, and extravasation of circulating neurotoxic proteins. An improved understanding of mechanisms for ROS-associated alterations in BBB function during ischemia/reperfusion (I/R) injury can lead to improved treatment paradigms for ischemic stroke. Unfortunately, progress in developing ROS targeted therapeutics that are effective for stroke treatment has been slow. Here, we review how ROS are produced in response to I/R injury, their effects on BBB integrity (i.e., tight junction protein complexes, transporters), and the utilization of antioxidant treatments in ischemic stroke clinical trials. Overall, knowledge in this area provides a strong translational framework for discovery of novel drugs for stroke and/or improved strategies to mitigate I/R injury in stroke patients.
Collapse
Affiliation(s)
- Jeffrey J Lochhead
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ 85724, USA.
| | - Patrick T Ronaldson
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Thomas P Davis
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| |
Collapse
|
5
|
Csiszar A, Ungvari A, Patai R, Gulej R, Yabluchanskiy A, Benyo Z, Kovacs I, Sotonyi P, Kirkpartrick AC, Prodan CI, Liotta EM, Zhang XA, Toth P, Tarantini S, Sorond FA, Ungvari Z. Atherosclerotic burden and cerebral small vessel disease: exploring the link through microvascular aging and cerebral microhemorrhages. GeroScience 2024; 46:5103-5132. [PMID: 38639833 PMCID: PMC11336042 DOI: 10.1007/s11357-024-01139-7] [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: 02/20/2024] [Accepted: 03/14/2024] [Indexed: 04/20/2024] Open
Abstract
Cerebral microhemorrhages (CMHs, also known as cerebral microbleeds) are a critical but frequently underestimated aspect of cerebral small vessel disease (CSVD), bearing substantial clinical consequences. Detectable through sensitive neuroimaging techniques, CMHs reveal an extensive pathological landscape. They are prevalent in the aging population, with multiple CMHs often being observed in a given individual. CMHs are closely associated with accelerated cognitive decline and are increasingly recognized as key contributors to the pathogenesis of vascular cognitive impairment and dementia (VCID) and Alzheimer's disease (AD). This review paper delves into the hypothesis that atherosclerosis, a prevalent age-related large vessel disease, extends its pathological influence into the cerebral microcirculation, thereby contributing to the development and progression of CSVD, with a specific focus on CMHs. We explore the concept of vascular aging as a continuum, bridging macrovascular pathologies like atherosclerosis with microvascular abnormalities characteristic of CSVD. We posit that the same risk factors precipitating accelerated aging in large vessels (i.e., atherogenesis), primarily through oxidative stress and inflammatory pathways, similarly instigate accelerated microvascular aging. Accelerated microvascular aging leads to increased microvascular fragility, which in turn predisposes to the formation of CMHs. The presence of hypertension and amyloid pathology further intensifies this process. We comprehensively overview the current body of evidence supporting this interconnected vascular hypothesis. Our review includes an examination of epidemiological data, which provides insights into the prevalence and impact of CMHs in the context of atherosclerosis and CSVD. Furthermore, we explore the shared mechanisms between large vessel aging, atherogenesis, microvascular aging, and CSVD, particularly focusing on how these intertwined processes contribute to the genesis of CMHs. By highlighting the role of vascular aging in the pathophysiology of CMHs, this review seeks to enhance the understanding of CSVD and its links to systemic vascular disorders. Our aim is to provide insights that could inform future therapeutic approaches and research directions in the realm of neurovascular health.
Collapse
Affiliation(s)
- Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Ungvari
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary.
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Zoltan Benyo
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN, Semmelweis University, 1094, Budapest, Hungary
| | - Illes Kovacs
- Department of Ophthalmology, Semmelweis University, 1085, Budapest, Hungary
- Department of Ophthalmology, Weill Cornell Medical College, New York, NY, 10021, USA
| | - Peter Sotonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Angelia C Kirkpartrick
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Calin I Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Eric M Liotta
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Xin A Zhang
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Peter Toth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- Neurotrauma Research Group, Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- ELKH-PTE Clinical Neuroscience MR Research Group, University of Pecs, Pecs, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Farzaneh A Sorond
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
| |
Collapse
|
6
|
da Silva JF, Polk FD, Martin PE, Thai SH, Savu A, Gonzales M, Kath AM, Gee MT, Pires PW. Sex-specific mechanisms of cerebral microvascular BK Ca dysfunction in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.06.543962. [PMID: 37333104 PMCID: PMC10274786 DOI: 10.1101/2023.06.06.543962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
BACKGROUND Cerebral microvascular dysfunction and nitro-oxidative stress are present in patients with Alzheimer's disease (AD) and may contribute to disease progression and severity. Large conductance Ca 2+ -activated K + channels (BK Ca ) play an essential role in vasodilatory responses and maintenance of myogenic tone in resistance arteries. BK Ca impairment can lead to microvascular dysfunction and hemodynamic deficits in the brain. We hypothesized that reduced BK Ca function in cerebral arteries mediates microvascular and neurovascular responses in the 5x-FAD model of AD. METHODS BK Ca activity in the cerebral microcirculation was assessed by patch clamp electrophysiology and pressure myography, in situ Ca 2+ sparks by spinning disk confocal microscopy, hemodynamics by laser speckle contrast imaging. Molecular and biochemical analyses were conducted by affinity-purification assays, qPCR, Western blots and immunofluorescence. RESULTS We observed that pial arteries from 5-6 months-old male and female 5x-FAD mice exhibited a hyper-contractile phenotype than wild-type (WT) littermates, which was linked to lower vascular BK Ca activity and reduced open probability. In males, BK Ca dysfunction is likely a consequence of an observed lower expression of the pore-forming subunit BKα and blunted frequency of Ca 2+ sparks, which are required for BK Ca activity. However, in females, impaired BK Ca function is, in part, a consequence of reversible nitro-oxidative changes in the BK α subunit, which reduces its open probability and regulation of vascular tone. We further show that BK Ca function is involved in neurovascular coupling in mice, and its dysfunction is linked to neurovascular dysfunction in the model. CONCLUSION These data highlight the central role played by BK Ca in cerebral microvascular and neurovascular regulation, as well as sex-dependent mechanisms underlying its dysfunction in a mouse model of AD.
Collapse
|
7
|
Pacholko A, Iadecola C. Hypertension, Neurodegeneration, and Cognitive Decline. Hypertension 2024; 81:991-1007. [PMID: 38426329 PMCID: PMC11023809 DOI: 10.1161/hypertensionaha.123.21356] [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] [Indexed: 03/02/2024]
Abstract
Elevated blood pressure is a well-established risk factor for age-related cognitive decline. Long linked to cognitive impairment on vascular bases, increasing evidence suggests a potential association of hypertension with the neurodegenerative pathology underlying Alzheimer disease. Hypertension is well known to disrupt the structural and functional integrity of the cerebral vasculature. However, the mechanisms by which these alterations lead to brain damage, enhance Alzheimer pathology, and promote cognitive impairment remain to be established. Furthermore, critical questions concerning whether lowering blood pressure by antihypertensive medications prevents cognitive impairment have not been answered. Recent developments in neurovascular biology, brain imaging, and epidemiology, as well as new clinical trials, have provided insights into these critical issues. In particular, clinical and basic findings on the link between neurovascular dysfunction and the pathobiology of neurodegeneration have shed new light on the overlap between vascular and Alzheimer pathology. In this review, we will examine the progress made in the relationship between hypertension and cognitive impairment and, after a critical evaluation of the evidence, attempt to identify remaining knowledge gaps and future research directions that may advance our understanding of one of the leading health challenges of our time.
Collapse
Affiliation(s)
- Anthony Pacholko
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| |
Collapse
|
8
|
Parashar A, Jha D, Mehta V, Chauhan B, Ghosh P, Deb PK, Jaiswal M, Prajapati SK. Sonic hedgehog signalling pathway contributes in age-related disorders and Alzheimer's disease. Ageing Res Rev 2024; 96:102271. [PMID: 38492808 DOI: 10.1016/j.arr.2024.102271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Alzheimer's disease (AD) is caused by the aging process and manifested by cognitive deficits and progressive memory loss. During aging, several conditions, including hypertension, diabetes, and cholesterol, have been identified as potential causes of AD by affecting Sonic hedgehog (Shh) signalling. In addition to being essential for cell differentiation and proliferation, Shh signalling is involved in tissue repair and the prevention of neurodegeneration. Neurogenesis is dependent on Shh signalling; inhibition of this pathway results in neurodegeneration. Several protein-protein interactions that are involved in Shh signalling are implicated in the pathophysiology of AD like overexpression of the protein nexin-1 inhibits the Shh pathway in AD. A protein called Growth Arrest Specific-1 works with another protein called cysteine dioxygenase (CDO) to boost Shh signalling. CDO is involved in the development of the central nervous system (CNS). Shh signalling strengthened the blood brain barrier and therefore prevent the entry of amyloid beta and other toxins to the brain from periphery. Further, several traditional remedies used for AD and dementia, including Epigallocatechin gallate, yokukansan, Lycium barbarum polysaccharides, salvianolic acid, and baicalin, are known to stimulate the Shh pathway. In this review, we elaborated that the Shh signalling exerts a substantial influence on the pathogenesis of AD. In this article, we have tried to explore the various possible connections between the Shh signalling and various known pathologies of AD.
Collapse
Affiliation(s)
- Arun Parashar
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan 173 212, India.
| | - Dhruv Jha
- Birla Institute of Technology, India
| | - Vineet Mehta
- Department of Pharmacology, Government College of Pharmacy, Rohru, District Shimla, Himachal Pradesh 171207, India
| | - Bonney Chauhan
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan 173 212, India
| | - Pappu Ghosh
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan 173 212, India
| | - Prashanta Kumar Deb
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan 173 212, India
| | | | | |
Collapse
|
9
|
Villa RF, Ferrari F, Gorini A. Effects of Chronic Hypertension on the Energy Metabolism of Cerebral Cortex Mitochondria in Normotensive and in Spontaneously Hypertensive Rats During Aging. Neuromolecular Med 2024; 26:2. [PMID: 38393429 DOI: 10.1007/s12017-023-08772-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 12/02/2023] [Indexed: 02/25/2024]
Abstract
In this study the subcellular modifications undergone by cerebral cortex mitochondrial metabolism in chronic hypertension during aging were evaluated. The catalytic properties of regulatory energy-linked enzymes of Tricarboxylic Acid Cycle (TCA), Electron Transport Chain (ETC) and glutamate metabolism were assayed on non-synaptic mitochondria (FM, located in post-synaptic compartment) and on intra-synaptic mitochondria of pre-synaptic compartment, furtherly divided in "light" (LM) and "heavy" (HM) mitochondria, purified form cerebral cortex of normotensive Wistar Kyoto Rats (WKY) versus Spontaneously Hypertensive Rats (SHR) at 6, 12 and 18 months. During physiological aging, the metabolic machinery was differently expressed in pre- and post-synaptic compartments: LM and above all HM were more affected by aging, displaying lower ETC activities. In SHR at 6 months, FM and LM showed an uncoupling between TCA and ETC, likely as initial adaptive response to hypertension. During pathological aging, HM were particularly affected at 12 months in SHR, as if the adaptive modifications in FM and LM at 6 months granted a mitochondrial functional balance, while at 18 months all the neuronal mitochondria displayed decreased metabolic fluxes versus WKY. This study describes the effects of chronic hypertension on cerebral mitochondrial energy metabolism during aging through functional proteomics of enzymes at subcellular levels, i.e. in neuronal soma and synapses. In addition, this represents the starting point to envisage an experimental physiopathological model which could be useful also for pharmacological studies, to assess drug actions during the development of age-related pathologies that could coexist and/or are provoked by chronic hypertension.
