1
|
Joutel A. The Pathobiology of Cerebrovascular Lesions in CADASIL Small Vessel Disease. Basic Clin Pharmacol Toxicol 2025; 136:e70028. [PMID: 40145673 PMCID: PMC11948957 DOI: 10.1111/bcpt.70028] [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] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025]
Abstract
Cerebral small vessel disease (cSVD) is a significant global health issue, accounting for approximately 25% of ischemic strokes and 20% of all dementia cases. CADASIL, the most common monogenic form of cSVD, is caused by stereotyped mutations in the NOTCH3 receptor that alter the number of cysteine residues in its extracellular domain (Notch3ECD). The two hallmark features of CADASIL are the loss of arterial smooth muscle cells (SMCs) and the abnormal accumulation of Notch3ECD, without associated accumulation of its transmembrane intracellular domain. Notably, cysteine-altering mutations in NOTCH3 are prevalent in the general population, and although they are not directly associated with classical CADASIL disease, they are still linked to an elevated risk of stroke and dementia. NOTCH3 is predominantly expressed in the mural cells of small blood vessels and plays an essential role in the development, maintenance, function and survival of arterial SMCs. Recent research has challenged the loss-of-function hypothesis, instead implicating Notch3ECD aggregation, involving both mutant and wild-type NOTCH3, as the primary driver of vascular pathology in CADASIL. Consequently, therapeutic strategies targeting the reduction of Notch3ECD levels in brain arteries, such as antisense therapies, are considered highly promising for clinical development.
Collapse
Affiliation(s)
- Anne Joutel
- University Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266ParisFrance
- GHU‐Paris Psychiatry and NeurosciencesSt. Anne HospitalParisFrance
| |
Collapse
|
2
|
Longden T, Isaacs D. Pericyte Electrical Signalling and Brain Haemodynamics. Basic Clin Pharmacol Toxicol 2025; 136:e70030. [PMID: 40159653 PMCID: PMC11955720 DOI: 10.1111/bcpt.70030] [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] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/24/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025]
Abstract
Dynamic control of membrane potential lies at the nexus of a wide spectrum of biological processes, ranging from the control of individual cell secretions to the orchestration of complex thought and behaviour. Electrical signals in all vascular cell types (smooth muscle cells, endothelial cells and pericytes) contribute to the control of haemodynamics and energy delivery across spatiotemporal scales and throughout all tissues. Here, our goal is to review and synthesize key studies of electrical signalling within the brain vasculature and integrate these with recent data illustrating an important electrical signalling role for pericytes, in doing so attempting to work towards a holistic description of blood flow control in the brain by vascular electrical signalling. We use this as a framework for generating further questions that we believe are important to pursue. Drawing parallels with electrical signal integration in the nervous system may facilitate deeper insights into how signalling is organized within the vasculature and how it controls blood flow at the network level.
Collapse
Affiliation(s)
- Thomas A. Longden
- Department of Pharmacology and PhysiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and TechnologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Dominic Isaacs
- Department of Pharmacology and PhysiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and TechnologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
- Program in NeuroscienceUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
3
|
Ferris HR, Jeffrey DA, Guerrero MB, Birnbaumer L, Zheng F, Dabertrand F. Increased luminal pressure in brain capillaries drives TRPC3-dependent depolarization and constriction of transitional pericytes. Sci Signal 2025; 18:eads1903. [PMID: 40299956 DOI: 10.1126/scisignal.ads1903] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 04/09/2025] [Indexed: 05/01/2025]
Abstract
Cerebral autoregulation ensures constant blood flow, an essential condition of brain health. A fundamental parameter of the brain circulation is the dynamic regulation of microvessel diameter to allow for adjustments in resistance to blood pressure changes. Pericytes are a family of mural cells that wrap around the capillary endothelium and contribute to the dynamic control of capillary diameter. We sought to determine whether and how brain pericytes constrict in response to blood pressure elevation with in vivo two-photon microscopy, electrophysiology, and ex vivo arteriolar-capillary myography of mice with conditional mural cell knockout or with expression of a genetically encoded Ca2+ indicator. In first- to fourth-order capillaries, pericytes displayed a rapid and measurable response to pressure by decreasing luminal diameter, depolarizing membrane potentials, and increasing cytoplasmic Ca2+ signaling. Pharmacological and imaging approaches revealed that transient receptor potential channel 3 (TRPC3) and voltage-gated Ca2+ channels were sequentially activated to promote fast constriction. Genetic ablation of TRPC3 resulted in decreased currents, loss of membrane depolarization, and near-complete ablation of the generation of tone over a standard pressure curve in transitional pericytes but not in upstream arterioles. Together, our findings identify TRPC3 channel activation as critical for proximal pericyte depolarization and contraction in response to pressure, highlighting the signaling differences between arteriolar and capillary blood flow regulation.
Collapse
Affiliation(s)
- Hannah R Ferris
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Danielle A Jeffrey
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Mayra Bueno Guerrero
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Lutz Birnbaumer
- Signal Transduction, National Institute of Environmental Sciences, Research Triangle Park, NC 27709, USA
- Institute of Biomedical Research (BIOMED), School of Medical Sciences, Catholic University of Argentina, Buenos Aires C1107AAZ, Argentina
| | - Fang Zheng
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Fabrice Dabertrand
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
4
|
Aykan S, Lai JH, Sugimoto K, Aykan O, Fung WY, Ho D, Joutel A, Sakadzic S, Chung DY, Ayata C. Impaired Resting-State Functional Connectivity in Cerebral Autosomal-Dominant Arteriopathy, Subcortical Infarcts, and Leukoencephalopathy Mutant Mice. Stroke 2025; 56:987-995. [PMID: 39882613 PMCID: PMC11932795 DOI: 10.1161/strokeaha.124.049772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/02/2025] [Accepted: 01/15/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Cerebral autosomal-dominant arteriopathy, subcortical infarcts, and leukoencephalopathy is the most prevalent monogenic inherited cause of cerebral small vessel disease. Despite its prevalence, there is currently no proven therapy to prevent or reverse the progression of the disease. METHODS This study aimed to characterize the functional integrity of long white matter tracts in cerebral autosomal-dominant arteriopathy, subcortical infarcts, and leukoencephalopathy transgenic mice expressing R169C mutant Notch3 (Notch3R169C) compared with wild type littermates (Notch3WT), both with and without a superimposed focal white matter lesion in the corpus callosum, utilizing optical resting-state functional connectivity imaging alongside behavioral examinations. In addition, we examined the efficacy of tocotrienol, a neuroprotective derivative of vitamin E derived from palm oil, which has shown promise in preventing white matter disease progression in clinical trials involving patients with small vessel disease. RESULTS At baseline, resting-state interhemispheric and intrahemispheric functional connectivity was significantly lower in Notch3R169C than in Notch3WT (P=0.004), and the grid walk test revealed a higher number of foot faults in the Notch3R169C group compared with Notch3WT. Sex did not interact with the genotype on the primary outcomes. Introducing a lesion in the corpus callosum compromised functional connectivity and behavior outcomes in both genotypes to a similar extent; lesion volumes did not differ between the genotypes. Tocotrienol treatment did not show any protective effect on any end point. CONCLUSIONS These data show impaired resting-state functional connectivity and increased foot faults in the Notch3R169C mutant model of cerebral autosomal-dominant arteriopathy, subcortical infarcts, and leukoencephalopathy. Future work will aim to test therapeutic or preventive interventions in cerebral autosomal-dominant arteriopathy, subcortical infarcts, and leukoencephalopathy mutants using these measures.
Collapse
Affiliation(s)
- Sanem Aykan
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - James Han Lai
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Kazutaka Sugimoto
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Orhan Aykan
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | | | | | - Anne Joutel
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, 75014 Paris, France
| | - Sava Sakadzic
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - David Y. Chung
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
| | - Cenk Ayata
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, USA
- Stroke Service, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
5
|
Yuan L, Chen X, Jankovic J, Deng H. CADASIL: A NOTCH3-associated cerebral small vessel disease. J Adv Res 2024; 66:223-235. [PMID: 38176524 PMCID: PMC11674792 DOI: 10.1016/j.jare.2024.01.001] [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: 10/18/2023] [Revised: 12/16/2023] [Accepted: 01/01/2024] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common hereditary cerebral small vessel disease (CSVD), pathologically characterized by a non-atherosclerotic and non-amyloid diffuse angiopathy primarily involving small to medium-sized penetrating arteries and leptomeningeal arteries. In 1996, mutation in the notch receptor 3 gene (NOTCH3) was identified as the cause of CADASIL. However, since that time other genetic CSVDs have been described, including the HtrA serine peptidase 1 gene-associated CSVD and the cathepsin A gene-associated CSVD, that clinically mimic the original phenotype. Though NOTCH3-associated CSVD is now a well-recognized hereditary disorder and the number of studies investigating this disease is increasing, the role of NOTCH3 in the pathogenesis of CADASIL remains elusive. AIM OF REVIEW This review aims to provide insights into the pathogenesis and the diagnosis of hereditary CSVDs, as well as personalized therapy, predictive approach, and targeted prevention. In this review, we summarize the current progress in CADASIL, including the clinical, neuroimaging, pathological, genetic, diagnostic, and therapeutic aspects, as well as differential diagnosis, in which the role of NOTCH3 mutations is highlighted. KEY SCIENTIFIC CONCEPTS OF REVIEW In this review, CADASIL is revisited as a NOTCH3-associated CSVD along with other hereditary CSVDs.
Collapse
Affiliation(s)
- Lamei Yuan
- Health Management Center, the Third Xiangya Hospital, Central South University, Changsha, China; Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, China; Disease Genome Research Center, Central South University, Changsha, China; Department of Neurology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiangyu Chen
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, China; Disease Genome Research Center, Central South University, Changsha, China; Department of Pathology, Changsha Maternal and Child Health Care Hospital, Changsha, China
| | - Joseph Jankovic
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Hao Deng
- Health Management Center, the Third Xiangya Hospital, Central South University, Changsha, China; Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, China; Disease Genome Research Center, Central South University, Changsha, China; Department of Neurology, the Third Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
6
|
Esfandi H, Javidan M, Anderson RM, Pashaie R. Depth-Dependent Contributions of Various Vascular Zones to Cerebral Autoregulation and Functional Hyperemia: An In-Silico Analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.616950. [PMID: 39416222 PMCID: PMC11482864 DOI: 10.1101/2024.10.07.616950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Autoregulation and neurogliavascular coupling are key mechanisms that modulate myogenic tone (MT) in vessels to regulate cerebral blood flow (CBF) during resting state and periods of increased neural activity, respectively. To determine relative contributions of distinct vascular zones across different cortical depths in CBF regulation, we developed a simplified yet detailed and computationally efficient model of the mouse cerebrovasculature. The model integrates multiple simplifications and generalizations regarding vascular morphology, the hierarchical organization of mural cells, and potentiation/inhibition of MT in vessels. Our analysis showed that autoregulation is the result of the synergy between these factors, but achieving an optimal balance across all cortical depths and throughout the autoregulation range is a complex task. This complexity explains the non-uniformity observed experimentally in capillary blood flow at different cortical depths. In silico simulations of cerebral autoregulation support the idea that the cerebral vasculature does not maintain a plateau of blood flow throughout the autoregulatory range and consists of both flat and sloped phases. We learned that small-diameter vessels with large contractility, such as penetrating arterioles and precapillary arterioles, have major control over intravascular pressure at the entry points of capillaries and play a significant role in CBF regulation. However, temporal alterations in capillary diameter contribute moderately to cerebral autoregulation and minimally to functional hyperemia. In addition, hemodynamic analysis shows that while hemodynamics within capillaries remain relatively stable across all cortical depths throughout the entire autoregulation range, significant variability in hemodynamics can be observed within the first few branch orders of precapillary arterioles or transitional zone vessels. The computationally efficient cerebrovasculature model, proposed in this study, provides a novel framework for analyzing dynamics of the CBF regulation where hemodynamic and vasodynamic interactions are the foundation on which more sophisticated models can be developed.
Collapse
Affiliation(s)
- Hadi Esfandi
- Electrical Engineering and Computer Science Department, Florida Atlantic University, Boca Raton, FL, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Mahshad Javidan
- Electrical Engineering and Computer Science Department, Florida Atlantic University, Boca Raton, FL, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Rozalyn M. Anderson
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research, Education, and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Ramin Pashaie
- Electrical Engineering and Computer Science Department, Florida Atlantic University, Boca Raton, FL, USA
| |
Collapse
|
7
|
A genome-wide association meta-analysis of all-cause and vascular dementia. Alzheimers Dement 2024; 20:5973-5995. [PMID: 39046104 PMCID: PMC11497727 DOI: 10.1002/alz.14115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 04/30/2024] [Accepted: 05/20/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Dementia is a multifactorial disease with Alzheimer's disease (AD) and vascular dementia (VaD) pathologies making the largest contributions. Yet, most genome-wide association studies (GWAS) focus on AD. METHODS We conducted a GWAS of all-cause dementia (ACD) and examined the genetic overlap with VaD. Our dataset includes 800,597 individuals, with 46,902 and 8702 cases of ACD and VaD, respectively. Known AD loci for ACD and VaD were replicated. Bioinformatic analyses prioritized genes that are likely functionally relevant and shared with closely related traits and risk factors. RESULTS For ACD, novel loci identified were associated with energy transport (SEMA4D), neuronal excitability (ANO3), amyloid deposition in the brain (RBFOX1), and magnetic resonance imaging markers of small vessel disease (SVD; HBEGF). Novel VaD loci were associated with hypertension, diabetes, and neuron maintenance (SPRY2, FOXA2, AJAP1, and PSMA3). DISCUSSION Our study identified genetic risks underlying ACD, demonstrating overlap with neurodegenerative processes, vascular risk factors, and cerebral SVD. HIGHLIGHTS We conducted the largest genome-wide association study of all-cause dementia (ACD) and vascular dementia (VaD). Known genetic variants associated with AD were replicated for ACD and VaD. Functional analyses identified novel loci for ACD and VaD. Genetic risks of ACD overlapped with neurodegeneration, vascular risk factors, and cerebral small vessel disease.
