1
|
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
|
2
|
Levites Y, Dammer EB, Ran Y, Tsering W, Duong D, Abreha M, Gadhavi J, Lolo K, Trejo-Lopez J, Phillips J, Iturbe A, Erquizi A, Moore BD, Ryu D, Natu A, Dillon K, Torrellas J, Moran C, Ladd T, Afroz F, Islam T, Jagirdar J, Funk CC, Robinson M, Rangaraju S, Borchelt DR, Ertekin-Taner N, Kelly JW, Heppner FL, Johnson ECB, McFarland K, Levey AI, Prokop S, Seyfried NT, Golde TE. Integrative proteomics identifies a conserved Aβ amyloid responsome, novel plaque proteins, and pathology modifiers in Alzheimer's disease. Cell Rep Med 2024; 5:101669. [PMID: 39127040 PMCID: PMC11384960 DOI: 10.1016/j.xcrm.2024.101669] [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: 08/03/2023] [Revised: 04/15/2024] [Accepted: 07/10/2024] [Indexed: 08/12/2024]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder that develops over decades. AD brain proteomics reveals vast alterations in protein levels and numerous altered biologic pathways. Here, we compare AD brain proteome and network changes with the brain proteomes of amyloid β (Aβ)-depositing mice to identify conserved and divergent protein networks with the conserved networks identifying an Aβ amyloid responsome. Proteins in the most conserved network (M42) accumulate in plaques, cerebrovascular amyloid (CAA), and/or dystrophic neuronal processes, and overexpression of two M42 proteins, midkine (Mdk) and pleiotrophin (PTN), increases the accumulation of Aβ in plaques and CAA. M42 proteins bind amyloid fibrils in vitro, and MDK and PTN co-accumulate with cardiac transthyretin amyloid. M42 proteins appear intimately linked to amyloid deposition and can regulate amyloid deposition, suggesting that they are pathology modifiers and thus putative therapeutic targets. We posit that amyloid-scaffolded accumulation of numerous M42+ proteins is a central mechanism mediating downstream pathophysiology in AD.
Collapse
Affiliation(s)
- Yona Levites
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric B Dammer
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA, USA; Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Yong Ran
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Wangchen Tsering
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL, USA; Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Duc Duong
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA, USA; Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Measho Abreha
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA, USA; Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Joshna Gadhavi
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA, USA; Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Kiara Lolo
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL, USA; Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Jorge Trejo-Lopez
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL, USA; Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Jennifer Phillips
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL, USA; Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Andrea Iturbe
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA; Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Aya Erquizi
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA; Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Brenda D Moore
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Danny Ryu
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Aditya Natu
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA, USA; Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Kristy Dillon
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA; Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jose Torrellas
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA; Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Corey Moran
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA; Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Thomas Ladd
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| | - Farhana Afroz
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Tariful Islam
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Jaishree Jagirdar
- Department of Pathology and Laboratory Medicine, Emory University Hospital, Atlanta, GA, USA
| | - Cory C Funk
- Institute for Systems Biology, Seattle, WA, USA
| | | | | | - David R Borchelt
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA; Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Nilüfer Ertekin-Taner
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL, USA; Mayo Clinic, Department of Neurology, Jacksonville, FL, USA
| | - Jeffrey W Kelly
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Frank L Heppner
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 110117 Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE) Berlin, 110117 Berlin, Germany; Cluster of Excellence, NeuroCure, Charitéplatz, 110117 Berlin, Germany
| | - Erik C B Johnson
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA; Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Karen McFarland
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Allan I Levey
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA; Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Stefan Prokop
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL, USA; Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA; Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA.
| | - Nicholas T Seyfried
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA, USA; Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, USA.
| | - Todd E Golde
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA; Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
3
|
Beaufort N, Ingendahl L, Merdanovic M, Schmidt A, Podlesainski D, Richter T, Neumann T, Kuszner M, Vetter IR, Stege P, Burston SG, Filipovic A, Ruiz-Blanco YB, Bravo-Rodriguez K, Mieres-Perez J, Beuck C, Uebel S, Zobawa M, Schillinger J, Malik R, Todorov-Völgyi K, Rey J, Roberti A, Hagemeier B, Wefers B, Müller SA, Wurst W, Sanchez-Garcia E, Zimmermann A, Hu XY, Clausen T, Huber R, Lichtenthaler SF, Schmuck C, Giese M, Kaiser M, Ehrmann M, Dichgans M. Rational correction of pathogenic conformational defects in HTRA1. Nat Commun 2024; 15:5944. [PMID: 39013852 PMCID: PMC11252331 DOI: 10.1038/s41467-024-49982-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/24/2024] [Indexed: 07/18/2024] Open
Abstract
Loss-of-function mutations in the homotrimeric serine protease HTRA1 cause cerebral vasculopathy. Here, we establish independent approaches to achieve the functional correction of trimer assembly defects. Focusing on the prototypical R274Q mutation, we identify an HTRA1 variant that promotes trimer formation thus restoring enzymatic activity in vitro. Genetic experiments in Htra1R274Q mice further demonstrate that expression of this protein-based corrector in trans is sufficient to stabilize HtrA1-R274Q and restore the proteomic signature of the brain vasculature. An alternative approach employs supramolecular chemical ligands that shift the monomer-trimer equilibrium towards proteolytically active trimers. Moreover, we identify a peptidic ligand that activates HTRA1 monomers. Our findings open perspectives for tailored protein repair strategies.