Collapse
Affiliation(s)
- Roberto Federico Villa
- Department of Biology and Biotechnology, Laboratory of Pharmacology and Molecular Medicine of Central Nervous System, University of Pavia, Via Ferrata, 9, 27100, Pavia, Italy.
| | - Federica Ferrari
- Department of Biology and Biotechnology, Laboratory of Pharmacology and Molecular Medicine of Central Nervous System, University of Pavia, Via Ferrata, 9, 27100, Pavia, Italy
- School of Neurology, Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi. 21, 27100, Pavia, Italy
| | - Antonella Gorini
- Department of Biology and Biotechnology, Laboratory of Pharmacology and Molecular Medicine of Central Nervous System, University of Pavia, Via Ferrata, 9, 27100, Pavia, Italy
| |
Collapse
|
10
|
Ma H, Wang L, Sun H, Yu Q, Yang T, Wang Y, Niu B, Jia Y, Liu Y, Liang Z, An M, Guo J. MIR-107/HMGB1/FGF-2 axis responds to excessive mechanical stretch to promote rapid repair of vascular endothelial cells. Arch Biochem Biophys 2023:109686. [PMID: 37406794 DOI: 10.1016/j.abb.2023.109686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/01/2023] [Accepted: 07/02/2023] [Indexed: 07/07/2023]
Abstract
The increase of vascular wall tension can lead to endothelial injury during hypertension, but its potential mechanism remains to be studied. Our results of previous study showed that HUVECs could induce changes in HMGB1/RAGE to resist abnormal mechanical environments in pathological mechanical stretching. In this study, we applied two different kinds of mechanical tension to endothelial cells using the in vitro mechanical loading system FlexCell-5000T and focused on exploring the expression of miR-107 related pathways in HUVECs with excessive mechanical tension. The results showed that miR-107 negatively regulated the expression of the HMGB1/RAGE axis under excessive mechanical tension. Excessive mechanical stretching reduced the expression of miR-107 in HUVECs, and increased the expression of the HMGB1/RAGE axis. When miR-107 analog was transfected into HUVECs with lipo3000 reagent, the overexpression of miR-107 slowed down the increase of the HMGB1/RAGE axis caused by excessive mechanical stretching. At the same time, the overexpression of miR-107 inhibited the proliferation and migration of HUVECs to a certain extent. On the contrary, when miR-107 was silent, the proliferation and migration of HUVECs showed an upward trend. In addition, the study also showed that under excessive mechanical tension, miR-107 could regulate the expression of FGF-2 by HMGB1. In conclusion, these findings suggest that pathological mechanical stretching promote resistance to abnormal mechanical stimulation on HUVECs through miR-107/HMGB1/RAGE/FGF-2 pathway, thus promote vascular repair after endothelial injury. The suggest that miR-107 is a potential therapeutic target for hypertension.
Collapse
Affiliation(s)
- Haiyang Ma
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi, 030024, PR China
| | - Li Wang
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi, 030024, PR China
| | - Haoyu Sun
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi, 030024, PR China
| | - Qing Yu
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi, 030024, PR China
| | - Tiantian Yang
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi, 030024, PR China
| | - Yajing Wang
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi, 030024, PR China
| | - Bin Niu
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi, 030024, PR China
| | - Yaru Jia
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi, 030024, PR China
| | - Yang Liu
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi, 030024, PR China
| | - Ziwei Liang
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi, 030024, PR China
| | - Meiwen An
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi, 030024, PR China.
| | - Jiqiang Guo
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, PR China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China; Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi, 030024, PR China.
| |
Collapse
|
11
|
Nunes JM, Kell DB, Pretorius E. Cardiovascular and haematological pathology in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS): A role for viruses. Blood Rev 2023; 60:101075. [PMID: 36963989 PMCID: PMC10027292 DOI: 10.1016/j.blre.2023.101075] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023]
Abstract
ME/CFS is a debilitating chronic condition that often develops after viral or bacterial infection. Insight from the study of Long COVID/Post Acute Sequelae of COVID-19 (PASC), the post-viral syndrome associated with SARS-CoV-2 infection, might prove to be useful for understanding pathophysiological mechanisms of ME/CFS. Disease presentation is similar between the two conditions, and a subset of Long COVID patients meet the diagnostic criteria for ME/CFS. Since Long COVID is characterized by significant vascular pathology - including endothelial dysfunction, coagulopathy, and vascular dysregulation - the question of whether or not the same biological abnormalities are of significance in ME/CFS arises. Cardiac abnormalities have for a while now been documented in ME/CFS cohorts, with recent studies demonstrating major deficits in cerebral blood flow, and hence vascular dysregulation. A growing body of research is demonstrating that ME/CFS is accompanied by platelet hyperactivation, anomalous clotting, a procoagulant phenotype, and endothelial dysfunction. Endothelial damage and dysregulated clotting can impair substance exchange between blood and tissues, and result in hypoperfusion, which may contribute to the manifestation of certain ME/CFS symptoms. Here we review the ME/CFS literature to summarize cardiovascular and haematological findings documented in patients with the condition, and, in this context, briefly discuss the potential role of previously-implicated pathogens. Overall, cardiac and haematological abnormalities are present within ME/CFS cohorts. While atherosclerotic heart disease is not significantly associated with ME/CFS, suboptimal cardiovascular function defined by reduced cardiac output, impaired cerebral blood flow, and vascular dysregulation are, and these abnormalities do not appear to be influenced by deconditioning. Rather, these cardiac abnormalities may result from dysfunction in the (autonomic) nervous system. Plenty of recently published studies are demonstrating significant platelet hyperactivity and endothelial dysfunction in ME/CFS, as well as anomalous clotting processes. It is of particular importance to determine to what extent these cardiovascular and haematological abnormalities contribute to symptom severity, and if these two systems can be targeted for therapeutic purposes. Viral reservoirs of herpesviruses exist in ME/CFS, and most likely contribute to cardiovascular and haematological dysfunction directly or indirectly. This review highlights the potential of studying cardiac functioning, the vasculature, and coagulation system in ME/CFS.
Collapse
Affiliation(s)
- Jean M Nunes
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa.
| | - Douglas B Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa; Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool L69 7ZB, UK; The Novo Nordisk Foundation Centre for Biosustainability, Building 220, Chemitorvet 200, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa; Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool L69 7ZB, UK.
| |
Collapse
|
12
|
Uribe P, Barra J, Painen K, Zambrano F, Schulz M, Moya C, Isachenko V, Isachenko E, Mallmann P, Sánchez R. FeTPPS, a Peroxynitrite Decomposition Catalyst, Ameliorates Nitrosative Stress in Human Spermatozoa. Antioxidants (Basel) 2023; 12:1272. [PMID: 37372002 DOI: 10.3390/antiox12061272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/01/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
Excessive levels of reactive nitrogen species (RNS), such as peroxynitrite, promote nitrosative stress, which is an important cause of impaired sperm function. The metalloporphyrin FeTPPS is highly effective in catalyzing the decomposition of peroxynitrite, reducing its toxic effects in vivo and in vitro. FeTPPS has significant therapeutic potential in peroxynitrite-related diseases; however, its effects on human spermatozoa under nitrosative stress have not been described. This work aimed to evaluate the in vitro effect of FeTPPS against peroxynitrite-mediated nitrosative stress in human spermatozoa. For this purpose, spermatozoa from normozoospermic donors were exposed to 3-morpholinosydnonimine, a molecule that generates peroxynitrite. First, the FeTPPS-mediated peroxynitrite decomposition catalysis was analyzed. Then, its individual effect on sperm quality parameters was evaluated. Finally, the effect of FeTPPS on ATP levels, motility, mitochondrial membrane potential, thiol oxidation, viability, and DNA fragmentation was analyzed in spermatozoa under nitrosative stress conditions. The results showed that FeTPPS effectively catalyzes the decomposition of peroxynitrite without affecting sperm viability at concentrations up to 50 μmol/L. Furthermore, FeTPPS mitigates the deleterious effects of nitrosative stress on all sperm parameters analyzed. These results highlight the therapeutic potential of FeTPPS in reducing the negative impact of nitrosative stress in semen samples with high RNS levels.
Collapse
Affiliation(s)
- Pamela Uribe
- Center of Excellence in Translational Medicine-Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Faculty of Medicine, Universidad de La Frontera, Temuco 4810296, Chile
- Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Temuco 4781176, Chile
| | - Javiera Barra
- Center of Excellence in Translational Medicine-Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Faculty of Medicine, Universidad de La Frontera, Temuco 4810296, Chile
| | - Kevin Painen
- Center of Excellence in Translational Medicine-Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Faculty of Medicine, Universidad de La Frontera, Temuco 4810296, Chile
| | - Fabiola Zambrano
- Center of Excellence in Translational Medicine-Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Faculty of Medicine, Universidad de La Frontera, Temuco 4810296, Chile
- Department of Preclinical Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4781176, Chile
| | - Mabel Schulz
- Center of Excellence in Translational Medicine-Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Faculty of Medicine, Universidad de La Frontera, Temuco 4810296, Chile
- Department of Preclinical Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4781176, Chile
| | - Claudia Moya
- Center of Excellence in Translational Medicine-Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Faculty of Medicine, Universidad de La Frontera, Temuco 4810296, Chile
| | - Vladimir Isachenko
- Research Group in Reproductive Medicine, Department of Obstetrics and Gynecology, Cologne University, 50923 Köln, Germany
| | - Evgenia Isachenko
- Research Group in Reproductive Medicine, Department of Obstetrics and Gynecology, Cologne University, 50923 Köln, Germany
| | - Peter Mallmann
- Research Group in Reproductive Medicine, Department of Obstetrics and Gynecology, Cologne University, 50923 Köln, Germany
| | - Raúl Sánchez
- Center of Excellence in Translational Medicine-Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Faculty of Medicine, Universidad de La Frontera, Temuco 4810296, Chile
- Department of Preclinical Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4781176, Chile
| |
Collapse
|
13
|
Waigi EW, Webb RC, Moss MA, Uline MJ, McCarthy CG, Wenceslau CF. Soluble and insoluble protein aggregates, endoplasmic reticulum stress, and vascular dysfunction in Alzheimer's disease and cardiovascular diseases. GeroScience 2023; 45:1411-1438. [PMID: 36823398 PMCID: PMC10400528 DOI: 10.1007/s11357-023-00748-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/28/2023] [Indexed: 02/25/2023] Open
Abstract
Dementia refers to a particular group of symptoms characterized by difficulties with memory, language, problem-solving, and other thinking skills that affect a person's ability to perform everyday activities. Alzheimer's disease (AD) is the most common form of dementia, affecting about 6.2 million Americans aged 65 years and older. Likewise, cardiovascular diseases (CVDs) are a major cause of disability and premature death, impacting 126.9 million adults in the USA, a number that increases with age. Consequently, CVDs and cardiovascular risk factors are associated with an increased risk of AD and cognitive impairment. They share important age-related cardiometabolic and lifestyle risk factors, that make them among the leading causes of death. Additionally, there are several premises and hypotheses about the mechanisms underlying the association between AD and CVD. Although AD and CVD may be considered deleterious to health, the study of their combination constitutes a clinical challenge, and investigations to understand the mechanistic pathways for the cause-effect and/or shared pathology between these two disease constellations remains an active area of research. AD pathology is propagated by the amyloid β (Aβ) peptides. These peptides give rise to small, toxic, and soluble Aβ oligomers (SPOs) that are nonfibrillar, and it is their levels that show a robust correlation with the extent of cognitive impairment. This review will elucidate the interplay between the effects of accumulating SPOs in AD and CVDs, the resulting ER stress response, and their role in vascular dysfunction. We will also address the potential underlying mechanisms, including the possibility that SPOs are among the causes of vascular injury in CVD associated with cognitive decline. By revealing common mechanistic underpinnings of AD and CVD, we hope that novel experimental therapeutics can be designed to reduce the burden of these devastating diseases. Graphical abstract Alzheimer's disease (AD) pathology leads to the release of Aβ peptides, and their accumulation in the peripheral organs has varying effects on various components of the cardiovascular system including endoplasmic reticulum (ER) stress and vascular damage. Image created with BioRender.com.
Collapse
Affiliation(s)
- Emily W Waigi
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - R Clinton Webb
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
| | - Melissa A Moss
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
- Department of Chemical Engineering, University of South Carolina, Columbia, SC, USA
| | - Mark J Uline
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
- Department of Chemical Engineering, University of South Carolina, Columbia, SC, USA
| | - Cameron G McCarthy
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
| | - Camilla Ferreira Wenceslau
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA.
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA.
| |
Collapse
|
14
|
Maryam K, Ali H. Aerobic and resistance exercises affect the BDNF/TrkB signaling pathway, and hippocampal neuron density of high-fat diet-induced obese elderly rats. Physiol Behav 2023; 264:114140. [PMID: 36870384 DOI: 10.1016/j.physbeh.2023.114140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/05/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Obesity, caused by a high-fat diet (HFD) in elderly, is a risk factor for insulin resistance and a precursor to diabetes and can lead to impaired cognitive function. Physical exercise has positive effects on decrease obesity and improvement brain function. We investigated which type of aerobic (AE) or resistance (RE) exercise can be more effective in reducing HFD-induced cognitive dysfunction in obese elderly rats. 48 male Wistar rats (19-monthold) were divided into six groups: Healthy control (CON), CON+AE, CON+RE, HFD, HFD+AE, and HFD+RE. Obesity was induced by 5 months of HFD feeding in older rats. After obesity confirmation, RT (with a range of 50% to 100%1RM/3 days/week) and AE (running at 8-m/min for 15-min to 26-m/min for 60-min /5 days/week) was performed for 12-weeks. Morris water maze Test was used to evaluate cognitive performance. All data were analyzed using two-way statistical test of variance. The results showed that obesity had a negative effect on glycemic index, increased inflammation, decreased antioxidant levels, decreased BDNF/TrkB and decreased nerve density in hippocampal tissue. The Morris water maze results clearly showed cognitive impairment in the obesity group. But 12 weeks after AE and RE, all the measured variables were on the improvement path, and in general, no difference was observed between the two exercise methods. Two mods of exercise (AE and RE) may be having same effects on nerve cell density, inflammatory, antioxidant and functional status of hippocampus of obese rats. Each of the AE and RE can create beneficial effects on the cognitive function of the elderly.