Collapse
|
8
|
Sancho M, Klug NR, Harraz OF, Hill-Eubanks D, Nelson MT. Distinct potassium channel types in brain capillary pericytes. Biophys J 2024; 123:2110-2121. [PMID: 38444160 PMCID: PMC11309962 DOI: 10.1016/j.bpj.2024.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/25/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024] Open
Abstract
Capillaries, composed of electrically coupled endothelial cells and overlying pericytes, constitute the vast majority of blood vessels in the brain. The most arteriole-proximate three to four branches of the capillary bed are covered by α-actin-expressing, contractile pericytes. These mural cells have a distinctive morphology and express different markers compared with their smooth muscle cell (SMC) cousins but share similar excitation-coupling contraction machinery. Despite this similarity, pericytes are considerably more depolarized than SMCs at low intravascular pressures. We have recently shown that pericytes, such as SMCs, possess functional voltage-dependent Ca2+ channels and ATP-sensitive K+ channels. Here, we further investigate the complement of pericyte ion channels, focusing on members of the K+ channel superfamily. Using NG2-DsRed-transgenic mice and diverse configurations of the patch-clamp technique, we demonstrate that pericytes display robust inward-rectifier K+ currents that are primarily mediated by the Kir2 family, based on their unique biophysical characteristics and sensitivity to micromolar concentrations of Ba2+. Moreover, multiple lines of evidence, including characteristic kinetics, sensitivity to specific blockers, biophysical attributes, and distinctive single-channel properties, established the functional expression of two voltage-dependent K+ channels: KV1 and BKCa. Although these three types of channels are also present in SMCs, they exhibit distinctive current density and kinetics profiles in pericytes. Collectively, these findings underscore differences in the operation of shared molecular features between pericytes and SMCs and highlight the potential contribution of these three K+ ion channels in setting pericyte membrane potential, modulating capillary hemodynamics, and regulating cerebral blood flow.
Collapse
Affiliation(s)
- Maria Sancho
- Department of Pharmacology, University of Vermont, Burlington, Vermont; Department of Physiology, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain.
| | - Nicholas R Klug
- Department of Pharmacology, University of Vermont, Burlington, Vermont
| | - Osama F Harraz
- Department of Pharmacology, University of Vermont, Burlington, Vermont; Vermont Center for Cardiovascular and Brain Health, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | | | - Mark T Nelson
- Department of Pharmacology, University of Vermont, Burlington, Vermont; Vermont Center for Cardiovascular and Brain Health, Larner College of Medicine, University of Vermont, Burlington, Vermont; Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
9
|
Kang M, Mun SY, Zhuang W, Park M, Jeong J, Park H, Jung WK, Choi IW, Na S, Park WS. Inhibition of voltage-gated potassium channel by aripiprazole in rabbit coronary arterial smooth muscle cells. Eur J Pharmacol 2024; 973:176610. [PMID: 38663541 DOI: 10.1016/j.ejphar.2024.176610] [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: 03/24/2024] [Accepted: 04/22/2024] [Indexed: 05/08/2024]
Abstract
Aripiprazole, a third-generation antipsychotic, has been widely used to treat schizophrenia. In this study, we evaluated the effect of aripiprazole on voltage-gated potassium (Kv) channels in rabbit coronary arterial smooth muscle cells using the patch clamp technique. Aripiprazole reduced the Kv current in a concentration-dependent manner with a half-maximal inhibitory concentration of 0.89 ± 0.20 μM and a Hill coefficient of 1.30 ± 0.25. The inhibitory effect of aripiprazole on Kv channels was voltage-dependent, and an additional aripiprazole-induced decrease in the Kv current was observed in the voltage range of full channel activation. The decay rate of Kv channel inactivation was accelerated by aripiprazole. Aripiprazole shifted the steady-state activation curve to the right and the inactivation curve to the left. Application of a repetitive train of pulses (1 and 2 Hz) promoted inhibition of the Kv current by aripiprazole. Furthermore, the recovery time constant from inactivation increased in the presence of aripiprazole. Pretreatment of Kv1.5 subtype inhibitor reduced the inhibitory effect of aripiprazole. However, pretreatment with Kv 7 and Kv2.1 subtype inhibitors did not change the degree of aripiprazole-induced inhibition of the Kv current. We conclude that aripiprazole inhibits Kv channels in a concentration-, voltage-, time-, and use (state)-dependent manner by affecting the gating properties of the channels.
Collapse
Affiliation(s)
- Minji Kang
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Seo-Yeong Mun
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Wenwen Zhuang
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Minju Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Junsu Jeong
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Hongzoo Park
- Institute of Medical Sciences, Department of Urology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Won-Kyo Jung
- Department of Biomedical Engineering, and Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan, 48513, South Korea
| | - Il-Whan Choi
- Department of Microbiology, College of Medicine, Inje University, Busan, 48516, South Korea
| | - Sunghun Na
- Institute of Medical Sciences, Department of Obstetrics and Gynecology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Won Sun Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea.
| |
Collapse
|
10
|
Dupré N, Drieu A, Joutel A. Pathophysiology of cerebral small vessel disease: a journey through recent discoveries. J Clin Invest 2024; 134:e172841. [PMID: 38747292 PMCID: PMC11093606 DOI: 10.1172/jci172841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024] Open
Abstract
Cerebral small vessel disease (cSVD) encompasses a heterogeneous group of age-related small vessel pathologies that affect multiple regions. Disease manifestations range from lesions incidentally detected on neuroimaging (white matter hyperintensities, small deep infarcts, microbleeds, or enlarged perivascular spaces) to severe disability and cognitive impairment. cSVD accounts for approximately 25% of ischemic strokes and the vast majority of spontaneous intracerebral hemorrhage and is also the most important vascular contributor to dementia. Despite its high prevalence and potentially long therapeutic window, there are still no mechanism-based treatments. Here, we provide an overview of the recent advances in this field. We summarize recent data highlighting the remarkable continuum between monogenic and multifactorial cSVDs involving NOTCH3, HTRA1, and COL4A1/A2 genes. Taking a vessel-centric view, we discuss possible cause-and-effect relationships between risk factors, structural and functional vessel changes, and disease manifestations, underscoring some major knowledge gaps. Although endothelial dysfunction is rightly considered a central feature of cSVD, the contributions of smooth muscle cells, pericytes, and other perivascular cells warrant continued investigation.
Collapse
Affiliation(s)
- Nicolas Dupré
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France
| | - Antoine Drieu
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France
| | - Anne Joutel
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France
| |
Collapse
|
11
|
Djurich S, Secomb TW. Analysis of potassium ion diffusion from neurons to capillaries: Effects of astrocyte endfeet geometry. Eur J Neurosci 2024; 59:323-332. [PMID: 38123136 PMCID: PMC10872621 DOI: 10.1111/ejn.16232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 11/25/2023] [Accepted: 12/02/2023] [Indexed: 12/23/2023]
Abstract
Neurovascular coupling (NVC) refers to a local increase in cerebral blood flow in response to increased neuronal activity. Mechanisms of communication between neurons and blood vessels remain unclear. Astrocyte endfeet almost completely cover cerebral capillaries, suggesting that astrocytes play a role in NVC by releasing vasoactive substances near capillaries. An alternative hypothesis is that direct diffusion through the extracellular space of potassium ions (K+ ) released by neurons contributes to NVC. Here, the goal is to determine whether astrocyte endfeet present a barrier to K+ diffusion from neurons to capillaries. Two simplified 2D geometries of extracellular space, clefts between endfeet, and perivascular space are used: (i) a source 1 μm from a capillary; (ii) a neuron 15 μm from a capillary. K+ release is modelled as a step increase in [K+ ] at the outer boundary of the extracellular space. The time-dependent diffusion equation is solved numerically. In the first geometry, perivascular [K+ ] approaches its final value within 0.05 s. Decreasing endfeet cleft width or increasing perivascular space width slows the rise in [K+ ]. In the second geometry, the increase in perivascular [K+ ] occurs within 0.5 s and is insensitive to changes in cleft width or perivascular space width. Predicted levels of perivascular [K+ ] are sufficient to cause vasodilation, and the rise time is within the time for flow increase in NVC. These results suggest that direct diffusion of K+ through the extracellular space is a possible NVC signalling mechanism.
Collapse
Affiliation(s)
- Sara Djurich
- Department of Physiology, University of Arizona, Tucson, Arizona, USA
| | - Timothy W Secomb
- Department of Physiology, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
12
|
Yamasaki E, Thakore P, Ali S, Solano AS, Wang X, Gao X, Labelle-Dumais C, Chaumeil MM, Gould DB, Earley S. Impaired intracellular Ca 2+ signaling contributes to age-related cerebral small vessel disease in Col4a1 mutant mice. Sci Signal 2023; 16:eadi3966. [PMID: 37963192 PMCID: PMC10726848 DOI: 10.1126/scisignal.adi3966] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 10/25/2023] [Indexed: 11/16/2023]
Abstract
Humans and mice with mutations in COL4A1 and COL4A2 manifest hallmarks of cerebral small vessel disease (cSVD). Mice with a missense mutation in Col4a1 at amino acid 1344 (Col4a1+/G1344D) exhibit age-dependent intracerebral hemorrhages (ICHs) and brain lesions. Here, we report that this pathology was associated with the loss of myogenic vasoconstriction, an intrinsic vascular response essential for the autoregulation of cerebral blood flow. Electrophysiological analyses showed that the loss of myogenic constriction resulted from blunted pressure-induced smooth muscle cell (SMC) membrane depolarization. Furthermore, we found that dysregulation of membrane potential was associated with impaired Ca2+-dependent activation of large-conductance Ca2+-activated K+ (BK) and transient receptor potential melastatin 4 (TRPM4) cation channels linked to disruptions in sarcoplasmic reticulum (SR) Ca2+ signaling. Col4a1 mutations impair protein folding, which can cause SR stress. Treating Col4a1+/G1344D mice with 4-phenylbutyrate, a compound that promotes the trafficking of misfolded proteins and alleviates SR stress, restored SR Ca2+ signaling, maintained BK and TRPM4 channel activity, prevented loss of myogenic tone, and reduced ICHs. We conclude that alterations in SR Ca2+ handling that impair ion channel activity result in dysregulation of SMC membrane potential and loss of myogenic tone and contribute to age-related cSVD in Col4a1+/G1344D mice.
Collapse
Affiliation(s)
- Evan Yamasaki
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV 89557-0318, USA
| | - Pratish Thakore
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV 89557-0318, USA
| | - Sher Ali
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV 89557-0318, USA
| | - Alfredo Sanchez Solano
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV 89557-0318, USA
| | - Xiaowei Wang
- Department of Ophthalmology, UCSF School of Medicine, San Francisco, CA 94158, USA
| | - Xiao Gao
- Department of Physical Therapy and Rehabilitation Science, UCSF School of Medicine, San Francisco, CA 94143, USA
- Department of Radiology and Biomedical Imaging, UCSF School of Medicine, San Francisco, CA 94143, USA
| | | | - Myriam M. Chaumeil
- Department of Physical Therapy and Rehabilitation Science, UCSF School of Medicine, San Francisco, CA 94143, USA
- Department of Radiology and Biomedical Imaging, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Douglas B. Gould
- Department of Ophthalmology, UCSF School of Medicine, San Francisco, CA 94158, USA
- Department of Anatomy, Institute for Human Genetics, Cardiovascular Research Institute, Bakar Aging Research Institute, UCSF School of Medicine, San Francisco, CA 94158, USA
| | - Scott Earley
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV 89557-0318, USA
| |
Collapse
|
13
|
Ferris HR, Stine NC, Hill-Eubanks DC, Nelson MT, Wellman GC, Koide M. Epidermal Growth Factor Receptors in Vascular Endothelial Cells Contribute to Functional Hyperemia in the Brain. Int J Mol Sci 2023; 24:16284. [PMID: 38003472 PMCID: PMC10671586 DOI: 10.3390/ijms242216284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/06/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
Functional hyperemia-activity-dependent increases in local blood perfusion-underlies the on-demand delivery of blood to regions of enhanced neuronal activity, a process that is crucial for brain health. Importantly, functional hyperemia deficits have been linked to multiple dementia risk factors, including aging, chronic hypertension, and cerebral small vessel disease (cSVD). We previously reported crippled functional hyperemia in a mouse model of genetic cSVD that was likely caused by depletion of phosphatidylinositol 4,5-bisphosphate (PIP2) in capillary endothelial cells (EC) downstream of impaired epidermal growth factor receptor (EGFR) signaling. Here, using EC-specific EGFR-knockout (KO) mice, we directly examined the role of endothelial EGFR signaling in functional hyperemia, assessed by measuring increases in cerebral blood flow in response to contralateral whisker stimulation using laser Doppler flowmetry. Molecular characterizations showed that EGFR expression was dramatically decreased in freshly isolated capillaries from EC-EGFR-KO mice, as expected. Notably, whisker stimulation-induced functional hyperemia was significantly impaired in these mice, an effect that was rescued by administration of PIP2, but not by the EGFR ligand, HB-EGF. These data suggest that the deletion of the EGFR specifically in ECs attenuates functional hyperemia, likely via depleting PIP2 and subsequently incapacitating Kir2.1 channel functionality in capillary ECs. Thus, our study underscores the role of endothelial EGFR signaling in functional hyperemia of the brain.
Collapse
Affiliation(s)
- Hannah R. Ferris
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT 05405, USA; (H.R.F.); (N.C.S.)
| | - Nathan C. Stine
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT 05405, USA; (H.R.F.); (N.C.S.)
| | - David C. Hill-Eubanks
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT 05405, USA; (H.R.F.); (N.C.S.)
| | - Mark T. Nelson
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT 05405, USA; (H.R.F.); (N.C.S.)
- Vermont Center for Cardiovascular and Brain Health, Larner College of Medicine University of Vermont, Burlington, VT 05405, USA
- Division of Cardiovascular Sciences, University of Manchester, Manchester M13 9PL, UK
| | - George C. Wellman
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT 05405, USA; (H.R.F.); (N.C.S.)
| | - Masayo Koide
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT 05405, USA; (H.R.F.); (N.C.S.)
- Vermont Center for Cardiovascular and Brain Health, Larner College of Medicine University of Vermont, Burlington, VT 05405, USA
| |
Collapse
|
14
|
Ferris HR, Hill-Eubanks DC, Nelson MT, Wellman GC, Koide M. Epidermal growth factor receptors in vascular endothelial cells contribute to functional hyperemia in the brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.15.557981. [PMID: 37745396 PMCID: PMC10516026 DOI: 10.1101/2023.09.15.557981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Functional hyperemia - activity-dependent increases in local blood perfusion - underlies the on-demand delivery of blood to regions of enhanced neuronal activity, a process that is crucial for brain health. Importantly, functional hyperemia deficits have been linked to multiple dementia risk factors, including aging, chronic hypertension, and cerebral small vessel disease (cSVD). We previously reported crippled functional hyperemia in a mouse model of genetic cSVD that was likely caused by depletion of phosphatidylinositol 4,5-bisphosphate (PIP2) in capillary endothelial cells (EC) downstream of impaired epidermal growth factor receptor (EGFR) signaling. Here, using EC-specific EGFR-knockout (KO) mice, we directly examined the role of endothelial EGFR signaling in functional hyperemia, assessed by measuring increases in cerebral blood flow in response to contralateral whisker stimulation using laser Doppler flowmetry. Molecular characterizations showed that EGFR expression was dramatically decreased in freshly isolated capillaries from EC-EGFR-KO mice, as expected. Notably, whisker stimulation-induced functional hyperemia was significantly impaired in these mice, an effect that was rescued by exogenous administration of PIP2, but not by the EGFR ligand, HB-EGF. These data suggest that the deletion of the EGFR specifically in ECs depletes PIP2 and attenuates functional hyperemia, underscoring the central role of the endothelial EGFR signaling in cerebral blood flow regulation.
Collapse
Affiliation(s)
- Hannah R. Ferris
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT, USA
| | - David C. Hill-Eubanks
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT, USA
| | - Mark T. Nelson
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, Larner College of Medicine University of Vermont, Burlington, VT, USA
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - George C. Wellman
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT, USA
| | - Masayo Koide
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, Larner College of Medicine University of Vermont, Burlington, VT, USA
| |
Collapse
|
15
|
Thakore P, Yamasaki E, Ali S, Sanchez Solano A, Labelle-Dumais C, Gao X, Chaumeil MM, Gould DB, Earley S. PI3K block restores age-dependent neurovascular coupling defects associated with cerebral small vessel disease. Proc Natl Acad Sci U S A 2023; 120:e2306479120. [PMID: 37607233 PMCID: PMC10467353 DOI: 10.1073/pnas.2306479120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/17/2023] [Indexed: 08/24/2023] Open
Abstract
Neurovascular coupling (NVC), a vital physiological process that rapidly and precisely directs localized blood flow to the most active regions of the brain, is accomplished in part by the vast network of cerebral capillaries acting as a sensory web capable of detecting increases in neuronal activity and orchestrating the dilation of upstream parenchymal arterioles. Here, we report a Col4a1 mutant mouse model of cerebral small vessel disease (cSVD) with age-dependent defects in capillary-to-arteriole dilation, functional hyperemia in the brain, and memory. The fundamental defect in aged mutant animals was the depletion of the minor membrane phospholipid phosphatidylinositol 4,5 bisphosphate (PIP2) in brain capillary endothelial cells, leading to the loss of inwardly rectifying K+ (Kir2.1) channel activity. Blocking phosphatidylinositol-3-kinase (PI3K), an enzyme that diminishes the bioavailability of PIP2 by converting it to phosphatidylinositol (3, 4, 5)-trisphosphate (PIP3), restored Kir2.1 channel activity, capillary-to-arteriole dilation, and functional hyperemia. In longitudinal studies, chronic PI3K inhibition also improved the memory function of aged Col4a1 mutant mice. Our data suggest that PI3K inhibition is a viable therapeutic strategy for treating defective NVC and cognitive impairment associated with cSVD.
Collapse
Affiliation(s)
- Pratish Thakore
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System University of Nevada, Reno School of Medicine, Reno, NV89557-0318
| | - Evan Yamasaki
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System University of Nevada, Reno School of Medicine, Reno, NV89557-0318
| | - Sher Ali
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System University of Nevada, Reno School of Medicine, Reno, NV89557-0318
| | - Alfredo Sanchez Solano
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System University of Nevada, Reno School of Medicine, Reno, NV89557-0318
| | - Cassandre Labelle-Dumais
- Department of Ophthalmology and Anatomy, Institute for Human Genetics, University of California San Francisco School of Medicine, San Francisco, CA94143
| | - Xiao Gao
- Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA94158
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA94143-0628
| | - Myriam M. Chaumeil
- Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA94158
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA94143-0628
| | - Douglas B. Gould
- Department of Ophthalmology and Anatomy, Institute for Human Genetics, University of California San Francisco School of Medicine, San Francisco, CA94143
| | - Scott Earley
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System University of Nevada, Reno School of Medicine, Reno, NV89557-0318
| |
Collapse
|
16
|
Jeffrey DA, Russell A, Guerrero MB, Fontaine JT, Romero P, Rosehart AC, Dabertrand F. Estrogen regulates myogenic tone in hippocampal arterioles by enhanced basal release of nitric oxide and endothelial SK Ca channel activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.15.553442. [PMID: 37645715 PMCID: PMC10462022 DOI: 10.1101/2023.08.15.553442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Arteries and arterioles exhibit myogenic tone, a partially constricted state that allows further constriction or dilation in response to moment-to-moment fluctuations in blood pressure. The vascular endothelium that lines the internal surface of all blood vessels controls a wide variety of essential functions, including the contractility of the adjacent smooth muscle cells by providing a tonic vasodilatory influence. Studies conducted on large (pial) arteries on the surface of the brain have shown that estrogen lowers myogenic tone in female mice by enhancing nitric oxide (NO) release from the endothelium, however, whether this difference extends to the intracerebral microcirculation remains ambiguous. The existing incomplete picture of sex differences in cerebrovascular physiology combined with a deficiency in treatments that fully restore cognitive function after cerebrovascular accidents places heavy emphasis on the necessity to investigate myogenic tone regulation in the microcirculation from both male and female mice. We hypothesized that sex-linked hormone regulation of myogenic tone extends its influence on the microcirculation level, and sought to characterize it in isolated arterioles from the hippocampus, a major cognitive brain area. Using diameter measurements both in vivo (acute cranial window vascular diameter) and ex vivo (pressure myography experiments), we measured lower myogenic tone responses in hippocampal arterioles from female than male mice. By using a combined surgical and pharmacological approach, we found myogenic tone in ovariectomized (OVX) female mice matches that of males, as well as in endothelium-denuded arterioles. Interestingly, eNOS inhibition induced a larger constriction in female arterioles but only partially abolished the difference in tone. We identified that the remnant difference was mediated by a higher activity and expression of the small-conductance Ca 2+ -sensitive K + (SK) channels. Collectively, these data indicate that eNOS and SK channels exert greater vasodilatory influence over myogenic tone in female mice at physiological pressures.
Collapse
|
17
|
Hanson A, McClenaghan C, Weng KC, Colijn S, Stratman AN, Halabi CM, Grange DK, Silva JR, Nichols CG. Electrophysiology of human iPSC-derived vascular smooth muscle cells and cell autonomous consequences of Cantu Syndrome mutations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.29.547088. [PMID: 37425756 PMCID: PMC10327170 DOI: 10.1101/2023.06.29.547088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Objective Cantu Syndrome (CS), a multisystem disease with a complex cardiovascular phenotype, is caused by GoF variants in the Kir6.1/SUR2 subunits of ATP-sensitive potassium (K ATP ) channels, and is characterized by low systemic vascular resistance, as well as tortuous, dilated vessels, and decreased pulse-wave velocity. Thus, CS vascular dysfunction is multifactorial, with distinct hypomyotonic and hyperelastic components. To dissect whether such complexities arise cell-autonomously within vascular smooth muscle cells (VSMCs), or as secondary responses to the pathophysiological milieu, we assessed electrical properties and gene expression in human induced pluripotent stem cell-derived VSMCs (hiPSC-VSMCs), differentiated from control and CS patient-derived hiPSCs, and in native mouse control and CS VSMCs. Approach and Results Whole-cell voltage-clamp of isolated aortic and mesenteric VSMCs isolated from wild type (WT) and Kir6.1[V65M] (CS) mice revealed no difference in voltage-gated K + (K v ) or Ca 2+ currents. K v and Ca 2+ currents were also not different between validated hiPSC-VSMCs differentiated from control and CS patient-derived hiPSCs. Pinacidil-sensitive K ATP currents in control hiPSC-VSMCs were consistent with those in WT mouse VSMCs, and were considerably larger in CS hiPSC-VSMCs. Consistent with lack of any compensatory modulation of other currents, this resulted in membrane hyperpolarization, explaining the hypomyotonic basis of CS vasculopathy. Increased compliance and dilation in isolated CS mouse aortae, was associated with increased elastin mRNA expression. This was consistent with higher levels of elastin mRNA in CS hiPSC-VSMCs, suggesting that the hyperelastic component of CS vasculopathy is a cell-autonomous consequence of vascular K ATP GoF. Conclusions The results show that hiPSC-VSMCs reiterate expression of the same major ion currents as primary VSMCs, validating the use of these cells to study vascular disease. The results further indicate that both the hypomyotonic and hyperelastic components of CS vasculopathy are cell-autonomous phenomena driven by K ATP overactivity within VSMCs.
Collapse
|
18
|
Thakore P, Yamasaki E, Ali S, Solano AS, Labelle-Dumais C, Gao X, Chaumeil MM, Gould DB, Earley S. PI3K block restores age-dependent neurovascular coupling defects associated with cerebral small vessel disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.03.531032. [PMID: 36945616 PMCID: PMC10028793 DOI: 10.1101/2023.03.03.531032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
Neurovascular coupling (NVC), a vital physiological process that rapidly and precisely directs localized blood flow to the most active regions of the brain, is accomplished in part by the vast network of cerebral capillaries acting as a sensory web capable of detecting increases in neuronal activity and orchestrating the dilation of upstream parenchymal arterioles. Here, we report a Col4a1 mutant mouse model of cerebral small vessel disease (cSVD) with age-dependent defects in capillary-to-arteriole dilation, functional hyperemia in the brain, and memory. The fundamental defect in aged mutant animals was the depletion of the minor membrane phospholipid phosphatidylinositol 4,5 bisphosphate (PIP 2 ) in brain capillary endothelial cells, leading to the loss of inwardly rectifier K + (Kir2.1) channel activity. Blocking phosphatidylinositol-3-kinase (PI3K), an enzyme that diminishes the bioavailability of PIP 2 by converting it to phosphatidylinositol (3,4,5)-trisphosphate (PIP 3 ), restored Kir2.1 channel activity, capillary-to-arteriole dilation, and functional hyperemia. In longitudinal studies, chronic PI3K inhibition also improved the memory function of aged Col4a1 mutant mice. Our data suggest that PI3K inhibition is a viable therapeutic strategy for treating defective NVC and cognitive impairment associated with cSVD. One-sentence summary PI3K inhibition rescues neurovascular coupling defects in cerebral small vessel disease.
Collapse
|
19
|
Johnson AC. Hippocampal Vascular Supply and Its Role in Vascular Cognitive Impairment. Stroke 2023; 54:673-685. [PMID: 36848422 PMCID: PMC9991081 DOI: 10.1161/strokeaha.122.038263] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/22/2022] [Indexed: 03/01/2023]
Abstract
The incidence of age-related dementia is increasing as the world population ages and due to lack of effective treatments for dementia. Vascular contributions to cognitive impairment and dementia are increasing as the prevalence of pathologies associated with cerebrovascular disease rise, including chronic hypertension, diabetes, and ischemic stroke. The hippocampus is a bilateral deep brain structure that is central to learning, memory, and cognitive function and highly susceptible to hypoxic/ischemic injury. Compared with cortical brain regions such as the somatosensory cortex, less is known about the function of the hippocampal vasculature that is critical in maintaining neurocognitive health. This review focuses on the hippocampal vascular supply, presenting what is known about hippocampal hemodynamics and blood-brain barrier function during health and disease, and discusses evidence that supports its contribution to vascular cognitive impairment and dementia. Understanding vascular-mediated hippocampal injury that contributes to memory dysfunction during healthy aging and cerebrovascular disease is essential to develop effective treatments to slow cognitive decline. The hippocampus and its vasculature may represent one such therapeutic target to mitigate the dementia epidemic.