Collapse
Affiliation(s)
- Keshvari Maryam
- Faculty of Sport Sciences, exercise physiology department, Bu-Ali Sina University, Hamadan, Iran
| | - Heidarianpour Ali
- Faculty of Sport Sciences, exercise physiology department, Bu-Ali Sina University, Hamadan, Iran.
| |
Collapse
|
15
|
Kujovic M, Lipka T, Zalman M, Baumann L, Jänner M, Baumann B. Treatment of hypertension and obstructive sleep apnea counteracts cognitive decline in common neurocognitive disorders in diagnosis-related patterns. Sci Rep 2023; 13:7556. [PMID: 37160982 PMCID: PMC10169815 DOI: 10.1038/s41598-023-33701-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/18/2023] [Indexed: 05/11/2023] Open
Abstract
The aim of this study was to investigate the effect of arterial hypertension (AH) and of obstructive sleep apnea (OSA) on cognitive course in the neurocognitive disorder (NCD) cohort RIFADE which enrolled patients with NCD due to Alzheimer's disease (AD), vascular NCD (vNCD), and mixed NCD (AD + vNCD = mNCD). Multiple risk factors (RF), including AH and OSA, that contribute to the development of various kinds of dementia have been identified in previous studies. Studies that observed AH lacked investigation of long-term effects and did not isolate it from other RF. Studies involving OSA as a risk factor did not include participants with all stages of NCD. 126 subjects were screened for AH and OSA. Repeated cognitive measurements were performed with the DemTect as primary outcome and the clock drawing test as secondary outcome measure. 90 patients had AH (71.4%) and 40 patients had OSA (31.7%). RF-status had a significant effect on cognitive outcome in models with RF as single factors (AH p = 0.027, OSA p < 0.001), a 2-factor analysis with AH × OSA (AH as main factor p = 0.027) as well as a model including the 3 factors AH × OSA × diagnosis (p = 0.038). Similarly, a 3-factor model was significant for the clock-drawing test, whereas single factor-models remained insignificant. AH and OSA appear to be risk factors in common NCD and cognitive decline can be mitigated by treatment of these RF.
Collapse
Affiliation(s)
- Milenko Kujovic
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany.
- Department of Neuropsychiatry, Centre for Neurology and Neuropsychiatry, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany.
| | - Tim Lipka
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Mark Zalman
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Leonie Baumann
- Department of Mental Health, University Hospital of Münster, Munster, Germany
| | - Michaela Jänner
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Bruno Baumann
- Department of Mental Health, University Hospital of Münster, Munster, Germany
| |
Collapse
|
16
|
Neurovascular Coupling in Hypertension Is Impaired by IL-17A through Oxidative Stress. Int J Mol Sci 2023; 24:ijms24043959. [PMID: 36835372 PMCID: PMC9967204 DOI: 10.3390/ijms24043959] [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: 12/22/2022] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Hypertension, a multifactorial chronic inflammatory condition, is an important risk factor for neurovascular and neurodegenerative diseases, including stroke and Alzheimer's disease. These diseases have been associated with higher concentrations of circulating interleukin (IL)-17A. However, the possible role that IL-17A plays in linking hypertension with neurodegenerative diseases remains to be established. Cerebral blood flow regulation may be the crossroads of these conditions because regulating mechanisms may be altered in hypertension, including neurovascular coupling (NVC), known to participate in the pathogenesis of stroke and Alzheimer's disease. In the present study, the role of IL-17A on NVC impairment induced by angiotensin (Ang) II in the context of hypertension was examined. Neutralization of IL-17A or specific inhibition of its receptor prevents the NVC impairment (p < 0.05) and cerebral superoxide anion production (p < 0.05) induced by Ang II. Chronic administration of IL-17A impairs NVC (p < 0.05) and increases superoxide anion production. Both effects were prevented with Tempol and NADPH oxidase 2 gene deletion. These findings suggest that IL-17A, through superoxide anion production, is an important mediator of cerebrovascular dysregulation induced by Ang II. This pathway is thus a putative therapeutic target to restore cerebrovascular regulation in hypertension.
Collapse
|
17
|
Brain Bioenergetics in Chronic Hypertension: Risk Factor for Acute Ischemic Stroke. Biochem Pharmacol 2022; 205:115260. [PMID: 36179931 DOI: 10.1016/j.bcp.2022.115260] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/23/2022]
Abstract
Chronic hypertension is one of the key modifiable risk factors for acute ischemic stroke, also contributing to determine greater neurological deficits and worse functional outcome when an acute cerebrovascular event would occur. A tight relationship exists between cerebrovascular autoregulation, neuronal activity and brain bioenergetics. In chronic hypertension, progressive adaptations of these processes occur as an attempt to cope with the demanding necessity of brain functions, creating a new steady-state homeostatic condition. However, these adaptive modifications are insufficient to grant an adequate response to possible pathological perturbations of the established fragile hemodynamic and metabolic homeostasis. In this narrative review, we will discuss the main mechanisms by which alterations in brain bioenergetics and mitochondrial function in chronic hypertension could lead to increased risk of acute ischemic stroke, stressing the interconnections between hemodynamic factors (i.e. cerebral autoregulation and neurovascular coupling) and metabolic processes. Both experimental and clinical pieces of evidence will be discussed. Moreover, the potential role of mitochondrial dysfunction in determining, or at least sustaining, the pathogenesis and progression of chronic neurogenic hypertension will be considered. In the perspective of novel therapeutic strategies aiming at improving brain bioenergetics, we propose some determinant factors to consider in future studies focused on the cause-effect relationships between chronic hypertension and brain bioenergetic abnormalities (and vice versa), so to help translational research in this so-far unfilled gap.
Collapse
|
18
|
Barloese MCJ, Bauer C, Petersen ET, Hansen CS, Madsbad S, Siebner HR. Neurovascular Coupling in Type 2 Diabetes With Cognitive Decline. A Narrative Review of Neuroimaging Findings and Their Pathophysiological Implications. Front Endocrinol (Lausanne) 2022; 13:874007. [PMID: 35860697 PMCID: PMC9289474 DOI: 10.3389/fendo.2022.874007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/17/2022] [Indexed: 01/21/2023] Open
Abstract
Type 2 diabetes causes substantial long-term damage in several organs including the brain. Cognitive decline is receiving increased attention as diabetes has been established as an independent risk factor along with the identification of several other pathophysiological mechanisms. Early detection of detrimental changes in cerebral blood flow regulation may represent a useful clinical marker for development of cognitive decline for at-risk persons. Technically, reliable evaluation of neurovascular coupling is possible with several caveats but needs further development before it is clinically convenient. Different modalities including ultrasound, positron emission tomography and magnetic resonance are used preclinically to shed light on the many influences on vascular supply to the brain. In this narrative review, we focus on the complex link between type 2 diabetes, cognition, and neurovascular coupling and discuss how the disease-related pathology changes neurovascular coupling in the brain from the organ to the cellular level. Different modalities and their respective pitfalls are covered, and future directions suggested.
Collapse
Affiliation(s)
- Mads C. J. Barloese
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
- Department of Clinical Physiology and Nuclear Imaging, Center for Functional and Diagnostic Imaging, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Christian Bauer
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
- Radiography, Department of Technology, University College Copenhagen, Copenhagen, Denmark
| | - Esben Thade Petersen
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
- Center for Magnetic Resonance, Department of Electrical Engineering, Technical University of Denmark, Lyngby, Denmark
| | | | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hartwig Roman Siebner
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology, Copenhagen University Hospital - Bispebjerg and Fredriksberg, Copenhagen, Denmark
| |
Collapse
|
19
|
Alfieri A, Koudelka J, Li M, Scheffer S, Duncombe J, Caporali A, Kalaria RN, Smith C, Shah AM, Horsburgh K. Nox2 underpins microvascular inflammation and vascular contributions to cognitive decline. J Cereb Blood Flow Metab 2022; 42:1176-1191. [PMID: 35102790 PMCID: PMC9207496 DOI: 10.1177/0271678x221077766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 11/26/2021] [Accepted: 12/06/2021] [Indexed: 11/16/2022]
Abstract
Chronic microvascular inflammation and oxidative stress are inter-related mechanisms underpinning white matter disease and vascular cognitive impairment (VCI). A proposed mediator is nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (Nox2), a major source of reactive oxygen species (ROS) in the brain. To assess the role of Nox2 in VCI, we studied a tractable model with white matter pathology and cognitive impairment induced by bilateral carotid artery stenosis (BCAS). Mice with genetic deletion of Nox2 (Nox2 KO) were compared to wild-type (WT) following BCAS. Sustained BCAS over 12 weeks in WT mice induced Nox2 expression, indices of microvascular inflammation and oxidative damage, along with white matter pathology culminating in a marked cognitive impairment, which were all protected by Nox2 genetic deletion. Neurovascular coupling was impaired in WT mice post-BCAS and restored in Nox2 KO mice. Increased vascular expression of chemoattractant mediators, cell-adhesion molecules and endothelial activation factors in WT mice post-BCAS were ameliorated by Nox2 deficiency. The clinical relevance was confirmed by increased vascular Nox2 and indices of microvascular inflammation in human post-mortem subjects with cerebral vascular disease. Our results support Nox2 activity as a critical determinant of VCI, whose targeting may be of therapeutic benefit in cerebral vascular disease.
Collapse
Affiliation(s)
- Alessio Alfieri
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- National Heart and Lung Institute, Vascular Science, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - Juraj Koudelka
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Mosi Li
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Sanny Scheffer
- Department of Pathology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Jessica Duncombe
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Andrea Caporali
- British Heart Foundation Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Rajesh N Kalaria
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle-Upon-Tyne, UK
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Ajay M Shah
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, London, UK
| | - Karen Horsburgh
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
20
|
Barkaway A, Attwell D, Korte N. Immune-vascular mural cell interactions: consequences for immune cell trafficking, cerebral blood flow, and the blood-brain barrier. NEUROPHOTONICS 2022; 9:031914. [PMID: 35581998 PMCID: PMC9107322 DOI: 10.1117/1.nph.9.3.031914] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/13/2022] [Indexed: 06/15/2023]
Abstract
Brain barriers are crucial sites for cerebral energy supply, waste removal, immune cell migration, and solute exchange, all of which maintain an appropriate environment for neuronal activity. At the capillary level, where the largest area of brain-vascular interface occurs, pericytes adjust cerebral blood flow (CBF) by regulating capillary diameter and maintain the blood-brain barrier (BBB) by suppressing endothelial cell (EC) transcytosis and inducing tight junction expression between ECs. Pericytes also limit the infiltration of circulating leukocytes into the brain where resident microglia confine brain injury and provide the first line of defence against invading pathogens. Brain "waste" is cleared across the BBB into the blood, phagocytosed by microglia and astrocytes, or removed by the flow of cerebrospinal fluid (CSF) through perivascular routes-a process driven by respiratory motion and the pulsation of the heart, arteriolar smooth muscle, and possibly pericytes. "Dirty" CSF exits the brain and is probably drained around olfactory nerve rootlets and via the dural meningeal lymphatic vessels and possibly the skull bone marrow. The brain is widely regarded as an immune-privileged organ because it is accessible to few antigen-primed leukocytes. Leukocytes enter the brain via the meninges, the BBB, and the blood-CSF barrier. Advances in genetic and imaging tools have revealed that neurological diseases significantly alter immune-brain barrier interactions in at least three ways: (1) the brain's immune-privileged status is compromised when pericytes are lost or lymphatic vessels are dysregulated; (2) immune cells release vasoactive molecules to regulate CBF, modulate arteriole stiffness, and can plug and eliminate capillaries which impairs CBF and possibly waste clearance; and (3) immune-vascular interactions can make the BBB leaky via multiple mechanisms, thus aggravating the influx of undesirable substances and cells. Here, we review developments in these three areas and briefly discuss potential therapeutic avenues for restoring brain barrier functions.