Collapse
Affiliation(s)
- Abbie C Johnson
- Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington
| |
Collapse
|
20
|
Klug NR, Sancho M, Gonzales AL, Heppner TJ, O’Brien RIC, Hill-Eubanks D, Nelson MT. Intraluminal pressure elevates intracellular calcium and contracts CNS pericytes: Role of voltage-dependent calcium channels. Proc Natl Acad Sci U S A 2023; 120:e2216421120. [PMID: 36802432 PMCID: PMC9992766 DOI: 10.1073/pnas.2216421120] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/24/2023] [Indexed: 02/23/2023] Open
Abstract
Arteriolar smooth muscle cells (SMCs) and capillary pericytes dynamically regulate blood flow in the central nervous system in the face of fluctuating perfusion pressures. Pressure-induced depolarization and Ca2+ elevation provide a mechanism for regulation of SMC contraction, but whether pericytes participate in pressure-induced changes in blood flow remains unknown. Here, utilizing a pressurized whole-retina preparation, we found that increases in intraluminal pressure in the physiological range induce contraction of both dynamically contractile pericytes in the arteriole-proximate transition zone and distal pericytes of the capillary bed. We found that the contractile response to pressure elevation was slower in distal pericytes than in transition zone pericytes and arteriolar SMCs. Pressure-evoked elevation of cytosolic Ca2+ and contractile responses in SMCs were dependent on voltage-dependent Ca2+ channel (VDCC) activity. In contrast, Ca2+ elevation and contractile responses were partially dependent on VDCC activity in transition zone pericytes and independent of VDCC activity in distal pericytes. In both transition zone and distal pericytes, membrane potential at low inlet pressure (20 mmHg) was approximately -40 mV and was depolarized to approximately -30 mV by an increase in pressure to 80 mmHg. The magnitude of whole-cell VDCC currents in freshly isolated pericytes was approximately half that measured in isolated SMCs. Collectively, these results indicate a loss of VDCC involvement in pressure-induced constriction along the arteriole-capillary continuum. They further suggest that alternative mechanisms and kinetics of Ca2+ elevation, contractility, and blood flow regulation exist in central nervous system capillary networks, distinguishing them from neighboring arterioles.
Collapse
Affiliation(s)
- Nicholas R. Klug
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT05405
| | - Maria Sancho
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT05405
| | - Albert L. Gonzales
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT05405
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV89557
| | - Thomas J. Heppner
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT05405
| | | | - David Hill-Eubanks
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT05405
| | - Mark T. Nelson
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT05405
- Division of Cardiovascular Sciences, University of Manchester, ManchesterM13 9PL, UK
| |
Collapse
|
21
|
Yamasaki E, Ali S, Sanchez Solano A, Thakore P, Smith M, Wang X, Labelle-Dumais C, Gould DB, Earley S. Faulty TRPM4 channels underlie age-dependent cerebral vascular dysfunction in Gould syndrome. Proc Natl Acad Sci U S A 2023; 120:e2217327120. [PMID: 36693102 PMCID: PMC9945977 DOI: 10.1073/pnas.2217327120] [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] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/27/2022] [Indexed: 01/25/2023] Open
Abstract
Gould syndrome is a rare multisystem disorder resulting from autosomal dominant mutations in the collagen-encoding genes COL4A1 and COL4A2. Human patients and Col4a1 mutant mice display brain pathology that typifies cerebral small vessel diseases (cSVDs), including white matter hyperintensities, dilated perivascular spaces, lacunar infarcts, microbleeds, and spontaneous intracerebral hemorrhage. The underlying pathogenic mechanisms are unknown. Using the Col4a1+/G394V mouse model, we found that vasoconstriction in response to internal pressure-the vascular myogenic response-is blunted in cerebral arteries from middle-aged (12 mo old) but not young adult (3 mo old) animals, revealing age-dependent cerebral vascular dysfunction. The defect in the myogenic response was associated with a significant decrease in depolarizing cation currents conducted by TRPM4 (transient receptor potential melastatin 4) channels in native cerebral artery smooth muscle cells (SMCs) isolated from mutant mice. The minor membrane phospholipid phosphatidylinositol 4,5 bisphosphate (PIP2) is necessary for TRPM4 activity. Dialyzing SMCs with PIP2 and selective blockade of phosphoinositide 3-kinase (PI3K), an enzyme that converts PIP2 to phosphatidylinositol (3, 4, 5)-trisphosphate (PIP3), restored TRPM4 currents. Acute inhibition of PI3K activity and blockade of transforming growth factor-beta (TGF-β) receptors also rescued the myogenic response, suggesting that hyperactivity of TGF-β signaling pathways stimulates PI3K to deplete PIP2 and impair TRPM4 channels. We conclude that age-related cerebral vascular dysfunction in Col4a1+/G394V mice is caused by the loss of depolarizing TRPM4 currents due to PIP2 depletion, revealing an age-dependent mechanism of cSVD.
Collapse
Affiliation(s)
- Evan Yamasaki
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV89557-0318
| | - Sher Ali
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV89557-0318
| | - Alfredo Sanchez Solano
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV89557-0318
| | - Pratish Thakore
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV89557-0318
| | - Megan Smith
- Departments of Ophthalmology, Institute for Human Genetics, UCSF School of Medicine, San Francisco, CA94158
| | - Xiaowei Wang
- Departments of Ophthalmology, Institute for Human Genetics, UCSF School of Medicine, San Francisco, CA94158
| | - Cassandre Labelle-Dumais
- Departments of Ophthalmology, Institute for Human Genetics, UCSF School of Medicine, San Francisco, CA94158
| | - Douglas B. Gould
- Departments of Ophthalmology, Institute for Human Genetics, UCSF School of Medicine, San Francisco, CA94158
- Department of Anatomy, Institute for Human Genetics, Cardiovascular Research Institute, Bakar Aging Research Institute, UCSF School of Medicine, San Francisco, CA94158
| | - Scott Earley
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV89557-0318
| |
Collapse
|
22
|
van den Brink H, Kopczak A, Arts T, Onkenhout L, Siero JCW, Zwanenburg JJM, Hein S, Hübner M, Gesierich B, Duering M, Stringer MS, Hendrikse J, Wardlaw JM, Joutel A, Dichgans M, Biessels GJ. CADASIL Affects Multiple Aspects of Cerebral Small Vessel Function on 7T-MRI. Ann Neurol 2023; 93:29-39. [PMID: 36222455 DOI: 10.1002/ana.26527] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 10/05/2022] [Accepted: 10/07/2022] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Cerebral small vessel diseases (cSVDs) are a major cause of stroke and dementia. We used cutting-edge 7T-MRI techniques in patients with Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL), to establish which aspects of cerebral small vessel function are affected by this monogenic form of cSVD. METHODS We recruited 23 CADASIL patients (age 51.1 ± 10.1 years, 52% women) and 13 age- and sex-matched controls (46.1 ± 12.6, 46% women). Small vessel function measures included: basal ganglia and centrum semiovale perforating artery blood flow velocity and pulsatility, vascular reactivity to a visual stimulus in the occipital cortex and reactivity to hypercapnia in the cortex, subcortical gray matter, white matter, and white matter hyperintensities. RESULTS Compared with controls, CADASIL patients showed lower blood flow velocity and higher pulsatility index within perforating arteries of the centrum semiovale (mean difference - 0.09 cm/s, p = 0.03 and 0.20, p = 0.009) and basal ganglia (mean difference - 0.98 cm/s, p = 0.003 and 0.17, p = 0.06). Small vessel reactivity to a short visual stimulus was decreased (blood-oxygen-level dependent [BOLD] mean difference -0.21%, p = 0.04) in patients, while reactivity to hypercapnia was preserved in the cortex, subcortical gray matter, and normal appearing white matter. Among patients, reactivity to hypercapnia was decreased in white matter hyperintensities compared to normal appearing white matter (BOLD mean difference -0.29%, p = 0.02). INTERPRETATION Multiple aspects of cerebral small vessel function on 7T-MRI were abnormal in CADASIL patients, indicative of increased arteriolar stiffness and regional abnormalities in reactivity, locally also in relation to white matter injury. These observations provide novel markers of cSVD for mechanistic and intervention studies. ANN NEUROL 2023;93:29-39.
Collapse
Affiliation(s)
- Hilde van den Brink
- Department of Neurology and Neurosurgery, University Medical Center Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anna Kopczak
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Tine Arts
- Department of Radiology, Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Laurien Onkenhout
- Department of Neurology and Neurosurgery, University Medical Center Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jeroen C W Siero
- Department of Radiology, Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands.,Spinoza Centre for Neuroimaging Amsterdam, Amsterdam, The Netherlands
| | - Jaco J M Zwanenburg
- Department of Radiology, Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sandra Hein
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Mathias Hübner
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Benno Gesierich
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany.,Medical Image Analysis Center (MIAC AG) and qbig, Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Marco Duering
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany.,Medical Image Analysis Center (MIAC AG) and qbig, Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Michael S Stringer
- Brain Research Imaging Centre, Centre for Clinical Brain Sciences, UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, UK
| | - Jeroen Hendrikse
- Department of Radiology, Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Joanna M Wardlaw
- Brain Research Imaging Centre, Centre for Clinical Brain Sciences, UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, UK
| | - Anne Joutel
- Institute of Psychiatry and Neurosciences of Paris, Université de Paris, Inserm U1266, Paris, France
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,German Center for Neurodegenerative Disease (DZNE), Munich, Germany
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, University Medical Center Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
23
|
Branyan K, Labelle-Dumais C, Wang X, Hayashi G, Lee B, Peltz Z, Gorman S, Li BQ, Mao M, Gould DB. Elevated TGFβ signaling contributes to cerebral small vessel disease in mouse models of Gould syndrome. Matrix Biol 2023; 115:48-70. [PMID: 36435425 PMCID: PMC10393528 DOI: 10.1016/j.matbio.2022.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
Cerebral small vessel disease (CSVD) is a leading cause of stroke and vascular cognitive impairment and dementia. Studying monogenic CSVD can reveal pathways that are dysregulated in common sporadic forms of the disease and may represent therapeutic targets. Mutations in collagen type IV alpha 1 (COL4A1) and alpha 2 (COL4A2) cause highly penetrant CSVD as part of a multisystem disorder referred to as Gould syndrome. COL4A1 and COL4A2 form heterotrimers [a1α1α2(IV)] that are fundamental constituents of basement membranes. However, their functions are poorly understood and the mechanism(s) by which COL4A1 and COL4A2 mutations cause CSVD are unknown. We used histological, molecular, genetic, pharmacological, and in vivo imaging approaches to characterize central nervous system (CNS) vascular pathologies in Col4a1 mutant mouse models of monogenic CSVD to provide insight into underlying pathogenic mechanisms. We describe developmental CNS angiogenesis abnormalities characterized by impaired retinal vascular outgrowth and patterning, increased numbers of mural cells with abnormal morphologies, altered contractile protein expression in vascular smooth muscle cells (VSMCs) and age-related loss of arteriolar VSMCs in Col4a1 mutant mice. Importantly, we identified elevated TGFβ signaling as a pathogenic consequence of Col4a1 mutations and show that genetically suppressing TGFβ signaling ameliorated CNS vascular pathologies, including partial rescue of retinal vascular patterning defects, prevention of VSMC loss, and significant reduction of intracerebral hemorrhages in Col4a1 mutant mice aged up to 8 months. This study identifies a novel biological role for collagen α1α1α2(IV) as a regulator of TGFβ signaling and demonstrates that elevated TGFβ signaling contributes to CNS vascular pathologies caused by Col4a1 mutations. Our findings suggest that pharmacologically suppressing TGFβ signaling could reduce the severity of CSVD, and potentially other manifestations associated with Gould syndrome and have important translational implications that could extend to idiopathic forms of CSVD.
Collapse
Affiliation(s)
- Kayla Branyan
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Cassandre Labelle-Dumais
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Xiaowei Wang
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Genki Hayashi
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Bryson Lee
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Zoe Peltz
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Seán Gorman
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Bo Qiao Li
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Mao Mao
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Douglas B Gould
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States; Department of Anatomy, Cardiovascular Research Institute, Bakar Aging Research Institute, and Institute for Human Genetics, University of California, San Francisco, United States.
| |
Collapse
|
24
|
A midposition NOTCH3 truncation in inherited cerebral small vessel disease may affect the protein interactome. J Biol Chem 2022; 299:102772. [PMID: 36470429 PMCID: PMC9808000 DOI: 10.1016/j.jbc.2022.102772] [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: 09/21/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 12/07/2022] Open
Abstract
Mutations in NOTCH3 underlie cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), the most common inherited cerebral small vessel disease. Two cleavages of NOTCH3 protein, at Asp80 and Asp121, were previously described in CADASIL pathological samples. Using monoclonal antibodies developed against a NOTCH3 neoepitope, we identified a third cleavage at Asp964 between an Asp-Pro sequence. We characterized the structural requirements for proteolysis at Asp964 and the vascular distribution of the cleavage event. A proteome-wide analysis was performed to find proteins that interact with the cleavage product. Finally, we investigated the biochemical determinants of this third cleavage event. Cleavage at Asp964 was critically dependent on the proline adjacent to the aspartate residue. In addition, the cleavage product was highly enriched in CADASIL brain tissue and localized to the media of degenerating arteries, where it deposited with the two additional NOTCH3 cleavage products. Recombinant NOTCH3 terminating at Asp964 was used to probe protein microarrays. We identified multiple molecules that bound to the cleaved NOTCH3 more than to uncleaved protein, suggesting that cleavage may alter the local protein interactome within disease-affected blood vessels. The cleavage of purified NOTCH3 protein at Asp964 in vitro was activated by reducing agents and NOTCH3 protein; cleavage was inhibited by specific dicarboxylic acids, as seen with cleavage at Asp80 and Asp121. Overall, we propose homologous redox-driven Asp-Pro cleavages and alterations in protein interactions as potential mechanisms in inherited small vessel disease; similarities in protein cleavage characteristics may indicate common biochemical modulators of pathological NOTCH3 processing.