Collapse
Affiliation(s)
- Anna Barkaway
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| | - David Attwell
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| | - Nils Korte
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| |
Collapse
|
21
|
Kinzenbaw DA, Langmack L, Faraci FM. Angiotensin II-induced endothelial dysfunction: Impact of sex, genetic background, and rho kinase. Physiol Rep 2022; 10:e15336. [PMID: 35681278 PMCID: PMC9184751 DOI: 10.14814/phy2.15336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 06/15/2023] Open
Abstract
The renin-angiotensin system (RAS) contributes to vascular disease with multiple cardiovascular risk factors including hypertension. As a major effector within the RAS, angiotensin II (Ang II) activates diverse signaling mechanisms that affect vascular biology. Despite the impact of such vascular pathophysiology, our understanding of the effects of Ang II in relation to the function of endothelial cells is incomplete. Because genetic background and biological sex can be determinants of vascular disease, we performed studies examining the direct effects of Ang II using carotid arteries from male and female mice on two genetic backgrounds, C57BL/6J and FVB/NJ. Although FVB/NJ mice are much less susceptible to atherosclerosis than C57BL/6J, the effects of Ang II on endothelial cells in FVB/NJ are poorly defined. Overnight incubation of isolated arteries with Ang II (10 nmol/L), impaired endothelial function in both strains and sexes by approximately one-half (p < 0.05). To examine the potential mechanistic contribution of Rho kinase (ROCK), we treated arteries with SLX-2119, an inhibitor with high selectivity for ROCK2. In both male and female mice of both strains, SLX-2119 largely restored endothelial function to normal, compared to vessels treated with vehicle. Thus, Ang II-induced endothelial dysfunction was observed in both FVB/NJ and C57BL/6J mice. This effect was sex-independent. In all groups, effects of Ang II were reversed by inhibition of ROCK2 with SLX-2119. These studies provide the first evidence that ROCK2 may be a key contributor to Ang II-induced endothelial dysfunction in both sexes and in mouse strains that differ in relation to other major aspects of vascular disease.
Collapse
Affiliation(s)
- Dale A. Kinzenbaw
- Departments of Internal MedicineFrancois M. Abboud Cardiovascular CenterThe University of Iowa Carver College of MedicineIowa CityIowaUSA
| | - Lucy Langmack
- Departments of Internal MedicineFrancois M. Abboud Cardiovascular CenterThe University of Iowa Carver College of MedicineIowa CityIowaUSA
| | - Frank M. Faraci
- Departments of Internal MedicineFrancois M. Abboud Cardiovascular CenterThe University of Iowa Carver College of MedicineIowa CityIowaUSA
- Departments of Neuroscience and PharmacologyThe University of Iowa Carver College of MedicineIowa CityIowaUSA
| |
Collapse
|
22
|
Knipper M, Singer W, Schwabe K, Hagberg GE, Li Hegner Y, Rüttiger L, Braun C, Land R. Disturbed Balance of Inhibitory Signaling Links Hearing Loss and Cognition. Front Neural Circuits 2022; 15:785603. [PMID: 35069123 PMCID: PMC8770933 DOI: 10.3389/fncir.2021.785603] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/08/2021] [Indexed: 12/19/2022] Open
Abstract
Neuronal hyperexcitability in the central auditory pathway linked to reduced inhibitory activity is associated with numerous forms of hearing loss, including noise damage, age-dependent hearing loss, and deafness, as well as tinnitus or auditory processing deficits in autism spectrum disorder (ASD). In most cases, the reduced central inhibitory activity and the accompanying hyperexcitability are interpreted as an active compensatory response to the absence of synaptic activity, linked to increased central neural gain control (increased output activity relative to reduced input). We here suggest that hyperexcitability also could be related to an immaturity or impairment of tonic inhibitory strength that typically develops in an activity-dependent process in the ascending auditory pathway with auditory experience. In these cases, high-SR auditory nerve fibers, which are critical for the shortest latencies and lowest sound thresholds, may have either not matured (possibly in congenital deafness or autism) or are dysfunctional (possibly after sudden, stressful auditory trauma or age-dependent hearing loss linked with cognitive decline). Fast auditory processing deficits can occur despite maintained basal hearing. In that case, tonic inhibitory strength is reduced in ascending auditory nuclei, and fast inhibitory parvalbumin positive interneuron (PV-IN) dendrites are diminished in auditory and frontal brain regions. This leads to deficits in central neural gain control linked to hippocampal LTP/LTD deficiencies, cognitive deficits, and unbalanced extra-hypothalamic stress control. Under these conditions, a diminished inhibitory strength may weaken local neuronal coupling to homeostatic vascular responses required for the metabolic support of auditory adjustment processes. We emphasize the need to distinguish these two states of excitatory/inhibitory imbalance in hearing disorders: (i) Under conditions of preserved fast auditory processing and sustained tonic inhibitory strength, an excitatory/inhibitory imbalance following auditory deprivation can maintain precise hearing through a memory linked, transient disinhibition that leads to enhanced spiking fidelity (central neural gain⇑) (ii) Under conditions of critically diminished fast auditory processing and reduced tonic inhibitory strength, hyperexcitability can be part of an increased synchronization over a broader frequency range, linked to reduced spiking reliability (central neural gain⇓). This latter stage mutually reinforces diminished metabolic support for auditory adjustment processes, increasing the risks for canonical dementia syndromes.
Collapse
Affiliation(s)
- Marlies Knipper
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
- *Correspondence: Marlies Knipper,
| | - Wibke Singer
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Kerstin Schwabe
- Experimental Neurosurgery, Department of Neurosurgery, Hannover Medical School, Hanover, Germany
| | - Gisela E. Hagberg
- Department of Biomedical Magnetic Resonance, University Hospital Tübingen (UKT), Tübingen, Germany
- High-Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Yiwen Li Hegner
- MEG Center, University of Tübingen, Tübingen, Germany
- Center of Neurology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Lukas Rüttiger
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Christoph Braun
- MEG Center, University of Tübingen, Tübingen, Germany
- Center of Neurology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Rüdiger Land
- Department of Experimental Otology, Institute for Audioneurotechnology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
23
|
De Silva TM, Sobey CG. Cerebral Vascular Biology in Health and Disease. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00001-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
24
|
Youwakim J, Girouard H. Inflammation: A Mediator Between Hypertension and Neurodegenerative Diseases. Am J Hypertens 2021; 34:1014-1030. [PMID: 34136907 DOI: 10.1093/ajh/hpab094] [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: 10/15/2020] [Revised: 05/03/2021] [Accepted: 06/15/2021] [Indexed: 12/14/2022] Open
Abstract
Hypertension is the most prevalent and modifiable risk factor for stroke, vascular cognitive impairment, and Alzheimer's disease. However, the mechanistic link between hypertension and neurodegenerative diseases remains to be understood. Recent evidence indicates that inflammation is a common pathophysiological trait for both hypertension and neurodegenerative diseases. Low-grade chronic inflammation at the systemic and central nervous system levels is now recognized to contribute to the physiopathology of hypertension. This review speculates that inflammation represents a mediator between hypertension and neurodegenerative diseases, either by a decrease in cerebral blood flow or a disruption of the blood-brain barrier which will, in turn, let inflammatory cells and neurotoxic molecules enter the brain parenchyma. This may impact brain functions including cognition and contribute to neurodegenerative diseases. This review will thus discuss the relationship between hypertension, systemic inflammation, cerebrovascular functions, neuroinflammation, and brain dysfunctions. The potential clinical future of immunotherapies against hypertension and associated cerebrovascular risks will also be presented.
Collapse
Affiliation(s)
- Jessica Youwakim
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
- Centre interdisciplinaire de recherche sur le cerveau et l’apprentissage (CIRCA); Montreal, QC, Canada
- Groupe de Recherche sur le Système Nerveux Central, Montreal, QC, Canada
| | - Hélène Girouard
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
- Centre interdisciplinaire de recherche sur le cerveau et l’apprentissage (CIRCA); Montreal, QC, Canada
- Groupe de Recherche sur le Système Nerveux Central, Montreal, QC, Canada
- Centre de recherche de l’Institut Universitaire de Gériaterie de Montréal, Montreal, QC, Canada
| |
Collapse
|
25
|
Virtuoso A, Colangelo AM, Maggio N, Fennig U, Weinberg N, Papa M, De Luca C. The Spatiotemporal Coupling: Regional Energy Failure and Aberrant Proteins in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:11304. [PMID: 34768733 PMCID: PMC8583302 DOI: 10.3390/ijms222111304] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/15/2021] [Accepted: 10/17/2021] [Indexed: 12/14/2022] Open
Abstract
The spatial and temporal coordination of each element is a pivotal characteristic of systems, and the central nervous system (CNS) is not an exception. Glial elements and the vascular interface have been considered more recently, together with the extracellular matrix and the immune system. However, the knowledge of the single-element configuration is not sufficient to predict physiological or pathological long-lasting changes. Ionic currents, complex molecular cascades, genomic rearrangement, and the regional energy demand can be different even in neighboring cells of the same phenotype, and their differential expression could explain the region-specific progression of the most studied neurodegenerative diseases. We here reviewed the main nodes and edges of the system, which could be studied to develop a comprehensive knowledge of CNS plasticity from the neurovascular unit to the synaptic cleft. The future goal is to redefine the modeling of synaptic plasticity and achieve a better understanding of neurological diseases, pointing out cellular, subcellular, and molecular components that couple in specific neuroanatomical and functional regions.
Collapse
Affiliation(s)
- Assunta Virtuoso
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (C.D.L.)
| | - Anna Maria Colangelo
- SYSBIO Centre of Systems Biology ISBE-IT, University of Milano-Bicocca, 20126 Milan, Italy;
- Laboratory of Neuroscience “R. Levi-Montalcini”, Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Nicola Maggio
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel; (N.M.); (U.F.); (N.W.)
- Department of Neurology, The Chaim Sheba Medical Center at Tel HaShomer, Ramat Gan 52662, Israel
| | - Uri Fennig
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel; (N.M.); (U.F.); (N.W.)
- Department of Neurology, The Chaim Sheba Medical Center at Tel HaShomer, Ramat Gan 52662, Israel
| | - Nitai Weinberg
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel; (N.M.); (U.F.); (N.W.)
- Department of Neurology, The Chaim Sheba Medical Center at Tel HaShomer, Ramat Gan 52662, Israel
| | - Michele Papa
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (C.D.L.)
- SYSBIO Centre of Systems Biology ISBE-IT, University of Milano-Bicocca, 20126 Milan, Italy;
| | - Ciro De Luca
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (C.D.L.)
| |
Collapse
|
26
|
Ungvari Z, Toth P, Tarantini S, Prodan CI, Sorond F, Merkely B, Csiszar A. Hypertension-induced cognitive impairment: from pathophysiology to public health. Nat Rev Nephrol 2021; 17:639-654. [PMID: 34127835 PMCID: PMC8202227 DOI: 10.1038/s41581-021-00430-6] [Citation(s) in RCA: 222] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
Hypertension affects two-thirds of people aged >60 years and significantly increases the risk of both vascular cognitive impairment and Alzheimer's disease. Hypertension compromises the structural and functional integrity of the cerebral microcirculation, promoting microvascular rarefaction, cerebromicrovascular endothelial dysfunction and neurovascular uncoupling, which impair cerebral blood supply. In addition, hypertension disrupts the blood-brain barrier, promoting neuroinflammation and exacerbation of amyloid pathologies. Ageing is characterized by multifaceted homeostatic dysfunction and impaired cellular stress resilience, which exacerbate the deleterious cerebromicrovascular effects of hypertension. Neuroradiological markers of hypertension-induced cerebral small vessel disease include white matter hyperintensities, lacunar infarcts and microhaemorrhages, all of which are associated with cognitive decline. Use of pharmaceutical and lifestyle interventions that reduce blood pressure, in combination with treatments that promote microvascular health, have the potential to prevent or delay the pathogenesis of vascular cognitive impairment and Alzheimer's disease in patients with hypertension.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Toth
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Calin I Prodan
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Farzaneh Sorond
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bela Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
27
|
Boily M, Li L, Vallerand D, Girouard H. Angiotensin II Disrupts Neurovascular Coupling by Potentiating Calcium Increases in Astrocytic Endfeet. J Am Heart Assoc 2021; 10:e020608. [PMID: 34459216 PMCID: PMC8649296 DOI: 10.1161/jaha.120.020608] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 07/09/2021] [Indexed: 11/16/2022]
Abstract
Background Angiotensin II (Ang II), a critical mediator of hypertension, impairs neurovascular coupling. Since astrocytes are key regulators of neurovascular coupling, we sought to investigate whether Ang II impairs neurovascular coupling through modulation of astrocytic Ca2+ signaling. Methods and Results Using laser Doppler flowmetry, we found that Ang II attenuates cerebral blood flow elevations induced by whisker stimulation or the metabotropic glutamate receptors agonist, 1S, 3R-1-aminocyclopentane-trans-1,3-dicarboxylic acid (P<0.01). In acute brain slices, Ang II shifted the vascular response induced by 1S, 3R-1-aminocyclopentane-trans-1,3-dicarboxylic acid towards vasoconstriction (P<0.05). The resting and 1S, 3R-1-aminocyclopentane-trans-1,3-dicarboxylic acid-induced Ca2+ levels in the astrocytic endfeet were more elevated in the presence of Ang II (P<0.01). Both effects were reversed by the AT1 receptor antagonist, candesartan (P<0.01 for diameter and P<0.05 for calcium levels). Using photolysis of caged Ca2+ in astrocytic endfeet or pre-incubation of 1,2-Bis(2-aminophenoxy)ethane-N,N,N',N'-tetra-acetic acid tetrakis (acetoxymethyl ester), we demonstrated the link between potentiated Ca2+ elevation and impaired vascular response in the presence of Ang II (P<0.001 and P<0.05, respectively). Both intracellular Ca2+ mobilization and Ca2+ influx through transient receptor potential vanilloid 4 mediated Ang II-induced astrocytic Ca2+ elevation, since blockade of these pathways significantly prevented the intracellular Ca2+ in response to 1S, 3R-1-aminocyclopentane-trans-1,3-dicarboxylic acid (P<0.05). Conclusions These results suggest that Ang II through its AT1 receptor potentiates the astrocytic Ca2+ responses to a level that promotes vasoconstriction over vasodilation, thus altering cerebral blood flow increases in response to neuronal activity.
Collapse
Affiliation(s)
- Michaël Boily
- Department of Pharmacology and PhysiologyFaculty of MedicineUniversité de MontréalMontréalQuébecCanada
- Groupe de Recherche sur le Système Nerveux Central (GRSNC)Université de MontréalMontréalQuébecCanada
| | - Lin Li
- Department of Pharmacology and PhysiologyFaculty of MedicineUniversité de MontréalMontréalQuébecCanada
- Groupe de Recherche sur le Système Nerveux Central (GRSNC)Université de MontréalMontréalQuébecCanada
| | - Diane Vallerand
- Department of Pharmacology and PhysiologyFaculty of MedicineUniversité de MontréalMontréalQuébecCanada
- Groupe de Recherche sur le Système Nerveux Central (GRSNC)Université de MontréalMontréalQuébecCanada
- Centre interdisciplinaire de recherche sur le cerveau et l’apprentissage (CIRCA)Université de MontréalMontréalQuébecCanada
| | - Hélène Girouard
- Department of Pharmacology and PhysiologyFaculty of MedicineUniversité de MontréalMontréalQuébecCanada
- Groupe de Recherche sur le Système Nerveux Central (GRSNC)Université de MontréalMontréalQuébecCanada
- Centre interdisciplinaire de recherche sur le cerveau et l’apprentissage (CIRCA)Université de MontréalMontréalQuébecCanada
- Centre de Recherche de l’Institut de Gériatrie de MontréalMontréalQuébecCanada
| |
Collapse
|
28
|
Lazarov O, Minshall RD, Bonini MG. Harnessing neurogenesis in the adult brain-A role in type 2 diabetes mellitus and Alzheimer's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 155:235-269. [PMID: 32854856 DOI: 10.1016/bs.irn.2020.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Some metabolic disorders, such as type 2 diabetes mellitus (T2DM) are risk factors for the development of cognitive deficits and Alzheimer's disease (AD). Epidemiological studies suggest that in people with T2DM, the risk of developing dementia is 2.5 times higher than that in the non-diabetic population. The signaling pathways that underlie the increased risk and facilitate cognitive deficits are not fully understood. In fact, the cause of memory deficits in AD is not fully elucidated. The dentate gyrus of the hippocampus plays an important role in memory formation. Hippocampal neurogenesis is the generation of new neurons and glia in the adult brain throughout life. New neurons incorporate in the granular cell layer of the dentate gyrus and play a role in learning and memory and hippocampal plasticity. A large body of studies suggests that hippocampal neurogenesis is impaired in mouse models of AD and T2DM. Recent evidence shows that hippocampal neurogenesis is also impaired in human patients exhibiting mild cognitive impairment or AD. This review discusses the role of hippocampal neurogenesis in the development of cognitive deficits and AD, and considers inflammatory and endothelial signaling pathways in T2DM that may compromise hippocampal neurogenesis and cognitive function, leading to AD.
Collapse
Affiliation(s)
- Orly Lazarov
- Department of Anatomy and Cell Biology, The University of Illinois at Chicago, Chicago, IL, United States.
| | - Richard D Minshall
- Department of Pharmacology, The University of Illinois at Chicago, Chicago, IL, United States; Department of Anesthesiology, The University of Illinois at Chicago, Chicago, IL, United States
| | - Marcelo G Bonini
- Department of Medicine (Hematology/Oncology), Feinberg School of Medicine of Northwestern University and Basic Sciences Research, Robert H. Lurie Comprehensive Cancer Centre, Chicago, IL, United States
| |
Collapse
|
29
|
García F, Lobos P, Ponce A, Cataldo K, Meza D, Farías P, Estay C, Oyarzun-Ampuero F, Herrera-Molina R, Paula-Lima A, Ardiles ÁO, Hidalgo C, Adasme T, Muñoz P. Astaxanthin Counteracts Excitotoxicity and Reduces the Ensuing Increases in Calcium Levels and Mitochondrial Reactive Oxygen Species Generation. Mar Drugs 2020; 18:md18060335. [PMID: 32604880 PMCID: PMC7345213 DOI: 10.3390/md18060335] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/28/2020] [Accepted: 06/08/2020] [Indexed: 12/17/2022] Open
Abstract
Astaxanthin (ASX) is a carotenoid pigment with strong antioxidant properties. We have reported previously that ASX protects neurons from the noxious effects of amyloid-β peptide oligomers, which promote excessive mitochondrial reactive oxygen species (mROS) production and induce a sustained increase in cytoplasmic Ca2+ concentration. These properties make ASX a promising therapeutic agent against pathological conditions that entail oxidative and Ca2+ dysregulation. Here, we studied whether ASX protects neurons from N-methyl-D-aspartate (NMDA)-induced excitotoxicity, a noxious process which decreases cellular viability, alters gene expression and promotes excessive mROS production. Incubation of the neuronal cell line SH-SY5Y with NMDA decreased cellular viability and increased mitochondrial superoxide production; pre-incubation with ASX prevented these effects. Additionally, incubation of SH-SY5Y cells with ASX effectively reduced the basal mROS production and prevented hydrogen peroxide-induced cell death. In primary hippocampal neurons, transfected with a genetically encoded cytoplasmic Ca2+ sensor, ASX also prevented the increase in intracellular Ca2+ concentration induced by NMDA. We suggest that, by preventing the noxious mROS and Ca2+ increases that occur under excitotoxic conditions, ASX could be useful as a therapeutic agent in neurodegenerative pathologies that involve alterations in Ca2+ homeostasis and ROS generation.
Collapse
Affiliation(s)
- Francisca García
- Laboratory of Cellular and Molecular Plasticity, Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile; (F.G.); (A.P.); (K.C.); (D.M.); (P.F.); (C.E.); (Á.O.A.)
- Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Biomedical Research Center, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Pedro Lobos
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile; (P.L.); (A.P.-L.); (C.H.)
| | - Alejandra Ponce
- Laboratory of Cellular and Molecular Plasticity, Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile; (F.G.); (A.P.); (K.C.); (D.M.); (P.F.); (C.E.); (Á.O.A.)
- Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Biomedical Research Center, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Karla Cataldo
- Laboratory of Cellular and Molecular Plasticity, Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile; (F.G.); (A.P.); (K.C.); (D.M.); (P.F.); (C.E.); (Á.O.A.)
- Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Biomedical Research Center, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Daniela Meza
- Laboratory of Cellular and Molecular Plasticity, Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile; (F.G.); (A.P.); (K.C.); (D.M.); (P.F.); (C.E.); (Á.O.A.)
- Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Biomedical Research Center, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Patricio Farías
- Laboratory of Cellular and Molecular Plasticity, Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile; (F.G.); (A.P.); (K.C.); (D.M.); (P.F.); (C.E.); (Á.O.A.)
- Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Biomedical Research Center, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Carolina Estay
- Laboratory of Cellular and Molecular Plasticity, Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile; (F.G.); (A.P.); (K.C.); (D.M.); (P.F.); (C.E.); (Á.O.A.)
- Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Biomedical Research Center, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Felipe Oyarzun-Ampuero
- Department of Technology and Pharmaceutical Sciences, Faculty of Chemical and Pharmaceutical Sciences, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santos Dumont 964, Independencia, Santiago 8380494, Chile;
| | - Rodrigo Herrera-Molina
- Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany;
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago 8370854, Chile
| | - Andrea Paula-Lima
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile; (P.L.); (A.P.-L.); (C.H.)
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago 8380000, Chile
| | - Álvaro O. Ardiles
- Laboratory of Cellular and Molecular Plasticity, Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile; (F.G.); (A.P.); (K.C.); (D.M.); (P.F.); (C.E.); (Á.O.A.)
- Interdisciplinary Center of Neuroscience of Valparaíso, Universidad de Valparaíso, Valparaíso 2381850, Chile
- Interdisciplinary Center for Health Studies, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Cecilia Hidalgo
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile; (P.L.); (A.P.-L.); (C.H.)
- Department of Neurosciences and Program of Physiology and Biophysics, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile
- Center for Exercise, Metabolism and Cancer Studies, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile
| | - Tatiana Adasme
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago 8370854, Chile
- Correspondence: (T.A.); (P.M.); Tel.: +56-29-786-496 (T.A.); +56-32-250-7368 (P.M.)
| | - Pablo Muñoz
- Laboratory of Cellular and Molecular Plasticity, Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile; (F.G.); (A.P.); (K.C.); (D.M.); (P.F.); (C.E.); (Á.O.A.)
- Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Biomedical Research Center, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Correspondence: (T.A.); (P.M.); Tel.: +56-29-786-496 (T.A.); +56-32-250-7368 (P.M.)
| |
Collapse
|
30
|
Kangussu LM, Marzano LAS, Souza CF, Dantas CC, Miranda AS, Simões e Silva AC. The Renin-Angiotensin System and the Cerebrovascular Diseases: Experimental and Clinical Evidence. Protein Pept Lett 2020; 27:463-475. [DOI: 10.2174/0929866527666191218091823] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/07/2019] [Accepted: 11/04/2019] [Indexed: 12/28/2022]
Abstract
Cerebrovascular Diseases (CVD) comprise a wide spectrum of disorders, all sharing an
acquired or inherited alteration of the cerebral vasculature. CVD have been associated with
important changes in systemic and tissue Renin-Angiotensin System (RAS). The aim of this review
was to summarize and to discuss recent findings related to the modulation of RAS components in
CVD. The role of RAS axes is more extensively studied in experimentally induced stroke. By
means of AT1 receptors in the brain, Ang II hampers cerebral blood flow and causes tissue
ischemia, inflammation, oxidative stress, cell damage and apoptosis. On the other hand, Ang-(1-7)
by stimulating Mas receptor promotes angiogenesis in brain tissue, decreases oxidative stress,
neuroinflammation, and improves cognition, cerebral blood flow, neuronal survival, learning and
memory. In regard to clinical studies, treatment with Angiotensin Converting Enzyme (ACE)
inhibitors and AT1 receptor antagonists exerts preventive and therapeutic effects on stroke. Besides
stroke, studies support a similar role of RAS molecules also in traumatic brain injury and cerebral
aneurysm. The literature supports a beneficial role for the alternative RAS axis in CVD. Further
studies are necessary to investigate the therapeutic potential of ACE2 activators and/or Mas
receptor agonists in patients with CVD.
Collapse
Affiliation(s)
- Lucas M. Kangussu
- Department of Morphology – Biological Science Institute, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lucas Alexandre Santos Marzano
- Interdisciplinary Laboratory of Medical Investigation - Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Cássio Ferraz Souza
- Interdisciplinary Laboratory of Medical Investigation - Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Carolina Couy Dantas
- Interdisciplinary Laboratory of Medical Investigation - Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Aline Silva Miranda
- Interdisciplinary Laboratory of Medical Investigation - Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana Cristina Simões e Silva
- Interdisciplinary Laboratory of Medical Investigation - Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
31
|
Forte M, Stanzione R, Cotugno M, Bianchi F, Marchitti S, Rubattu S. Vascular ageing in hypertension: Focus on mitochondria. Mech Ageing Dev 2020; 189:111267. [PMID: 32473170 DOI: 10.1016/j.mad.2020.111267] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/20/2020] [Accepted: 05/22/2020] [Indexed: 12/25/2022]
Abstract
Hypertension is a common age-related disease, along with vascular and neurodegenerative diseases. Vascular ageing increases during hypertension, but hypertension itself accelerates vascular ageing, thus creating a vicious circle. Vascular stiffening, endothelial dysfunction, impaired contractility and vasorelaxation are the main alterations related to vascular ageing, as a consequence of vascular smooth muscle and endothelial cells senescence. Several molecular mechanisms have been involved into the functional and morphological changes of the aged vessels. Among them, oxidative stress, inflammation, extracellular matrix deregulation and mitochondrial dysfunction are the best characterized. In the present review, we discuss relevant literature about the biology of vascular and cerebrovascular ageing with a particular focus on mitochondria signalling. We underline the therapeutic strategies, able to improve mitochondrial health, which may represent a promising tool to decrease vascular dysfunction associated with ageing and hypertension-related complications.