Collapse
|
25
|
Seo MS, Kang M, An JR, Heo R, Jung WK, Choi IW, Han ET, Han JH, Chun W, Park WS. Asenapine, an atypical antipsychotic, blocks voltage-gated potassium channels in rabbit coronary artery smooth muscle cells. Eur J Pharmacol 2022; 934:175318. [DOI: 10.1016/j.ejphar.2022.175318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/18/2022] [Accepted: 10/03/2022] [Indexed: 11/30/2022]
|
26
|
Blackwell JA, Silva JF, Louis EM, Savu A, Largent-Milnes TM, Brooks HL, Pires PW. Cerebral arteriolar and neurovascular dysfunction after chemically induced menopause in mice. Am J Physiol Heart Circ Physiol 2022; 323:H845-H860. [PMID: 36149767 PMCID: PMC9602916 DOI: 10.1152/ajpheart.00276.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 12/14/2022]
Abstract
Cognitive decline is linked to decreased cerebral blood flow, particularly in women after menopause. Impaired cerebrovascular function precedes the onset of dementia, possibly because of reduced functional dilation in parenchymal arterioles. These vessels are bottlenecks of the cerebral microcirculation, and dysfunction can limit functional hyperemia in the brain. Large-conductance Ca2+-activated K+ channels (BKCa) are the final effectors of several pathways responsible for functional hyperemia, and their expression is modulated by estrogen. However, it remains unknown whether BKCa function is altered in cerebral parenchymal arterioles after menopause. Using a chemically induced model of menopause, the 4-vinylcyclohexene diepoxide (VCD) model, which depletes follicles while maintaining intact ovaries, we hypothesized that menopause would be associated with reduced functional vasodilatory responses in cerebral parenchymal arterioles of wild-type mice via reduced BKCa function. Using pressure myography of isolated parenchymal arterioles, we observed that menopause (Meno) induced a significant increase in spontaneous myogenic tone. Endothelial function, assessed as nitric oxide production and dilation after cholinergic stimulation or endothelium-dependent hyperpolarization pathways, was unaffected by Meno. BKCa function was significantly impaired in Meno compared with control, without changes in voltage-gated K+ channel activity. Cerebral functional hyperemia, measured by laser-speckle contrast imaging during whisker stimulation, was significantly blunted in Meno mice, without detectable changes in basal perfusion. However, behavioral testing identified no change in cognition. These findings suggest that menopause induces cerebral microvascular and neurovascular deficits.NEW & NOTEWORTHY Cerebral parenchymal arterioles from menopause mice showed increased myogenic tone. We identified an impairment in smooth muscle cell BKCa channel activity, without a reduction in endothelium-dependent dilation or nitric oxide production. Microvascular dysfunction was associated with a reduction in neurovascular responses after somatosensory stimulation. Despite the neurovascular impairment, cognitive abilities were maintained in menopausal mice.
Collapse
Affiliation(s)
- Jade A Blackwell
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Josiane F Silva
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Emma M Louis
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Andrea Savu
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Tally M Largent-Milnes
- Department of Pharmacology, University of Arizona, Tucson, Arizona
- Bio5 Institute, University of Arizona, Tucson, Arizona
| | - Heddwen L Brooks
- Department of Physiology, University of Arizona, Tucson, Arizona
- Bio5 Institute, University of Arizona, Tucson, Arizona
- Sarver Heart Center, University of Arizona, Tucson, Arizona
| | - Paulo W Pires
- Department of Physiology, University of Arizona, Tucson, Arizona
- Bio5 Institute, University of Arizona, Tucson, Arizona
- Sarver Heart Center, University of Arizona, Tucson, Arizona
| |
Collapse
|
27
|
Oka F, Lee JH, Yuzawa I, Li M, von Bornstaedt D, Eikermann-Haerter K, Qin T, Chung DY, Sadeghian H, Seidel JL, Imai T, Vuralli D, Platt RF, Nelson MT, Joutel A, Sakadzic S, Ayata C. CADASIL mutations sensitize the brain to ischemia via spreading depolarizations and abnormal extracellular potassium homeostasis. J Clin Invest 2022; 132:149759. [PMID: 35202003 PMCID: PMC9012276 DOI: 10.1172/jci149759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 02/23/2022] [Indexed: 11/17/2022] Open
Abstract
Cerebral autosomal dominant arteriopathy, subcortical infarcts and leukoencephalopathy (CADASIL) is the most common monogenic form of small vessel disease characterized by migraine with aura, leukoaraiosis, strokes and dementia. CADASIL mutations cause cerebrovascular dysfunction in both animal models and humans. Here, we show that two different human CADASIL mutations (Notch3 R90C or R169C) worsen ischemic stroke outcomes in transgenic mice, explained by a higher blood flow threshold to maintain tissue viability. Both mutants developed larger infarcts and worse neurological deficits compared with wild type regardless of age or sex after filament middle cerebral artery occlusion. However, full-field laser speckle flowmetry during distal middle cerebral artery occlusion showed comparable perfusion deficits in mutants and their respective wild type controls. Circle of Willis anatomy and pial collateralization also did not differ among the genotypes. In contrast, mutants had a higher cerebral blood flow threshold below which infarction ensued, suggesting increased sensitivity of brain tissue to ischemia. Electrophysiological recordings revealed a 1.5- to 2-fold higher frequency of peri-infarct spreading depolarizations in CADASIL mutants. Higher extracellular K+ elevations during spreading depolarizations in the mutants implicated a defect in extracellular K+ clearance. Altogether, these data reveal a novel mechanism of enhanced vulnerability to ischemic injury linked to abnormal extracellular ion homeostasis and susceptibility to ischemic depolarizations in CADASIL.
Collapse
Affiliation(s)
- Fumiaki Oka
- Department of Neurosurgery, Yamaguchi Graduate School of Medicine, Ube, Japan
| | - Jeong Hyun Lee
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Korea, Democratic Peoples Republic of
| | - Izumi Yuzawa
- Department of Radiology, Harvard Medical School and Massachusetts General Hospital, Charlestown, United States of America
| | - Mei Li
- Department of Radiology, Harvard Medical School and Massachusetts General Hospital, Charlestown, United States of America
| | - Daniel von Bornstaedt
- Department of Radiology, Harvard Medical School and Massachusetts General Hospital, Charlestown, United States of America
| | - Katharina Eikermann-Haerter
- Department of Radiology, Harvard Medical School and Massachusetts General Hospital, Charlestown, United States of America
| | - Tao Qin
- Department of Radiology, Harvard Medical School and Massachusetts General Hospital, Charlestown, United States of America
| | - David Y Chung
- Department of Radiology, Harvard Medical School and Massachusetts General Hospital, Charlestown, United States of America
| | - Homa Sadeghian
- Department of Radiology, Harvard Medical School and Massachusetts General Hospital, Charlestown, United States of America
| | - Jessica L Seidel
- Department of Radiology, Harvard Medical School and Massachusetts General Hospital, Charlestown, United States of America
| | - Takahiko Imai
- Department of Radiology, Harvard Medical School and Massachusetts General Hospital, Charlestown, United States of America
| | - Doga Vuralli
- Department of Radiology, Harvard Medical School and Massachusetts General Hospital, Charlestown, United States of America
| | - Rosangela Fm Platt
- Department of Radiology, Harvard Medical School and Massachusetts General Hospital, Charlestown, United States of America
| | - Mark T Nelson
- Department of Pharmacology, University of Vermont, Burlington, United States of America
| | - Anne Joutel
- Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université de Paris, GHU Paris Psychiatrie et Neurosciences, Paris, France
| | - Sava Sakadzic
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States of America
| | - Cenk Ayata
- Department of Radiology, Harvard Medical School and Massachusetts General Hospital, Charlestown, United States of America
| |
Collapse
|
28
|
Abstract
Cerebral small vessel disease (cSVD) is a leading cause of ischaemic and haemorrhagic stroke and a major contributor to dementia. Covert cSVD, which is detectable with brain MRI but does not manifest as clinical stroke, is highly prevalent in the general population, particularly with increasing age. Advances in technologies and collaborative work have led to substantial progress in the identification of common genetic variants that are associated with cSVD-related stroke (ischaemic and haemorrhagic) and MRI-defined covert cSVD. In this Review, we provide an overview of collaborative studies - mostly genome-wide association studies (GWAS) - that have identified >50 independent genetic loci associated with the risk of cSVD. We describe how these associations have provided novel insights into the biological mechanisms involved in cSVD, revealed patterns of shared genetic variation across cSVD traits, and shed new light on the continuum between rare, monogenic and common, multifactorial cSVD. We consider how GWAS summary statistics have been leveraged for Mendelian randomization studies to explore causal pathways in cSVD and provide genetic evidence for drug effects, and how the combination of findings from GWAS with gene expression resources and drug target databases has enabled identification of putative causal genes and provided proof-of-concept for drug repositioning potential. We also discuss opportunities for polygenic risk prediction, multi-ancestry approaches and integration with other omics data.
Collapse
|
29
|
Cognition, mood and behavior in CADASIL. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2022; 3:100043. [PMID: 36324403 PMCID: PMC9616390 DOI: 10.1016/j.cccb.2022.100043] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/06/2022] [Accepted: 02/06/2022] [Indexed: 01/02/2023]
Abstract
CADASIL is responsible for cognitive, mood or behavior disturbances. Cognitive disturbances range from moderate cognitive slowing to impairment of executive functions and may progress to a global decrease of cognitive efficiency up to severe dementia. Mood disturbances are extremely variable in intensity, depression is the most frequent symptom. Behavioral changes may occur at all stage of the disease, but are often associated with the onset of cognitive alterations. Apathy is the most prominent behavior alteration.
CADASIL is the most common familial cerebral small vessel disease (cSVD). Stereotyped mutations of the NOTCH3 gene are responsible for this archetypal ischemic cSVD that can lead, at the very end stage, to severe dementia. Variable cognitive alterations, mood, or behavior disturbances are frequently observed during the course of the disease. In this review, these clinical manifestations, their occurrence, severity and duration are analyzed in relation to the disease progression. Also, the potential relationships with cerebral lesions and treatment options are discussed.
Collapse
|
30
|
Schoemaker D, Arboleda-Velasquez JF. Notch3 Signaling and Aggregation as Targets for the Treatment of CADASIL and Other NOTCH3-Associated Small-Vessel Diseases. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1856-1870. [PMID: 33895122 PMCID: PMC8647433 DOI: 10.1016/j.ajpath.2021.03.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/28/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022]
Abstract
Mutations in the NOTCH3 gene can lead to small-vessel disease in humans, including the well-characterized cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), a condition caused by NOTCH3 mutations altering the number of cysteine residues in the extracellular domain of Notch3. Growing evidence indicates that other types of mutations in NOTCH3, including cysteine-sparing missense mutations or frameshift and premature stop codons, can lead to small-vessel disease phenotypes of variable severity or penetrance. There are currently no disease-modifying therapies for small-vessel disease, including those associated with NOTCH3 mutations. A deeper understanding of underlying molecular mechanisms and clearly defined targets are needed to promote the development of therapies. This review discusses two key pathophysiological mechanisms believed to contribute to the emergence and progression of small-vessel disease associated with NOTCH3 mutations: abnormal Notch3 aggregation and aberrant Notch3 signaling. This review offers a summary of the literature supporting and challenging the relevance of these mechanisms, together with an overview of available preclinical experiments derived from these mechanisms. It highlights knowledge gaps and future research directions. In view of recent evidence demonstrating the relatively high frequency of NOTCH3 mutations in the population, and their potential role in promoting small-vessel disease, progress in the development of therapies for NOTCH3-associated small-vessel disease is urgently needed.
Collapse
Affiliation(s)
- Dorothee Schoemaker
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Schepens Eye Research Institute of the Mass Eye and Ear and Department of Ophthalmology of Harvard Medical School, Boston, Massachusetts.
| | - Joseph F Arboleda-Velasquez
- Schepens Eye Research Institute of the Mass Eye and Ear and Department of Ophthalmology of Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
31
|
Claassen JAHR, Thijssen DHJ, Panerai RB, Faraci FM. Regulation of cerebral blood flow in humans: physiology and clinical implications of autoregulation. Physiol Rev 2021; 101:1487-1559. [PMID: 33769101 PMCID: PMC8576366 DOI: 10.1152/physrev.00022.2020] [Citation(s) in RCA: 448] [Impact Index Per Article: 112.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Brain function critically depends on a close matching between metabolic demands, appropriate delivery of oxygen and nutrients, and removal of cellular waste. This matching requires continuous regulation of cerebral blood flow (CBF), which can be categorized into four broad topics: 1) autoregulation, which describes the response of the cerebrovasculature to changes in perfusion pressure; 2) vascular reactivity to vasoactive stimuli [including carbon dioxide (CO2)]; 3) neurovascular coupling (NVC), i.e., the CBF response to local changes in neural activity (often standardized cognitive stimuli in humans); and 4) endothelium-dependent responses. This review focuses primarily on autoregulation and its clinical implications. To place autoregulation in a more precise context, and to better understand integrated approaches in the cerebral circulation, we also briefly address reactivity to CO2 and NVC. In addition to our focus on effects of perfusion pressure (or blood pressure), we describe the impact of select stimuli on regulation of CBF (i.e., arterial blood gases, cerebral metabolism, neural mechanisms, and specific vascular cells), the interrelationships between these stimuli, and implications for regulation of CBF at the level of large arteries and the microcirculation. We review clinical implications of autoregulation in aging, hypertension, stroke, mild cognitive impairment, anesthesia, and dementias. Finally, we discuss autoregulation in the context of common daily physiological challenges, including changes in posture (e.g., orthostatic hypotension, syncope) and physical activity.