Collapse
Affiliation(s)
- Maurizio Forte
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy
| | | | - Maria Cotugno
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy
| | - Franca Bianchi
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy
| | | | - Speranza Rubattu
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy; Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy.
| |
Collapse
|
32
|
Aleksandrowicz M, Klapczynska K, Kozniewska E. Dysfunction of the endothelium and constriction of the isolated rat's middle cerebral artery in low sodium environment in the presence of vasopressin. Clin Exp Pharmacol Physiol 2019; 47:759-764. [PMID: 31876005 DOI: 10.1111/1440-1681.13242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 01/20/2023]
Abstract
Hyponatraemia, a water-electrolyte disorder diagnosed in patients with subarachnoid haemorrhage (SAH), increases a risk of persistent vasospasm. In majority of cases, hyponatraemia results from inappropriate secretion of vasopressin (AVP). The effect of AVP-associated hyponatraemia on cerebral vasculature is unknown. The present study aimed to elucidate the role of AVP in the response of the middle cerebral artery (MCA) of the rat to hyponatraemia. Isolated, cannulated, and pressurized rat MCAs were perfused/superfused with physiological (Na+ = 144 mmol/L) buffer or low-sodium (Na+ = 121 mmol/L) buffer containing either AVP or angiotensin II (ANG II). ANG II was used to check if the effect of low plasma sodium concentration combined with AVP on the MCA tone is unique to vasopressin. At physiological Na+ concentration, vasopressin (1.4 × 10-11 mol/L) or angiotensin II (10-9 mol/L) resulted in relaxation of the MCA. Substitution of low-sodium for the normal sodium buffer with the same concentration of AVP, resulted in the constriction of the MCA. This effect was absent after removal of the endothelium, administration of vasopressin V1 receptor antagonist or concomitant inhibition of endothelin-1 receptors and synthesis of thromboxane A2. In contrast, no constriction of the MCA in low-sodium buffer was observed when AVP was replaced with ANG II. Our data suggest that presence of vasopressin and low sodium ion concentration results in the change of endothelium phenotype from pro-vasodilatory to pro-vasoconstrictory. This phenomenon may be an overlooked factor contributing to vasospasm in SAH patients with hyponatraemia caused by inappropriate antidiuretic hormone secretion (SIADH).
Collapse
Affiliation(s)
- Marta Aleksandrowicz
- Laboratory of Experimental and Clinical Neurosurgery, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Klapczynska
- Faculty of Physical Education and Health Promotion, University of Szczecin, Szczecin, Poland
| | - Ewa Kozniewska
- Laboratory of Experimental and Clinical Neurosurgery, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
33
|
Lerman LO, Kurtz TW, Touyz RM, Ellison DH, Chade AR, Crowley SD, Mattson DL, Mullins JJ, Osborn J, Eirin A, Reckelhoff JF, Iadecola C, Coffman TM. Animal Models of Hypertension: A Scientific Statement From the American Heart Association. Hypertension 2019; 73:e87-e120. [PMID: 30866654 DOI: 10.1161/hyp.0000000000000090] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hypertension is the most common chronic disease in the world, yet the precise cause of elevated blood pressure often cannot be determined. Animal models have been useful for unraveling the pathogenesis of hypertension and for testing novel therapeutic strategies. The utility of animal models for improving the understanding of the pathogenesis, prevention, and treatment of hypertension and its comorbidities depends on their validity for representing human forms of hypertension, including responses to therapy, and on the quality of studies in those models (such as reproducibility and experimental design). Important unmet needs in this field include the development of models that mimic the discrete hypertensive syndromes that now populate the clinic, resolution of ongoing controversies in the pathogenesis of hypertension, and the development of new avenues for preventing and treating hypertension and its complications. Animal models may indeed be useful for addressing these unmet needs.
Collapse
|
34
|
Hachinski V, Einhäupl K, Ganten D, Alladi S, Brayne C, Stephan BCM, Sweeney MD, Zlokovic B, Iturria-Medina Y, Iadecola C, Nishimura N, Schaffer CB, Whitehead SN, Black SE, Østergaard L, Wardlaw J, Greenberg S, Friberg L, Norrving B, Rowe B, Joanette Y, Hacke W, Kuller L, Dichgans M, Endres M, Khachaturian ZS. Special topic section: linkages among cerebrovascular, cardiovascular, and cognitive disorders: Preventing dementia by preventing stroke: The Berlin Manifesto. Int J Stroke 2019:1747493019871915. [PMID: 31543058 DOI: 10.1177/1747493019871915] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
The incidence of stroke and dementia are diverging across the world, rising for those in low-and middle-income countries and falling in those in high-income countries. This suggests that whatever factors cause these trends are potentially modifiable. At the population level, neurological disorders as a group account for the largest proportion of disability-adjusted life years globally (10%). Among neurological disorders, stroke (42%) and dementia (10%) dominate. Stroke and dementia confer risks for each other and share some of the same, largely modifiable, risk and protective factors. In principle, 90% of strokes and 35% of dementias have been estimated to be preventable. Because a stroke doubles the chance of developing dementia and stroke is more common than dementia, more than a third of dementias could be prevented by preventing stroke. Developments at the pathological, pathophysiological, and clinical level also point to new directions. Growing understanding of brain pathophysiology has unveiled the reciprocal interaction of cerebrovascular disease and neurodegeneration identifying new therapeutic targets to include protection of the endothelium, the blood-brain barrier, and other components of the neurovascular unit. In addition, targeting amyloid angiopathy aspects of inflammation and genetic manipulation hold new testable promise. In the meantime, accumulating evidence suggests that whole populations experiencing improved education, and lower vascular risk factor profiles (e.g., reduced prevalence of smoking) and vascular disease, including stroke, have better cognitive function and lower dementia rates. At the individual levels, trials have demonstrated that anticoagulation of atrial fibrillation can reduce the risk of dementia by 48% and that systolic blood pressure lower than 140 mmHg may be better for the brain. Based on these considerations, the World Stroke Organization has issued a proclamation, endorsed by all the major international organizations focused on global brain and cardiovascular health, calling for the joint prevention of stroke and dementia. This article summarizes the evidence for translation into action. © 2019 the Alzheimer's Association and the World Stroke Organisation. Published by Elsevier Inc. All rights reserved.
Collapse
Affiliation(s)
- Vladimir Hachinski
- Department of Clinical Neurological Sciences, Western University, Ontario, Canada
| | - Karl Einhäupl
- Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Detlev Ganten
- Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Suvarna Alladi
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Carol Brayne
- Department of Public Health and Primary Care in the University of Cambridge, Cambridge, UK
| | - Blossom C M Stephan
- Institute of Mental Health, Division of Psychiatry and Applied Psychology, School of Medicine, University of Nottingham, Nottingham, UK
| | - Melanie D Sweeney
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Berislav Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yasser Iturria-Medina
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Nozomi Nishimura
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Chris B Schaffer
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Shawn N Whitehead
- Department of Anatomy and Cell Biology, Western University, Ontario, Canada
| | - Sandra E Black
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Leif Østergaard
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
- Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark
| | - Joanna Wardlaw
- Centre for Clinical Brain Sciences, Edinburgh Imaging, UK Dementia Research Institute, University of Edinburgh, Scotland, UK
| | - Steven Greenberg
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Leif Friberg
- Department of Clinical Sciences, Karolinska Institute, Stockholm, Sweden
| | - Bo Norrving
- Department of Clinical Sciences, Neurology, Lund University, Lund, Sweden
| | - Brian Rowe
- Department of Emergency Medicine and School of Public Health, University of Alberta, Edmonton, Alberta, Canada
| | - Yves Joanette
- Canadian Institute of Health and Research, Ottawa, Canada
| | - Werner Hacke
- Department of Neurology, Heidelberg University, Heidelberg, Germany
| | - Lewis Kuller
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-Universität LMU, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
| | - Matthias Endres
- Department of Neurology with Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- ExcellenceCluster NeuroCure, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), partner site Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | | |
Collapse
|
35
|
Hachinski V, Einhäupl K, Ganten D, Alladi S, Brayne C, Stephan BCM, Sweeney MD, Zlokovic B, Iturria-Medina Y, Iadecola C, Nishimura N, Schaffer CB, Whitehead SN, Black SE, Østergaard L, Wardlaw J, Greenberg S, Friberg L, Norrving B, Rowe B, Joanette Y, Hacke W, Kuller L, Dichgans M, Endres M, Khachaturian ZS. Preventing dementia by preventing stroke: The Berlin Manifesto. Alzheimers Dement 2019; 15:961-984. [PMID: 31327392 PMCID: PMC7001744 DOI: 10.1016/j.jalz.2019.06.001] [Citation(s) in RCA: 198] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The incidence of stroke and dementia are diverging across the world, rising for those in low- and middle-income countries and falling in those in high-income countries. This suggests that whatever factors cause these trends are potentially modifiable. At the population level, neurological disorders as a group account for the largest proportion of disability-adjusted life years globally (10%). Among neurological disorders, stroke (42%) and dementia (10%) dominate. Stroke and dementia confer risks for each other and share some of the same, largely modifiable, risk and protective factors. In principle, 90% of strokes and 35% of dementias have been estimated to be preventable. Because a stroke doubles the chance of developing dementia and stroke is more common than dementia, more than a third of dementias could be prevented by preventing stroke. Developments at the pathological, pathophysiological, and clinical level also point to new directions. Growing understanding of brain pathophysiology has unveiled the reciprocal interaction of cerebrovascular disease and neurodegeneration identifying new therapeutic targets to include protection of the endothelium, the blood-brain barrier, and other components of the neurovascular unit. In addition, targeting amyloid angiopathy aspects of inflammation and genetic manipulation hold new testable promise. In the meantime, accumulating evidence suggests that whole populations experiencing improved education, and lower vascular risk factor profiles (e.g., reduced prevalence of smoking) and vascular disease, including stroke, have better cognitive function and lower dementia rates. At the individual levels, trials have demonstrated that anticoagulation of atrial fibrillation can reduce the risk of dementia by 48% and that systolic blood pressure lower than 140 mmHg may be better for the brain. Based on these considerations, the World Stroke Organization has issued a proclamation, endorsed by all the major international organizations focused on global brain and cardiovascular health, calling for the joint prevention of stroke and dementia. This article summarizes the evidence for translation into action.
Collapse
Affiliation(s)
- Vladimir Hachinski
- Department of Clinical Neurological Sciences, Western University, Ontario, Canada.
| | - Karl Einhäupl
- Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Detlev Ganten
- Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Suvarna Alladi
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Carol Brayne
- Department of Public Health and Primary Care in the University of Cambridge, Cambridge, UK
| | - Blossom C M Stephan
- Institute of Mental Health, Division of Psychiatry and Applied Psychology, School of Medicine, University of Nottingham, Nottingham, UK
| | - Melanie D Sweeney
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Berislav Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yasser Iturria-Medina
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Nozomi Nishimura
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Chris B Schaffer
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Shawn N Whitehead
- Department of Anatomy and Cell Biology, Western University, Ontario, Canada
| | - Sandra E Black
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Leif Østergaard
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark; Department of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark
| | - Joanna Wardlaw
- Centre for Clinical Brain Sciences, Edinburgh Imaging, UK Dementia Research Institute, University of Edinburgh, Scotland, UK
| | - Steven Greenberg
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Leif Friberg
- Department of Clinical Sciences, Karolinska Institute, Stockholm, Sweden
| | - Bo Norrving
- Department of Clinical Sciences, Neurology, Lund University, Lund, Sweden
| | - Brian Rowe
- Department of Emergency Medicine and School of Public Health, University of Alberta, Edmonton, Alberta, Canada
| | - Yves Joanette
- Canadian Institute of Health and Research, Ottawa, Canada
| | - Werner Hacke
- Department of Neurology, Heidelberg University, Heidelberg, Germany
| | - Lewis Kuller
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-Universität LMU, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
| | - Matthias Endres
- Department of Neurology with Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany; Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany; ExcellenceCluster NeuroCure, Charité-Universitätsmedizin Berlin, Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE), partner site Berlin, Berlin, Germany; German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany
| | | |
Collapse
|
36
|
Abstract
Hypertension has emerged as a leading cause of age-related cognitive impairment. Long known to be associated with dementia caused by vascular factors, hypertension has more recently been linked also to Alzheimer disease-the major cause of dementia in older people. Thus, although midlife hypertension is a risk factor for late-life dementia, hypertension may also promote the neurodegenerative pathology underlying Alzheimer disease. The mechanistic bases of these harmful effects remain to be established. Hypertension is well known to alter in the structure and function of cerebral blood vessels, but how these cerebrovascular effects lead to cognitive impairment and promote Alzheimer disease pathology is not well understood. Furthermore, critical questions also concern whether treatment of hypertension prevents cognitive impairment, the blood pressure threshold for treatment, and the antihypertensive agents to be used. Recent advances in neurovascular biology, epidemiology, brain imaging, and biomarker development have started to provide new insights into these critical issues. In this review, we will examine the progress made to date, and, after a critical evaluation of the evidence, we will highlight questions still outstanding and seek to provide a path forward for future studies.