Collapse
Affiliation(s)
- Jurgen A H R Claassen
- Department of Geriatrics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, The Netherlands
| | - Dick H J Thijssen
- Department of Physiology, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Ronney B Panerai
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- >National Institute for Health Research Leicester Biomedical Research Centre, University of Leicester, Leicester, United Kingdom
| | - Frank M Faraci
- Departments of Internal Medicine, Neuroscience, and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
32
|
Rosehart AC, Longden TA, Weir N, Fontaine JT, Joutel A, Dabertrand F. Prostaglandin E 2 Dilates Intracerebral Arterioles When Applied to Capillaries: Implications for Small Vessel Diseases. Front Aging Neurosci 2021; 13:695965. [PMID: 34483880 PMCID: PMC8414797 DOI: 10.3389/fnagi.2021.695965] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/15/2021] [Indexed: 11/24/2022] Open
Abstract
Prostaglandin E2 (PGE2) has been widely proposed to mediate neurovascular coupling by dilating brain parenchymal arterioles through activation of prostanoid EP4 receptors. However, our previous report that direct application of PGE2 induces an EP1-mediated constriction strongly argues against its direct action on arterioles during neurovascular coupling, the mechanisms sustaining functional hyperemia. Recent advances have highlighted the role of capillaries in sensing neuronal activity and propagating vasodilatory signals to the upstream penetrating parenchymal arteriole. Here, we examined the effect of capillary stimulation with PGE2 on upstream arteriolar diameter using an ex vivo capillary-parenchymal arteriole preparation and in vivo cerebral blood flow measurements with two-photon laser-scanning microscopy. We found that PGE2 caused upstream arteriolar dilation when applied onto capillaries with an EC50 of 70 nM. The response was inhibited by EP1 receptor antagonist and was greatly reduced, but not abolished, by blocking the strong inward-rectifier K+ channel. We further observed a blunted dilatory response to capillary stimulation with PGE2 in a genetic mouse model of cerebral small vessel disease with impaired functional hyperemia. This evidence casts previous findings in a different light, indicating that capillaries are the locus of PGE2 action to induce upstream arteriolar dilation in the control of brain blood flow, thereby providing a paradigm-shifting view that nonetheless remains coherent with the broad contours of a substantial body of existing literature.
Collapse
Affiliation(s)
- Amanda C. Rosehart
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Thomas A. Longden
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Nick Weir
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Jackson T. Fontaine
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Anne Joutel
- INSERM, UMR 1266, GHU Paris Psychiatrie et Neurosciences, Institute of Psychiatry and Neuroscience of Paris, University of Paris, Paris, France
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Fabrice Dabertrand
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
33
|
PIP 2 corrects cerebral blood flow deficits in small vessel disease by rescuing capillary Kir2.1 activity. Proc Natl Acad Sci U S A 2021; 118:2025998118. [PMID: 33875602 PMCID: PMC8092380 DOI: 10.1073/pnas.2025998118] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cerebral small vessel diseases (SVDs) are a central link between stroke and dementia-two comorbidities without specific treatments. Despite the emerging consensus that SVDs are initiated in the endothelium, the early mechanisms remain largely unknown. Deficits in on-demand delivery of blood to active brain regions (functional hyperemia) are early manifestations of the underlying pathogenesis. The capillary endothelial cell strong inward-rectifier K+ channel Kir2.1, which senses neuronal activity and initiates a propagating electrical signal that dilates upstream arterioles, is a cornerstone of functional hyperemia. Here, using a genetic SVD mouse model, we show that impaired functional hyperemia is caused by diminished Kir2.1 channel activity. We link Kir2.1 deactivation to depletion of phosphatidylinositol 4,5-bisphosphate (PIP2), a membrane phospholipid essential for Kir2.1 activity. Systemic injection of soluble PIP2 rapidly restored functional hyperemia in SVD mice, suggesting a possible strategy for rescuing functional hyperemia in brain disorders in which blood flow is disturbed.
Collapse
|
34
|
van den Brink H, Kopczak A, Arts T, Onkenhout L, Siero JC, Zwanenburg JJ, Duering M, Blair GW, Doubal FN, Stringer MS, Thrippleton MJ, Kuijf HJ, de Luca A, Hendrikse J, Wardlaw JM, Dichgans M, Biessels GJ. Zooming in on cerebral small vessel function in small vessel diseases with 7T MRI: Rationale and design of the "ZOOM@SVDs" study. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2021; 2:100013. [PMID: 36324717 PMCID: PMC9616370 DOI: 10.1016/j.cccb.2021.100013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 06/01/2023]
Abstract
Background Cerebral small vessel diseases (SVDs) are a major cause of stroke and dementia. Yet, specific treatment strategies are lacking in part because of a limited understanding of the underlying disease processes. There is therefore an urgent need to study SVDs at their core, the small vessels themselves. Objective This paper presents the rationale and design of the ZOOM@SVDs study, which aims to establish measures of cerebral small vessel dysfunction on 7T MRI as novel disease markers of SVDs. Methods ZOOM@SVDs is a prospective observational cohort study with two years follow-up. ZOOM@SVDs recruits participants with Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL, N = 20), sporadic SVDs (N = 60), and healthy controls (N = 40). Participants undergo 7T brain MRI to assess different aspects of small vessel function including small vessel reactivity, cerebral perforating artery flow, and pulsatility. Extensive work-up at baseline and follow-up further includes clinical and neuropsychological assessment as well as 3T brain MRI to assess conventional SVD imaging markers. Measures of small vessel dysfunction are compared between patients and controls, and related to the severity of clinical and conventional MRI manifestations of SVDs. Discussion ZOOM@SVDs will deliver novel markers of cerebral small vessel function in patients with monogenic and sporadic forms of SVDs, and establish their relation with disease burden and progression. These small vessel markers can support etiological studies in SVDs and may serve as surrogate outcome measures in future clinical trials to show target engagement of drugs directed at the small vessels.
Collapse
Key Words
- ASL, Arterial Spin Labeling
- BOLD, Blood Oxygenation Level-Dependent
- CADASIL
- CADASIL, Cerebral Autosomal Dominant Arteriopathy with Leukoencephalopathy and Subcortical Infarcts
- CDR, Clinical Dementia Rating scale
- CERAD+, Consortium to Establish a Disease Registry for Alzheimer's Disease Plus battery
- CES-D, Center for Epidemiologic Studies Depression Scale
- CO2, Carbon Dioxide
- CSF, Cerebrospinal Fluid
- Cerebral small vessel disease
- DTI, Diffusion Tensor Imaging
- EPIC, European Prospective Investigation into Cancer and Nutrition
- EtCO2, End-tidal Carbon Dioxide
- FLAIR, Fluid Attenuated Inversion Recovery
- FOV, Field Of View
- FWHM, Full-Width-at-Half-Maximum
- GE, Gradient Echo
- GM, Grey Matter
- GPRS, General Packet Radio Service
- HRF, Hemodynamic Response Function
- High field strength MRI
- LMU, Ludwig-Maximilians-Universität
- MMSE, Mini-Mental State Examination
- NAWM, Normal Appearing White Matter
- NIHSS, National Institute for Health Stroke Scale
- PI, Pulsatility Index
- ROI, Region Of Interest
- SPPB, Short Physical Performance Battery
- SVDs, Small Vessel Diseases
- SWI, Susceptibility Weighted Imaging
- Small vessel function
- Sporadic SVD
- Stroke
- TE, Echo Time
- TI, Inversion Time
- TR, Repetition Time
- TSE, Turbo Spin Echo
- UMCU, University Medical Center Utrecht
- Vmax, Maximum velocity
- Vmean, Mean velocity
- Vmin, Minimum velocity
- WM, White Matter
- WMH, White Matter Hyperintensity
- fMRI, Functional Magnetic Resonance Imaging
Collapse
Affiliation(s)
- Hilde van den Brink
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3508 GA, the Netherlands
| | - Anna Kopczak
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Tine Arts
- Department of Radiology, Center for Image Sciences, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Laurien Onkenhout
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3508 GA, the Netherlands
| | - Jeroen C.W. Siero
- Department of Radiology, Center for Image Sciences, University Medical Center Utrecht, Utrecht, the Netherlands
- Spinoza Centre for Neuroimaging Amsterdam, Amsterdam, the Netherlands
| | - Jaco J.M. Zwanenburg
- Department of Radiology, Center for Image Sciences, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marco Duering
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Disease (DZNE), Munich, Germany
| | - Gordon W. Blair
- Brain Research Imaging Centre, Centre for Clinical Brain Sciences, UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, United Kingdom
| | - Fergus N. Doubal
- Brain Research Imaging Centre, Centre for Clinical Brain Sciences, UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, United Kingdom
| | - Michael S. Stringer
- Brain Research Imaging Centre, Centre for Clinical Brain Sciences, UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, United Kingdom
| | - Michael J. Thrippleton
- Brain Research Imaging Centre, Centre for Clinical Brain Sciences, UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, United Kingdom
| | - Hugo J. Kuijf
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Alberto de Luca
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3508 GA, the Netherlands
- Brain Research Imaging Centre, Centre for Clinical Brain Sciences, UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, United Kingdom
| | - Jeroen Hendrikse
- Department of Radiology, Center for Image Sciences, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Joanna M. Wardlaw
- Brain Research Imaging Centre, Centre for Clinical Brain Sciences, UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, United Kingdom
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Disease (DZNE), Munich, Germany
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3508 GA, the Netherlands
| | - SVDs@target group
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3508 GA, the Netherlands
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
- Department of Radiology, Center for Image Sciences, University Medical Center Utrecht, Utrecht, the Netherlands
- Spinoza Centre for Neuroimaging Amsterdam, Amsterdam, the Netherlands
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Disease (DZNE), Munich, Germany
- Brain Research Imaging Centre, Centre for Clinical Brain Sciences, UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, United Kingdom
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
35
|
Neves KB, Morris HE, Alves-Lopes R, Muir KW, Moreton F, Delles C, Montezano AC, Touyz RM. Peripheral arteriopathy caused by Notch3 gain-of-function mutation involves ER and oxidative stress and blunting of NO/sGC/cGMP pathway. Clin Sci (Lond) 2021; 135:753-773. [PMID: 33681964 DOI: 10.1042/cs20201412] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/24/2021] [Accepted: 03/08/2021] [Indexed: 12/30/2022]
Abstract
Notch3 mutations cause Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL), which predisposes to stroke and dementia. CADASIL is characterised by vascular dysfunction and granular osmiophilic material (GOM) accumulation in cerebral small vessels. Systemic vessels may also be impacted by Notch3 mutations. However vascular characteristics and pathophysiological processes remain elusive. We investigated mechanisms underlying the peripheral vasculopathy mediated by CADASIL-causing Notch3 gain-of-function mutation. We studied: (i) small arteries and vascular smooth muscle cells (VSMCs) from TgNotch3R169C mice (CADASIL model), (ii) VSMCs from peripheral arteries from CADASIL patients, and (iii) post-mortem brains from CADASIL individuals. TgNotch3R169C vessels exhibited GOM deposits, increased vasoreactivity and impaired vasorelaxation. Hypercontractile responses were normalised by fasudil (Rho kinase inhibitor) and 4-phenylbutyrate (4-PBA; endoplasmic-reticulum (ER) stress inhibitor). Ca2+ transients and Ca2+ channel expression were increased in CADASIL VSMCs, with increased expression of Rho guanine nucleotide-exchange factors (GEFs) and ER stress proteins. Vasorelaxation mechanisms were impaired in CADASIL, evidenced by decreased endothelial nitric oxide synthase (eNOS) phosphorylation and reduced cyclic guanosine 3',5'-monophosphate (cGMP) levels, with associated increased soluble guanylate cyclase (sGC) oxidation, decreased sGC activity and reduced levels of the vasodilator hydrogen peroxide (H2O2). In VSMCs from CADASIL patients, sGC oxidation was increased and cGMP levels decreased, effects normalised by fasudil and 4-PBA. Cerebral vessels in CADASIL patients exhibited significant oxidative damage. In conclusion, peripheral vascular dysfunction in CADASIL is associated with altered Ca2+ homoeostasis, oxidative stress and blunted eNOS/sGC/cGMP signaling, processes involving Rho kinase and ER stress. We identify novel pathways underlying the peripheral arteriopathy induced by Notch3 gain-of-function mutation, phenomena that may also be important in cerebral vessels.
Collapse
Affiliation(s)
- Karla B Neves
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Hannah E Morris
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Rhéure Alves-Lopes
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Keith W Muir
- Institute of Neuroscience and Psychology, University of Glasgow and Queen Elizabeth University Hospital, Glasgow, U.K
| | - Fiona Moreton
- Institute of Neuroscience and Psychology, University of Glasgow and Queen Elizabeth University Hospital, Glasgow, U.K
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| |
Collapse
|
36
|
Manini A, Pantoni L. CADASIL from Bench to Bedside: Disease Models and Novel Therapeutic Approaches. Mol Neurobiol 2021; 58:2558-2573. [PMID: 33464533 PMCID: PMC8128844 DOI: 10.1007/s12035-021-02282-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/05/2021] [Indexed: 12/21/2022]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is a monogenic disease caused by NOTCH3 mutations and characterized by typical clinical, neuroradiological, and pathological features. NOTCH3 belongs to a family of highly conserved transmembrane receptors rich of epidermal growth factor repeats, mostly expressed in vascular smooth muscle cells and pericytes, which perform essential developmental functions and are involved in tissues maintenance and renewal. To date, no therapeutic option for CADASIL is available except for few symptomatic treatments. Novel in vitro and in vivo models are continuously explored with the aim to investigate underlying pathogenic mechanisms and to test novel therapeutic approaches. In this scenario, knock-out, knock-in, and transgenic mice studies have generated a large amount of information on molecular and biological aspects of CADASIL, despite that they incompletely reproduce the human phenotype. Moreover, the field of in vitro models has been revolutionized in the last two decades by the introduction of induced pluripotent stem cells (iPSCs) technology. As a consequence, novel therapeutic approaches, including immunotherapy, growth factors administration, and antisense oligonucleotides, are currently under investigation. While waiting that further studies confirm the promising results obtained, the data reviewed suggest that our therapeutic approach to the disease could be transformed, generating new hope for the future.