Collapse
Affiliation(s)
- Costantino Iadecola
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York (C.I.)
| | - Rebecca F Gottesman
- Departments of Neurology (R.F.G.), Johns Hopkins University, Baltimore, MD
- Epidemiology (R.F.G.), Johns Hopkins University, Baltimore, MD
| |
Collapse
|
37
|
Diaz JR, Kim KJ, Brands MW, Filosa JA. Augmented astrocyte microdomain Ca 2+ dynamics and parenchymal arteriole tone in angiotensin II-infused hypertensive mice. Glia 2018; 67:551-565. [PMID: 30506941 DOI: 10.1002/glia.23564] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 10/22/2018] [Accepted: 10/22/2018] [Indexed: 11/09/2022]
Abstract
Hypertension is an important contributor to cognitive decline but the underlying mechanisms are unknown. Although much focus has been placed on the effect of hypertension on vascular function, less is understood of its effects on nonvascular cells. Because astrocytes and parenchymal arterioles (PA) form a functional unit (neurovascular unit), we tested the hypothesis that hypertension-induced changes in PA tone concomitantly increases astrocyte Ca2+ . We used cortical brain slices from 8-week-old mice to measure myogenic responses from pressurized and perfused PA. Chronic hypertension was induced in mice by 28-day angiotensin II (Ang II) infusion; PA resting tone and myogenic responses increased significantly. In addition, chronic hypertension significantly increased spontaneous Ca2+ events within astrocyte microdomains (MD). Similarly, a significant increase in astrocyte Ca2+ was observed during PA myogenic responses supporting enhanced vessel-to-astrocyte signaling. The transient potential receptor vanilloid 4 (TRPV4) channel, expressed in astrocyte processes in contact with blood vessels, namely endfeet, respond to hemodynamic stimuli such as increased pressure/flow. Supporting a role for TRPV4 channels in aberrant astrocyte Ca2+ dynamics in hypertension, cortical astrocytes from hypertensive mice showed augmented TRPV4 channel expression, currents and Ca2+ responses to the selective channel agonist GSK1016790A. In addition, pharmacological TRPV4 channel blockade or genetic deletion abrogated enhanced hypertension-induced increases in PA tone. Together, these data suggest chronic hypertension increases PA tone and Ca2+ events within astrocytes MD. We conclude that aberrant Ca2+ events in astrocyte constitute an early event toward the progression of cognitive decline.
Collapse
Affiliation(s)
| | - Ki Jung Kim
- Department of Physiology, Augusta University, Augusta, Georgia
| | | | | |
Collapse
|
38
|
Santisteban MM, Iadecola C. Hypertension, dietary salt and cognitive impairment. J Cereb Blood Flow Metab 2018; 38:2112-2128. [PMID: 30295560 PMCID: PMC6282225 DOI: 10.1177/0271678x18803374] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/31/2018] [Indexed: 12/12/2022]
Abstract
Dementia is growing at an alarming rate worldwide. Although Alzheimer disease is the leading cause, over 50% of individuals diagnosed with Alzheimer disease have vascular lesions at autopsy. There has been an increasing appreciation of the pathogenic role of vascular risk factors in cognitive impairment caused by neurodegeneration. Midlife hypertension is a leading risk factor for late-life dementia. Hypertension alters cerebrovascular structure, impairs the major factors regulating the cerebral microcirculation, and promotes Alzheimer pathology. Experimental studies have identified brain perivascular macrophages as the major free radical source mediating neurovascular dysfunction of hypertension. Recent evidence indicates that high dietary salt may also induce cognitive impairment. Contrary to previous belief, the effect is not necessarily associated with hypertension and is mediated by a deficit in endothelial nitric oxide. Collectively, the evidence suggests a remarkable cellular diversity of the impact of vascular risk factors on the cerebral vasculature and cognition. Whereas long-term longitudinal epidemiological studies are needed to resolve the temporal relationships between vascular risk factors and cognitive dysfunction, single-cell molecular studies of the vasculature in animal models will provide a fuller mechanistic understanding. This knowledge is critical for developing new preventive, diagnostic, and therapeutic approaches for these devastating diseases of the mind.
Collapse
Affiliation(s)
- Monica M Santisteban
- Feil Family Brain and Mind Research Institute Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
39
|
Wang F, Cao Y, Ma L, Pei H, Rausch WD, Li H. Dysfunction of Cerebrovascular Endothelial Cells: Prelude to Vascular Dementia. Front Aging Neurosci 2018; 10:376. [PMID: 30505270 PMCID: PMC6250852 DOI: 10.3389/fnagi.2018.00376] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/29/2018] [Indexed: 12/19/2022] Open
Abstract
Vascular dementia (VaD) is the second most common type of dementia after Alzheimer's disease (AD), characterized by progressive cognitive impairment, memory loss, and thinking or speech problems. VaD is usually caused by cerebrovascular disease, during which, cerebrovascular endothelial cells (CECs) are vulnerable. CEC dysfunction occurs before the onset of VaD and can eventually lead to dysregulation of cerebral blood flow and blood-brain barrier damage, followed by the activation of glia and inflammatory environment in the brain. White matter, neuronal axons, and synapses are compromised in this process, leading to cognitive impairment. The present review summarizes the mechanisms underlying CEC impairment during hypoperfusion and pathological role of CECs in VaD. Through the comprehensive examination and summarization, endothelial nitric oxide synthase (eNOS)/nitric oxide (NO) signaling pathway, Ras homolog gene family member A (RhoA) signaling pathway, and CEC-derived caveolin-1 (CAV-1) are proposed to serve as targets of new drugs for the treatment of VaD.
Collapse
Affiliation(s)
- Feixue Wang
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Cao
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Lina Ma
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Hui Pei
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Wolf Dieter Rausch
- Department for Biomedical Sciences, Institute of Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Hao Li
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
40
|
Koizumi K, Hattori Y, Ahn SJ, Buendia I, Ciacciarelli A, Uekawa K, Wang G, Hiller A, Zhao L, Voss HU, Paul SM, Schaffer C, Park L, Iadecola C. Apoε4 disrupts neurovascular regulation and undermines white matter integrity and cognitive function. Nat Commun 2018; 9:3816. [PMID: 30232327 PMCID: PMC6145902 DOI: 10.1038/s41467-018-06301-2] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022] Open
Abstract
The ApoE4 allele is associated with increased risk of small vessel disease, which is a cause of vascular cognitive impairment. Here, we report that mice with targeted replacement (TR) of the ApoE gene with human ApoE4 have reduced neocortical cerebral blood flow compared to ApoE3-TR mice, an effect due to reduced vascular density rather than slowing of microvascular red blood cell flow. Furthermore, homeostatic mechanisms matching local delivery of blood flow to brain activity are impaired in ApoE4-TR mice. In a model of cerebral hypoperfusion, these cerebrovascular alterations exacerbate damage to the white matter of the corpus callosum and worsen cognitive dysfunction. Using 3-photon microscopy we found that the increased white matter damage is linked to an enhanced reduction of microvascular flow resulting in local hypoxia. Such alterations may be responsible for the increased susceptibility to hypoxic-ischemic lesions in the subcortical white matter of individuals carrying the ApoE4 allele. ApoE4 is a risk factor for small vessel disease, which can lead to cognitive impairment. Here the authors assess the microvasculature of the corpus callosum using 3-photon microscopy and find that mice expressing the ApoE4 allele are more susceptible than wild-type to white matter injury and cognitive impairment in a model of hypoperfusion-induced hypoxia.
Collapse
Affiliation(s)
- Kenzo Koizumi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Yorito Hattori
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Sung Ji Ahn
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Izaskun Buendia
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Antonio Ciacciarelli
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Ken Uekawa
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Gang Wang
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Abigail Hiller
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Lingzhi Zhao
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Henning U Voss
- Department of Radiology, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Steven M Paul
- Department of Neurology and Psychiatry, Washington University in St. Louis, St. Louis, 63110, MO, USA
| | - Chris Schaffer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA.,Meinig School of Biomedical Engineering, Cornell University, Ithaca, 14853, NY, USA
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA.
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA.
| |
Collapse
|
41
|
Carvalho C, Moreira PI. Oxidative Stress: A Major Player in Cerebrovascular Alterations Associated to Neurodegenerative Events. Front Physiol 2018; 9:806. [PMID: 30018565 PMCID: PMC6037979 DOI: 10.3389/fphys.2018.00806] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/08/2018] [Indexed: 12/19/2022] Open
Abstract
The brain is one of the most exquisite organs in the body with high metabolic demands, and requires a tight regulation of the surrounding environment. This tight control is exerted by the neurovascular unit (NVU) comprising different cell types, where endothelial cells play the commander-in-chief role. Thus, it is assumable that even slight perturbations in NVU might affect, in some cases irreversibly, brain homeostasis and health. In this line, recent findings support the two-hit vascular hypothesis for neurodegenerative conditions, where vascular dysfunction underlies the development of neurodegenerative diseases, such as Alzheimer’s disease (AD). Knowing that endothelial cells are rich in mitochondria and nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, two major reactive oxygen species (ROS) sources, this review aims to gather information on how oxidative stress is in the front line of vascular alterations observed in brain aging and neurodegenerative conditions, particularly AD. Also, a brief discussion about the therapeutic strategies aimed to protect against cerebrovascular diseases is included.
Collapse
Affiliation(s)
- Cristina Carvalho
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Paula I Moreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Laboratory of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
42
|
Thorin-Trescases N, de Montgolfier O, Pinçon A, Raignault A, Caland L, Labbé P, Thorin E. Impact of pulse pressure on cerebrovascular events leading to age-related cognitive decline. Am J Physiol Heart Circ Physiol 2018; 314:H1214-H1224. [PMID: 29451817 DOI: 10.1152/ajpheart.00637.2017] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Aging is a modern concept: human life expectancy has more than doubled in less than 150 yr in Western countries. Longer life span, however, reveals age-related diseases, including cerebrovascular diseases. The vascular system is a prime target of aging: the "wear and tear" of large elastic arteries exposed to a lifelong pulsatile pressure causes arterial stiffening by fragmentation of elastin fibers and replacement by stiffer collagen. This arterial stiffening increases in return the amplitude of the pulse pressure (PP), its wave penetrating deeper into the microcirculation of low-resistance, high-flow organs such as the brain. Several studies have associated peripheral arterial stiffness responsible for the sustained increase in PP, with brain microvascular diseases such as cerebral small vessel disease, cortical gray matter thinning, white matter atrophy, and cognitive dysfunction in older individuals and prematurely in hypertensive and diabetic patients. The rarefaction of white matter is also associated with middle cerebral artery pulsatility that is strongly dependent on PP and artery stiffness. PP and brain damage are likely associated, but the sequence of mechanistic events has not been established. Elevated PP promotes endothelial dysfunction that may slowly develop in parallel with the accumulation of proinflammatory senescent cells and oxidative stress, generating cerebrovascular damage and remodeling, as well as brain structural changes. Here, we review data suggesting that age-related increased peripheral artery stiffness may promote the penetration of a high PP to cerebral microvessels, likely causing functional, structural, metabolic, and hemodynamic alterations that could ultimately promote neuronal dysfunction and cognitive decline.
Collapse
Affiliation(s)
| | - Olivia de Montgolfier
- Montreal Heart Institute, Research Center , Montreal, Quebec , Canada.,Department of Pharmacology, Faculty of Medicine, Université de Montréal , Montreal, Quebec , Canada
| | - Anthony Pinçon
- Montreal Heart Institute, Research Center , Montreal, Quebec , Canada.,Department of Pharmacology, Faculty of Medicine, Université de Montréal , Montreal, Quebec , Canada
| | - Adeline Raignault
- Montreal Heart Institute, Research Center , Montreal, Quebec , Canada
| | - Laurie Caland
- Montreal Heart Institute, Research Center , Montreal, Quebec , Canada.,Department of Pharmacology, Faculty of Medicine, Université de Montréal , Montreal, Quebec , Canada
| | - Pauline Labbé
- Montreal Heart Institute, Research Center , Montreal, Quebec , Canada.,Department of Pharmacology, Faculty of Medicine, Université de Montréal , Montreal, Quebec , Canada
| | - Eric Thorin
- Montreal Heart Institute, Research Center , Montreal, Quebec , Canada.,Department of Pharmacology, Faculty of Medicine, Université de Montréal , Montreal, Quebec , Canada.,Department of Surgery, Faculty of Medicine, Université de Montréal , Montreal, Quebec , Canada
| |
Collapse
|
43
|
Grochowski C, Litak J, Kamieniak P, Maciejewski R. Oxidative stress in cerebral small vessel disease. Role of reactive species. Free Radic Res 2017; 52:1-13. [PMID: 29166803 DOI: 10.1080/10715762.2017.1402304] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Cerebral small vessel disease (CSVD) is a wide term describing the condition affecting perforating arterial branches as well as arterioles, venules, and capillaries. Cerebral vascular net is one of the main targets of localised oxidative stress processes causing damage to vasculature, changes in the blood flow and blood-brain barrier and, in consequence, promoting neurodegenerative alterations in the brain tissue. Numerous studies report the fact of oxidation to proteins, sugars, lipids and nucleic acids, occurring in most neurodegenerative diseases mainly in the earliest stages and correlations with the development of cognitive and motor disturbances. The dysfunction of endothelium can be caused by oxidative stress and inflammatory mechanisms as a result of reactions and processes generating extensive reactive oxygen species (ROS) production such as high blood pressure, oxidised low density lipoproteins (oxLDL), very low density lipoproteins (vLDL), diabetes, homocysteinaemia, smoking, and infections. Several animal studies show positive aspects of ROS, especially within cerebral vasculature.