Collapse
Affiliation(s)
- Arianna Manini
- Stroke and Dementia Lab, "Luigi Sacco" Department of Biomedical and Clinical Sciences, University of Milan, Via Giovanni Battista Grassi 74, 20157, Milano, Italy
| | - Leonardo Pantoni
- Stroke and Dementia Lab, "Luigi Sacco" Department of Biomedical and Clinical Sciences, University of Milan, Via Giovanni Battista Grassi 74, 20157, Milano, Italy.
| |
Collapse
|
37
|
Hariharan A, Weir N, Robertson C, He L, Betsholtz C, Longden TA. The Ion Channel and GPCR Toolkit of Brain Capillary Pericytes. Front Cell Neurosci 2020; 14:601324. [PMID: 33390906 PMCID: PMC7775489 DOI: 10.3389/fncel.2020.601324] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
Brain pericytes reside on the abluminal surface of capillaries, and their processes cover ~90% of the length of the capillary bed. These cells were first described almost 150 years ago (Eberth, 1871; Rouget, 1873) and have been the subject of intense experimental scrutiny in recent years, but their physiological roles remain uncertain and little is known of the complement of signaling elements that they employ to carry out their functions. In this review, we synthesize functional data with single-cell RNAseq screens to explore the ion channel and G protein-coupled receptor (GPCR) toolkit of mesh and thin-strand pericytes of the brain, with the aim of providing a framework for deeper explorations of the molecular mechanisms that govern pericyte physiology. We argue that their complement of channels and receptors ideally positions capillary pericytes to play a central role in adapting blood flow to meet the challenge of satisfying neuronal energy requirements from deep within the capillary bed, by enabling dynamic regulation of their membrane potential to influence the electrical output of the cell. In particular, we outline how genetic and functional evidence suggest an important role for Gs-coupled GPCRs and ATP-sensitive potassium (KATP) channels in this context. We put forth a predictive model for long-range hyperpolarizing electrical signaling from pericytes to upstream arterioles, and detail the TRP and Ca2+ channels and Gq, Gi/o, and G12/13 signaling processes that counterbalance this. We underscore critical questions that need to be addressed to further advance our understanding of the signaling topology of capillary pericytes, and how this contributes to their physiological roles and their dysfunction in disease.
Collapse
Affiliation(s)
- Ashwini Hariharan
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Nick Weir
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Colin Robertson
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Liqun He
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Christer Betsholtz
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Medicine Huddinge (MedH), Karolinska Institutet & Integrated Cardio Metabolic Centre, Huddinge, Sweden
| | - Thomas A Longden
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
38
|
Quick S, Moss J, Rajani RM, Williams A. A Vessel for Change: Endothelial Dysfunction in Cerebral Small Vessel Disease. Trends Neurosci 2020; 44:289-305. [PMID: 33308877 DOI: 10.1016/j.tins.2020.11.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/24/2020] [Accepted: 11/11/2020] [Indexed: 01/08/2023]
Abstract
The blood vessels of the brain are lined with endothelial cells and it has been long known that these help to regulate blood flow to the brain. However, there is increasing evidence that these cells also interact with the surrounding brain tissue. These interactions change when the endothelial cells become dysfunctional and have an impact in diseases such as cerebral small vessel disease, the leading cause of vascular dementia. In this review, we focus on what endothelial dysfunction is, what causes it, how it leads to surrounding brain pathology, how researchers can investigate it with current models, and where this might lead in the future for dementia therapies.
Collapse
Affiliation(s)
- Sophie Quick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Jonathan Moss
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Rikesh M Rajani
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Anna Williams
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK.
| |
Collapse
|
39
|
Fontaine JT, Rosehart AC, Joutel A, Dabertrand F. HB-EGF depolarizes hippocampal arterioles to restore myogenic tone in a genetic model of small vessel disease. Mech Ageing Dev 2020; 192:111389. [PMID: 33127441 PMCID: PMC7683376 DOI: 10.1016/j.mad.2020.111389] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/16/2020] [Accepted: 10/22/2020] [Indexed: 12/26/2022]
Abstract
Vascular cognitive impairment, the second most common cause of dementia, profoundly affects hippocampal-dependent functions. However, while the growing literature covers complex neuronal interactions, little is known about the sustaining hippocampal microcirculation. Here we examined vasoconstriction to physiological pressures of hippocampal arterioles, a fundamental feature of small arteries, in a genetic mouse model of CADASIL, an archetypal cerebral small vessel disease. Using diameter and membrane potential recordings on isolated arterioles, we observed both blunted pressure-induced vasoconstriction and smooth muscle cell depolarization in CADASIL. This impairment was abolished in the presence of voltage-gated potassium (KV1) channel blocker 4-aminopyridine, or by application of heparin-binding EGF-like growth factor (HB-EGF), which promotes KV1 channel down-regulations. Interestingly, we observed that HB-EGF induced a depolarization of the myocyte plasma membrane within the arteriolar wall in CADASIL, but not wild-type, arterioles. Collectively, our results indicate that hippocampal arterioles in CADASIL mice display a blunted contractile response to luminal pressure, similar to the defect we previously reported in cortical arterioles and pial arteries, that is rescued by HB-EGF. Hippocampal vascular dysfunction in CADASIL could then contribute to the decreased vascular reserve associated with decreased cognitive performance, and its correction may provide a therapeutic option for treating vascular cognitive impairment.
Collapse
Affiliation(s)
- Jackson T Fontaine
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Amanda C Rosehart
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Anne Joutel
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT, USA; Institute of Psychiatry and Neurosciences of Paris, INSERM UMR1266, University of Paris, GHU Paris Psychiatrie et Neurosciences, France
| | - Fabrice Dabertrand
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
40
|
Ghali MGZ, Marchenko V, Yaşargil MG, Ghali GZ. Structure and function of the perivascular fluid compartment and vertebral venous plexus: Illumining a novel theory on mechanisms underlying the pathogenesis of Alzheimer's, cerebral small vessel, and neurodegenerative diseases. Neurobiol Dis 2020; 144:105022. [PMID: 32687942 DOI: 10.1016/j.nbd.2020.105022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 06/13/2020] [Accepted: 07/15/2020] [Indexed: 01/14/2023] Open
Abstract
Blood dynamically and richly supplies the cerebral tissue via microvessels invested in pia matter perforating the cerebral substance. Arteries penetrating the cerebral substance derive an investment from one or two successive layers of pia mater, luminally apposed to the pial-glial basal lamina of the microvasculature and abluminally apposed to a series of aquaporin IV-studded astrocytic end feet constituting the soi-disant glia limitans. The full investment of successive layers forms the variably continuous walls of the periarteriolar, pericapillary, and perivenular divisions of the perivascular fluid compartment. The pia matter disappears at the distal periarteriolar division of the perivascular fluid compartment. Plasma from arteriolar blood sequentially transudates into the periarteriolar division of the perivascular fluid compartment and subarachnoid cisterns in precession to trickling into the neural interstitium. Fluid from the neural interstitium successively propagates into the venules through the subarachnoid cisterns and perivenular division of the perivascular fluid compartment. Fluid fluent within the perivascular fluid compartment flows gegen the net direction of arteriovenular flow. Microvessel oscillations at the central tendency of the cerebral vasomotion generate corresponding oscillations of within the surrounding perivascular fluid compartment, interposed betwixt the abluminal surface of the vessels and internal surface of the pia mater. The precise microanatomy of this most fascinating among designable spaces has eluded the efforts of various investigators to interrogate its structure, though most authors non-consensusly concur the investing layers effectively and functionally segregate the perivascular and subarachnoid fluid compartments. Enlargement of the perivascular fluid compartment in a variety of neurological disorders, including senile dementia of the Alzheimer's type and cerebral small vessel disease, may alternately or coordinately constitute a correlative marker of disease severity and a possible cause implicated in the mechanistic pathogenesis of these conditions. Venular pressures modulating oscillatory dynamic flow within the perivascular fluid compartment may similarly contribute to the development of a variety among neurological disorders. An intimate understanding of subtle features typifying microanatomy and microphysiology of the investing structures and spaces of the cerebral microvasculature may powerfully inform mechanistic pathophysiology mediating a variety of neurovascular ischemic, neuroinfectious, neuroautoimmune, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Michael George Zaki Ghali
- Department of Neurological Surgery, University of California San Francisco, 505 Parnassus Street, San Francisco, CA 94143, United States; Department of Neurobiology and Anatomy, 2900 W. Queen Lane, Philadelphia, PA 19129, United States.
| | - Vitaliy Marchenko
- Department of Neurobiology and Anatomy, 2900 W. Queen Lane, Philadelphia, PA 19129, United States; Department of Neurophysiology, Bogomoletz Institute, Kyiv, Ukraine; Department of Neuroscience, Московский государственный университет имени М. В., Ломоносова GSP-1, Leninskie Gory, Moscow 119991, Russian Federation
| | - M Gazi Yaşargil
- Department of Neurosurgery, University Hospital Zurich Rämistrasse 100, 8091 Zurich, Switzerland
| | - George Zaki Ghali
- United States Environmental Protection Agency, Arlington, Virginia, USA; Emeritus Professor of Toxicology, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
41
|
The capillary Kir channel as sensor and amplifier of neuronal signals: Modeling insights on K +-mediated neurovascular communication. Proc Natl Acad Sci U S A 2020; 117:16626-16637. [PMID: 32601236 DOI: 10.1073/pnas.2000151117] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Neuronal activity leads to an increase in local cerebral blood flow (CBF) to allow adequate supply of oxygen and nutrients to active neurons, a process termed neurovascular coupling (NVC). We have previously shown that capillary endothelial cell (cEC) inwardly rectifying K+ (Kir) channels can sense neuronally evoked increases in interstitial K+ and induce rapid and robust dilations of upstream parenchymal arterioles, suggesting a key role of cECs in NVC. The requirements of this signal conduction remain elusive. Here, we utilize mathematical modeling to investigate how small outward currents in stimulated cECs can elicit physiologically relevant spread of vasodilatory signals within the highly interconnected brain microvascular network to increase local CBF. Our model shows that the Kir channel can act as an "on-off" switch in cECs to hyperpolarize the cell membrane as extracellular K+ increases. A local hyperpolarization can be amplified by the voltage-dependent activation of Kir in neighboring cECs. Sufficient Kir density enables robust amplification of the hyperpolarizing stimulus and produces responses that resemble action potentials in excitable cells. This Kir-mediated excitability can remain localized in the stimulated region or regeneratively propagate over significant distances in the microvascular network, thus dramatically increasing the efficacy of K+ for eliciting local hyperemia. Modeling results show how changes in cEC transmembrane current densities and gap junctional resistances can affect K+-mediated NVC and suggest a key role for Kir as a sensor of neuronal activity and an amplifier of retrograde electrical signaling in the cerebral vasculature.
Collapse
|
42
|
Chabriat H, Joutel A, Tournier‐Lasserve E, Bousser MG. CADASIL: yesterday, today, tomorrow. Eur J Neurol 2020; 27:1588-1595. [DOI: 10.1111/ene.14293] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 04/28/2020] [Indexed: 12/27/2022]
Affiliation(s)
- H. Chabriat
- Department of Neurology and CERVCO Reference Center for Rare Vascular Diseases of the Eye and Brain Hôpital Lariboisiére, APHP Paris France
- INSERM U 1141 Paris France
- University of Paris Paris France
| | - A. Joutel
- University of Paris Paris France
- Institute of Psychiatry and Neurosciences of Paris INSERM U1266 Paris France
| | - E. Tournier‐Lasserve
- INSERM U 1141 Paris France
- University of Paris Paris France
- Molecular Genetics Department and CERVCO Reference Center for Rare Vascular Diseases of the Eye and Brain Hopital Lariboisiére, APHP Paris France
| | - M. G. Bousser
- Department of Neurology and CERVCO Reference Center for Rare Vascular Diseases of the Eye and Brain Hôpital Lariboisiére, APHP Paris France
- University of Paris Paris France
| |
Collapse
|
43
|
Joutel A. Prospects for Diminishing the Impact of Nonamyloid Small-Vessel Diseases of the Brain. Annu Rev Pharmacol Toxicol 2020; 60:437-456. [PMID: 31425001 DOI: 10.1146/annurev-pharmtox-010818-021712] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Small-vessel diseases (SVDs) of the brain are involved in about one-fourth of ischemic strokes and a vast majority of intracerebral hemorrhages and are responsible for nearly half of dementia cases in the elderly. SVDs are a heavy burden for society, a burden that is expected to increase further in the absence of significant therapeutic advances, given the aging population. Here, we provide a critical appraisal of currently available therapeutic approaches for nonamyloid sporadic SVDs that are largely based on targeting modifiable risk factors. We review what is known about the pathogenic mechanisms of vascular risk factor-related SVDs and cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), the most frequent hereditary SVD, and elaborate on two mechanism-based therapeutic approaches worth exploring in sporadic SVD and CADASIL. We conclude by discussing opportunities and challenges that need to be tackled if efforts to achieve significant therapeutic advances for these diseases are to be successful.