Collapse
Affiliation(s)
- Cezary Grochowski
- a Department of Neurosurgery and Pediatric Neurosurgery , Medical University of Lublin , Lublin , Poland.,b Department of Human Anatomy , Medical University of Lublin , Lublin , Poland
| | - Jakub Litak
- a Department of Neurosurgery and Pediatric Neurosurgery , Medical University of Lublin , Lublin , Poland
| | - Piotr Kamieniak
- a Department of Neurosurgery and Pediatric Neurosurgery , Medical University of Lublin , Lublin , Poland
| | - Ryszard Maciejewski
- b Department of Human Anatomy , Medical University of Lublin , Lublin , Poland
| |
Collapse
|
44
|
The Neurovascular Unit Coming of Age: A Journey through Neurovascular Coupling in Health and Disease. Neuron 2017; 96:17-42. [PMID: 28957666 DOI: 10.1016/j.neuron.2017.07.030] [Citation(s) in RCA: 1391] [Impact Index Per Article: 173.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/20/2017] [Accepted: 07/25/2017] [Indexed: 02/07/2023]
Abstract
The concept of the neurovascular unit (NVU), formalized at the 2001 Stroke Progress Review Group meeting of the National Institute of Neurological Disorders and Stroke, emphasizes the intimate relationship between the brain and its vessels. Since then, the NVU has attracted the interest of the neuroscience community, resulting in considerable advances in the field. Here the current state of knowledge of the NVU will be assessed, focusing on one of its most vital roles: the coupling between neural activity and blood flow. The evidence supports a conceptual shift in the mechanisms of neurovascular coupling, from a unidimensional process involving neuronal-astrocytic signaling to local blood vessels to a multidimensional one in which mediators released from multiple cells engage distinct signaling pathways and effector systems across the entire cerebrovascular network in a highly orchestrated manner. The recently appreciated NVU dysfunction in neurodegenerative diseases, although still poorly understood, supports emerging concepts that maintaining neurovascular health promotes brain health.
Collapse
|
45
|
Csiszar A, Tarantini S, Fülöp GA, Kiss T, Valcarcel-Ares MN, Galvan V, Ungvari Z, Yabluchanskiy A. Hypertension impairs neurovascular coupling and promotes microvascular injury: role in exacerbation of Alzheimer's disease. GeroScience 2017; 39:359-372. [PMID: 28853030 PMCID: PMC5636770 DOI: 10.1007/s11357-017-9991-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/26/2017] [Indexed: 12/13/2022] Open
Abstract
Hypertension in the elderly substantially increases both the risk of vascular cognitive impairment (VCI) and Alzheimer's disease (AD); however, the underlying mechanisms are not completely understood. This review discusses the effects of hypertension on structural and functional integrity of cerebral microcirculation, including hypertension-induced alterations in neurovascular coupling responses, cellular and molecular mechanisms involved in microvascular damage (capillary rarefaction, blood-brain barrier disruption), and the genesis of cerebral microhemorrhages and their potential role in exacerbation of cognitive decline associated with AD. Understanding and targeting the hypertension-induced cerebromicrovascular alterations that are involved in the onset and progression of AD and contribute to cognitive impairment are expected to have a major role in preserving brain health in high-risk older individuals.
Collapse
Affiliation(s)
- Anna Csiszar
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Gábor A Fülöp
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamas Kiss
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - M Noa Valcarcel-Ares
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Veronica Galvan
- Department of Cellular and Integrative Physiology, Barshop Institute for Longevity and Aging Studies University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
46
|
Hypertension-induced synapse loss and impairment in synaptic plasticity in the mouse hippocampus mimics the aging phenotype: implications for the pathogenesis of vascular cognitive impairment. GeroScience 2017; 39:385-406. [PMID: 28664509 DOI: 10.1007/s11357-017-9981-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 05/30/2017] [Indexed: 12/22/2022] Open
Abstract
Strong epidemiological and experimental evidence indicates that hypertension has detrimental effects on the cerebral microcirculation and thereby promotes accelerated brain aging. Hypertension is an independent risk factor for both vascular cognitive impairment (VCI) and Alzheimer's disease (AD). However, the pathophysiological link between hypertension-induced cerebromicrovascular injury (e.g., blood-brain barrier disruption, increased microvascular oxidative stress, and inflammation) and cognitive decline remains elusive. The present study was designed to characterize neuronal functional and morphological alterations induced by chronic hypertension and compare them to those induced by aging. To achieve that goal, we induced hypertension in young C57BL/6 mice by chronic (4 weeks) infusion of angiotensin II. We found that long-term potentiation (LTP) of performant path synapses following high-frequency stimulation of afferent fibers was decreased in hippocampal slices obtained from hypertensive mice, mimicking the aging phenotype. Hypertension and advanced age were associated with comparable decline in synaptic density in the stratum radiatum of the mouse hippocampus. Hypertension, similar to aging, was associated with changes in mRNA expression of several genes involved in regulation of neuronal function, including down-regulation of Bdnf, Homer1, and Dlg4, which may have a role in impaired synaptic plasticity. Collectively, hypertension impairs synaptic plasticity, reduces synaptic density, and promotes dysregulation of genes involved in synaptic function in the mouse hippocampus mimicking the aging phenotype. These hypertension-induced neuronal alterations may impair establishment of memories in the hippocampus and contribute to the pathogenesis and clinical manifestation of both vascular cognitive impairment (VCI) and Alzheimer's disease (AD).
Collapse
|
47
|
Rodriguez-Grande B, Konsman JP. Gas Diffusion in the CNS. J Neurosci Res 2017; 96:207-218. [PMID: 28504343 DOI: 10.1002/jnr.24077] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/23/2017] [Accepted: 04/10/2017] [Indexed: 12/21/2022]
Abstract
Gases have been long known to have essential physiological functions in the CNS such as respiration or regulation of vascular tone. Since gases have been classically considered to freely diffuse, research in gas biology has so far focused on mechanisms of gas synthesis and gas reactivity, rather than gas diffusion and transport. However, the discovery of gas pores during the last two decades and the characterization of diverse diffusion patterns through different membranes has raised the possibility that modulation of gas diffusion is also a physiologically relevant parameter. Here we review the means of gas movement into and within the brain through "free" diffusion and gas pores, notably aquaporins, discussing the role that gas diffusion may play in the modulation of gas function. We highlight how diffusion is relevant to neuronal signaling, volume transmission, and cerebrovascular control in the case of NO, one of the most extensively studied gases. We point out how facilitated transport can be especially relevant for gases with low permeability in lipid membranes like NH3 and discuss the possible implications of NH3 -permeable channels in physiology and hyperammonemic encephalopathy. We identify novel research questions about how modulation of gas diffusion could intervene in CNS pathologies. This emerging area of research can provide novel and interesting insights in the field of gas biology.
Collapse
|
48
|
COPD and stroke: are systemic inflammation and oxidative stress the missing links? Clin Sci (Lond) 2017; 130:1039-50. [PMID: 27215677 PMCID: PMC4876483 DOI: 10.1042/cs20160043] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 03/07/2016] [Indexed: 12/12/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by progressive airflow limitation and loss of lung function, and is currently the third largest cause of death in the world. It is now well established that cardiovascular-related comorbidities such as stroke contribute to morbidity and mortality in COPD. The mechanisms linking COPD and stroke remain to be fully defined but are likely to be interconnected. The association between COPD and stroke may be largely dependent on shared risk factors such as aging and smoking, or the association of COPD with traditional stroke risk factors. In addition, we propose that COPD-related systemic inflammation and oxidative stress may play important roles by promoting cerebral vascular dysfunction and platelet hyperactivity. In this review, we briefly discuss the pathogenesis of COPD, acute exacerbations of COPD (AECOPD) and cardiovascular comorbidities associated with COPD, in particular stroke. We also highlight and discuss the potential mechanisms underpinning the link between COPD and stroke, with a particular focus on the roles of systemic inflammation and oxidative stress.
Collapse
|
49
|
Ungvari Z, Tarantini S, Hertelendy P, Valcarcel-Ares MN, Fülöp GA, Logan S, Kiss T, Farkas E, Csiszar A, Yabluchanskiy A. Cerebromicrovascular dysfunction predicts cognitive decline and gait abnormalities in a mouse model of whole brain irradiation-induced accelerated brain senescence. GeroScience 2017; 39:33-42. [PMID: 28299642 DOI: 10.1007/s11357-017-9964-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/25/2017] [Indexed: 01/02/2023] Open
Abstract
Whole brain irradiation (WBI) is a mainstream therapy for patients with both identifiable brain metastases and prophylaxis for microscopic malignancies. However, it also promotes accelerated senescence in healthy tissues and leads to progressive cognitive dysfunction in up to 50% of tumor patients surviving long term after treatment, due to γ-irradiation-induced cerebromicrovascular injury. Moment-to-moment adjustment of cerebral blood flow (CBF) via neuronal activity-dependent cerebromicrovascular dilation (functional hyperemia) has a critical role in maintenance of healthy cognitive function. To determine whether cognitive decline induced by WBI associates with impaired cerebromicrovascular function, C56BL/6 mice (3 months) subjected to a clinically relevant protocol of fractionated WBI (5 Gy twice weekly for 4 weeks) and control mice were compared. Mice were tested for spatial memory performance (radial arm water maze), sensorimotor coordination (computerized gait analysis, CatWalk), and cerebromicrovascular function (whisker-stimulation-induced increases in CBF, measured by laser Doppler flowmetry) at 3 to 6 months post-irradiation. We found that mice with WBI exhibited impaired cerebromicrovascular function at 3 months post-irradiation, which was associated with impaired performance in the radial arm water maze. At 6 months, post-irradiation progressive impairment in gait coordination (including changes in the regularity index and phase dispersion) was also evident. Collectively, our findings provide evidence for early and persisting neurovascular impairment after a clinically relevant protocol of fractionated WBI, which predict early manifestations of cognitive impairment.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Stefano Tarantini
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Hertelendy
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - M Noa Valcarcel-Ares
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Gabor A Fülöp
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sreemathi Logan
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
| | - Tamas Kiss
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Eszter Farkas
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Anna Csiszar
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Andriy Yabluchanskiy
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA.
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
50
|
Hu X, De Silva TM, Chen J, Faraci FM. Cerebral Vascular Disease and Neurovascular Injury in Ischemic Stroke. Circ Res 2017; 120:449-471. [PMID: 28154097 PMCID: PMC5313039 DOI: 10.1161/circresaha.116.308427] [Citation(s) in RCA: 280] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/13/2016] [Accepted: 10/26/2016] [Indexed: 12/13/2022]
Abstract
The consequences of cerebrovascular disease are among the leading health issues worldwide. Large and small cerebral vessel disease can trigger stroke and contribute to the vascular component of other forms of neurological dysfunction and degeneration. Both forms of vascular disease are driven by diverse risk factors, with hypertension as the leading contributor. Despite the importance of neurovascular disease and subsequent injury after ischemic events, fundamental knowledge in these areas lag behind our current understanding of neuroprotection and vascular biology in general. The goal of this review is to address select key structural and functional changes in the vasculature that promote hypoperfusion and ischemia, while also affecting the extent of injury and effectiveness of therapy. In addition, as damage to the blood-brain barrier is one of the major consequences of ischemia, we discuss cellular and molecular mechanisms underlying ischemia-induced changes in blood-brain barrier integrity and function, including alterations in endothelial cells and the contribution of pericytes, immune cells, and matrix metalloproteinases. Identification of cell types, pathways, and molecules that control vascular changes before and after ischemia may result in novel approaches to slow the progression of cerebrovascular disease and lessen both the frequency and impact of ischemic events.
Collapse
Affiliation(s)
- Xiaoming Hu
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - T. Michael De Silva
- Biomedicine Discovery Institute, Department of Pharmacology, 9 Ancora Imparo Way, Monash University, Clayton, Vic, Australia
| | - Jun Chen
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Frank M. Faraci
- Departments of Internal Medicine and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City Veterans Affairs Healthcare System, Iowa City, IA, USA
| |
Collapse
|