Collapse
Affiliation(s)
- Anne Joutel
- Institute of Psychiatry and Neurosciences of Paris, INSERM UMR1266, Paris Descartes University, 75014 Paris, France; .,DHU NeuroVasc, Sorbonne Paris Cité, 75010 Paris, France.,Department of Pharmacology, College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| |
Collapse
|
44
|
Rosehart AC, Johnson AC, Dabertrand F. Ex Vivo Pressurized Hippocampal Capillary-Parenchymal Arteriole Preparation for Functional Study. J Vis Exp 2019. [PMID: 31904015 DOI: 10.3791/60676] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
From subtle behavioral alterations to late-stage dementia, vascular cognitive impairment typically develops following cerebral ischemia. Stroke and cardiac arrest are remarkably sexually dimorphic diseases, and both induce cerebral ischemia. However, progress in understanding the vascular cognitive impairment, and then developing sex-specific treatments, has been partly limited by challenges in investigating the brain microcirculation from mouse models in functional studies. Here, we present an approach to examine the capillary-to-arteriole signaling in an ex vivo hippocampal capillary-parenchymal arteriole (HiCaPA) preparation from mouse brain. We describe how to isolate, cannulate, and pressurize the microcirculation to measure arteriolar diameter in response to capillary stimulation. We show which appropriate functional controls can be used to validate the HiCaPA preparation integrity and display typical results, including testing potassium as a neurovascular coupling agent and the effect of the recently characterized inhibitor of the Kir2 inward rectifying potassium channel family, ML133. Further, we compare the responses in preparations obtained from male and female mice. While these data reflect functional investigations, our approach can also be used in molecular biology, immunochemistry, and electrophysiology studies.
Collapse
Affiliation(s)
- Amanda C Rosehart
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus
| | - Abbie C Johnson
- Department of Neurological Sciences, University of Vermont Larner College of Medicine
| | - Fabrice Dabertrand
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus; Department of Pharmacology, University of Colorado Anschutz Medical Campus;
| |
Collapse
|
45
|
Neves KB, Harvey AP, Moreton F, Montezano AC, Rios FJ, Alves-Lopes R, Nguyen Dinh Cat A, Rocchicciolli P, Delles C, Joutel A, Muir K, Touyz RM. ER stress and Rho kinase activation underlie the vasculopathy of CADASIL. JCI Insight 2019; 4:131344. [PMID: 31647781 PMCID: PMC6962020 DOI: 10.1172/jci.insight.131344] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 10/18/2019] [Indexed: 12/21/2022] Open
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) leads to premature stroke and vascular dementia. Mechanism-specific therapies for this aggressive cerebral small vessel disease are lacking. CADASIL is caused by NOTCH3 mutations that influence vascular smooth muscle cell (VSMC) function through unknown processes. We investigated molecular mechanisms underlying the vasculopathy in CADASIL focusing on endoplasmic reticulum (ER) stress and RhoA/Rho kinase (ROCK). Peripheral small arteries and VSMCs were isolated from gluteal biopsies of CADASIL patients and mesentery of TgNotch3R169C mice (CADASIL model). CADASIL vessels exhibited impaired vasorelaxation, blunted vasoconstriction, and hypertrophic remodeling. Expression of NOTCH3 and ER stress target genes was amplified and ER stress response, Rho kinase activity, superoxide production, and cytoskeleton-associated protein phosphorylation were increased in CADASIL, processes associated with Nox5 upregulation. Aberrant vascular responses and signaling in CADASIL were ameliorated by inhibitors of Notch3 (γ-secretase inhibitor), Nox5 (mellitin), ER stress (4-phenylbutyric acid), and ROCK (fasudil). Observations in human CADASIL were recapitulated in TgNotch3R169C mice. These findings indicate that vascular dysfunction in CADASIL involves ER stress/ROCK interplay driven by Notch3-induced Nox5 activation and that NOTCH3 mutation-associated vascular pathology, typical in cerebral vessels, also manifests peripherally. We define Notch3-Nox5/ER stress/ROCK signaling as a putative mechanism-specific target and suggest that peripheral artery responses may be an accessible biomarker in CADASIL.
Collapse
Affiliation(s)
- Karla B. Neves
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - Adam P. Harvey
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - Fiona Moreton
- Institute of Neuroscience and Psychology, University of Glasgow and Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Augusto C. Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - Francisco J. Rios
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - Rhéure Alves-Lopes
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | | | | | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - Anne Joutel
- Institute of Psychiatry and Neurosciences of Paris Inserm, Paris Descartes University, Paris, France
| | - Keith Muir
- Institute of Neuroscience and Psychology, University of Glasgow and Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Rhian M. Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
- Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
46
|
Abstract
Cerebral small vessel disease (SVD) is characterized by changes in the pial and parenchymal microcirculations. SVD produces reductions in cerebral blood flow and impaired blood-brain barrier function, which are leading contributors to age-related reductions in brain health. End-organ effects are diverse, resulting in both cognitive and noncognitive deficits. Underlying phenotypes and mechanisms are multifactorial, with no specific treatments at this time. Despite consequences that are already considerable, the impact of SVD is predicted to increase substantially with the growing aging population. In the face of this health challenge, the basic biology, pathogenesis, and determinants of SVD are poorly defined. This review summarizes recent progress and concepts in this area, highlighting key findings and some major unanswered questions. We focus on phenotypes and mechanisms that underlie microvascular aging, the greatest risk factor for cerebrovascular disease and its subsequent effects.
Collapse
Affiliation(s)
- T Michael De Silva
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne Campus, Bundoora, Victoria 3086, Australia;
| | - Frank M Faraci
- Departments of Internal Medicine, Neuroscience, and Pharmacology, Francois M. Abboud Cardiovascular Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA;
| |
Collapse
|
47
|
Coupland K, Lendahl U, Karlström H. Role of NOTCH3 Mutations in the Cerebral Small Vessel Disease Cerebral Autosomal Dominant Arteriopathy With Subcortical Infarcts and Leukoencephalopathy. Stroke 2019; 49:2793-2800. [PMID: 30355220 DOI: 10.1161/strokeaha.118.021560] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Kirsten Coupland
- From the Department of Neurobiology, Care Sciences, and Society (K.C., H.K.), Karolinska Institutet Stockholm, Sweden
| | - Urban Lendahl
- Department of Cell and Molecular Biology (U.L.), Karolinska Institutet Stockholm, Sweden
| | - Helena Karlström
- From the Department of Neurobiology, Care Sciences, and Society (K.C., H.K.), Karolinska Institutet Stockholm, Sweden
| |
Collapse
|
48
|
Ling C, Zhang Z, Wu Y, Fang X, Kong Q, Zhang W, Wang Z, Yang Q, Yuan Y. Reduced Venous Oxygen Saturation Associates With Increased Dependence of Patients With Cerebral Autosomal Dominant Arteriopathy With Subcortical Infarcts and Leukoencephalopathy: A 7.0-T Magnetic Resonance Imaging Study. Stroke 2019; 50:3128-3134. [PMID: 31514698 DOI: 10.1161/strokeaha.119.026376] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose- Accumulating evidence has demonstrated hemodynamic abnormalities and cerebral hypoperfusion in patients with cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). Increased venous susceptibility assessed by susceptibility weighted imaging and mapping has been shown to indicate a decrease in venous oxygen saturation. This study aimed to investigate whether altered venous oxygen saturation is related to clinical phenotypes of CADASIL patients. Methods- Using 7.0-T susceptibility weighted imaging and mapping, we compared venous susceptibility of cortical veins between 41 CADASIL patients and 43 age- and sex-matched healthy controls. The magnetic resonance imaging lesion load, mini-mental state examination score, Barthel Index, and modified Rankin Scale were examined in the patient group, and the correlations between venous susceptibility and clinical characteristics were analyzed. Results- Venous susceptibility increased with age (r=0.508, P=0.001) and was higher in CADASIL patients than in healthy controls (t=-4.673; P<0.001). We found a positive association between venous susceptibility and the age-related white matter change scores (r=0.364; P=0.019), number of lacunar infarctions (r=0.520; P<0.001), number of cerebral microbleeds (ρ=0.445; P=0.004), and small-vessel disease scores (ρ=0.465; P=0.002) in CADASIL patients. Moreover, increased venous susceptibility was associated with higher modified Rankin Scale scores in CADASIL patients after adjustment for age- and small-vessel disease scores (odds ratio=3.178; 95% CI, 1.101-9.179; P=0.033). Conclusions- Our findings indicate that extensive cerebral hypoperfusion may induce central nervous system impairment in CADASIL, and susceptibility weighted imaging and mapping could be used clinically to assess the condition of CADASIL patients.
Collapse
Affiliation(s)
- Chen Ling
- From the Department of Neurology, Peking University First Hospital, Beijing, China (C.L., X.F., W.Z., Z.W., Y.Y.)
| | - Zihao Zhang
- From the Department of Neurology, Peking University First Hospital, Beijing, China (C.L., X.F., W.Z., Z.W., Y.Y.)
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China (Z.Z., Y.W., Q.K.)
- CAS Center for Excellence in Brain Science and Intelligence Technology, Beijing, China (Z.Z., Y.W., Q.K.)
- University of Chinese Academy of Sciences, Beijing, China (Z.Z., Y.W., Q.K.)
| | - Yue Wu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China (Z.Z., Y.W., Q.K.)
- CAS Center for Excellence in Brain Science and Intelligence Technology, Beijing, China (Z.Z., Y.W., Q.K.)
- University of Chinese Academy of Sciences, Beijing, China (Z.Z., Y.W., Q.K.)
| | - Xiaojing Fang
- Department of Neurology, Peking University International Hospital, Beijing, China (X.F.)
| | - Qingle Kong
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China (Z.Z., Y.W., Q.K.)
- CAS Center for Excellence in Brain Science and Intelligence Technology, Beijing, China (Z.Z., Y.W., Q.K.)
- University of Chinese Academy of Sciences, Beijing, China (Z.Z., Y.W., Q.K.)
| | - Wei Zhang
- From the Department of Neurology, Peking University First Hospital, Beijing, China (C.L., X.F., W.Z., Z.W., Y.Y.)
| | - Zhaoxia Wang
- From the Department of Neurology, Peking University First Hospital, Beijing, China (C.L., X.F., W.Z., Z.W., Y.Y.)
| | - Qi Yang
- Department of Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China (Q.Y.)
| | - Yun Yuan
- From the Department of Neurology, Peking University First Hospital, Beijing, China (C.L., X.F., W.Z., Z.W., Y.Y.)
| |
Collapse
|
49
|
Altered Brain Glucose Metabolism Assessed by 18F-FDG PET Imaging Is Associated with the Cognitive Impairment of CADASIL. Neuroscience 2019; 417:35-44. [PMID: 31394195 DOI: 10.1016/j.neuroscience.2019.07.048] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/26/2022]
Abstract
Recurrent stroke and cognitive impairment are the primary features of patients with cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). The cognitive deficits in these patients are known to be correlated with structural brain changes, such as white matter lesions and lacunae, and resting-state functional connectivity in brain networks. However, the associations between changes in brain glucose metabolism based on 18F-2-fluoro-2-deoxy-D-glucose (18F-FDG) positron emission tomography (PET) imaging and cognitive scores in CADASIL patients remain unclear. In the present study, 24 CADASIL patients and 24 matched healthy controls underwent 18F-FDG PET imaging. Brain glucose metabolism was measured in all subjects and Pearson's correlation analyses were performed to evaluate relationships between abnormal glucose metabolism in various brain areas and cognitive scores. Compared to controls, CADASIL patients exhibited significantly lower metabolism in the right cerebellar posterior lobe, left cerebellar anterior lobe, bilateral thalamus and left limbic lobe. Additionally, hypermetabolism was observed in the left precentral and postcentral gyri. Importantly, glucose metabolism in the left limbic lobe was positively associated with cognitive scores on the Mini-Mental State Examination (MMSE). Furthermore, glucose metabolism in the left precentral gyri was negatively correlated with cognitive scores on the Montreal Cognitive Assessment (MoCA). The present findings provide strong support for the presence of altered brain glucose metabolism in CADASIL patients as well as the associations between abnormal metabolism and cognitive scales in this population. The present findings suggest that patterns of brain glucose metabolism may become useful markers of cognitive impairment in CADASIL patients.
Collapse
|
50
|
Frösen J, Joutel A. Smooth muscle cells of intracranial vessels: from development to disease. Cardiovasc Res 2019; 114:501-512. [PMID: 29351598 DOI: 10.1093/cvr/cvy002] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 01/12/2018] [Indexed: 02/02/2023] Open
Abstract
Cerebrovascular diseases that cause ischaemic or haemorrhagic stroke with subsequent loss of life or functional capacity due to damage of the brain tissue are among the leading causes of human suffering and economic burden inflicted by diseases in the developed world. Diseases affecting intracranial vessels are significant contributors to ischaemic and haemorrhagic strokes. Brain arteriovenous malformations, which are a collection of abnormal blood vessels connecting arteries to veins, are the most common cause of intracranial haemorrhage in children and young adults. Saccular intracranial aneurysms, which are pathological saccular dilations mainly occurring at bifurcations of the large intracranial arteries near the circle of Willis, are highly prevalent in the middle-aged population, causing significant anxiety and concern; their rupture, although rare, is a significant cause of intracranial haemorrhage in those past middle age that is associated with a very sinister prognosis. Cerebral small-vessel disease, which comprise all pathological processes affecting vessels <500 microns in diameter, account for the majority of intracerebral haemorrhages and ∼25% of ischaemic strokes and 45% of dementias in the elderly. In this review, we summarize the developmental, structural, and functional features of intracranial vessels. We then describe the role of smooth muscle cells in brain arteriovenous malformations, intracranial aneurysms, and small-vessel diseases, and discuss how the peculiar ontogeny, structure, and function of intracranial vessels are related to the development of these diseases.
Collapse
Affiliation(s)
- Juhana Frösen
- Hemorrhagic Brain Pathology Research Group, NeuroCenter, Kuopio University Hospital, Kuopio 70029, Finland.,Department of Neurosurgery, Kuopio University Hospital, Kuopio 70029, Finland
| | - Anne Joutel
- Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, Université Paris Diderot-Paris 7, 10 av de Verdun, Paris 75010, France.,DHU NeuroVasc, Sorbonne Paris Cité, Paris 75010, France
| |
Collapse
|