Collapse
Affiliation(s)
- Nathalie Beaufort
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Linda Ingendahl
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Melisa Merdanovic
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Andree Schmidt
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), LMU Munich, Munich, Germany
| | - David Podlesainski
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Tim Richter
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Thorben Neumann
- Organic Chemistry, Faculty of Chemistry, University Duisburg-Essen, Essen, Germany
| | - Michael Kuszner
- Center of Medical Biotechnology, Faculty of Chemistry, University Duisburg-Essen, Essen, Germany
| | - Ingrid R Vetter
- Max-Planck-Institute of Molecular Physiology, Dortmund, Germany
| | - Patricia Stege
- Max-Planck-Institute of Molecular Physiology, Dortmund, Germany
| | - Steven G Burston
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, Bristol, UK
| | - Anto Filipovic
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Yasser B Ruiz-Blanco
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Kenny Bravo-Rodriguez
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
- Max-Planck-Institute of Molecular Physiology, Dortmund, Germany
| | - Joel Mieres-Perez
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
- Department of Biochemical and Chemical Engineering, Technical University Dortmund, Dortmund, Germany
| | - Christine Beuck
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Stephan Uebel
- Max-Planck-Institute of Biochemistry, Martinsried, Germany
| | - Monika Zobawa
- Max-Planck-Institute of Biochemistry, Martinsried, Germany
| | - Jasmin Schillinger
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Katalin Todorov-Völgyi
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Juliana Rey
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Annabell Roberti
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Birte Hagemeier
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Benedikt Wefers
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute of Developmental Genetics (IDG), Helmholtz Zentrum München, Neuherberg, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Wolfgang Wurst
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute of Developmental Genetics (IDG), Helmholtz Zentrum München, Neuherberg, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Technische Universität München-Weihenstephan, Freising, Germany
| | - Elsa Sanchez-Garcia
- Department of Biochemical and Chemical Engineering, Technical University Dortmund, Dortmund, Germany
| | - Alexander Zimmermann
- Center of Medical Biotechnology, Faculty of Chemistry, University Duisburg-Essen, Essen, Germany
| | - Xiao-Yu Hu
- College of Material Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing, China
| | - Tim Clausen
- Research Institute of Molecular Pathology (IMP), Vienna, Austria
| | - Robert Huber
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
- Max-Planck-Institute of Biochemistry, Martinsried, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Carsten Schmuck
- Center of Medical Biotechnology, Faculty of Chemistry, University Duisburg-Essen, Essen, Germany
| | - Michael Giese
- Organic Chemistry, Faculty of Chemistry, University Duisburg-Essen, Essen, Germany
| | - Markus Kaiser
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Michael Ehrmann
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany.
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University of Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
4
|
Boston G, Jobson D, Mizuno T, Ihara M, Kalaria RN. Most common NOTCH3 mutations causing CADASIL or CADASIL-like cerebral small vessel disease: A systematic review. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2024; 6:100227. [PMID: 38966425 PMCID: PMC11223087 DOI: 10.1016/j.cccb.2024.100227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 05/09/2024] [Accepted: 05/30/2024] [Indexed: 07/06/2024]
Abstract
Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL) is a monogenic disorder caused by mutations in the NOTCH3 gene. The main aim of our survey was to determine if there is an association between phenotypes and genotypes across the most common NOTCH3 mutations found in CADASIL patients. We systematically searched clinical studies and genomic databases from 1996 to 2023 to first identify the most common mutations responsible for CADASIL. We found the six most common NOTCH3 missense mutations globally were the p.R75P, p.R133C, p.R141C, p.R169C, p.R182C, and p.R544C, of which p.R133C was described to occur most often. Focusing on studies with comprehensive clinical records, our analysis further suggested that the p.R75P, p.R141C, p.R182C and p.R544C genotypes were highly congruent with the presence of white matter hyperintensities on magnetic resonance imaging (MRI), which was the most common phenotypic characteristic across all four mutations. We found the p.R141C mutation was associated with increased severity of disease. We also found the average age of onset in p.R544C carriers was more than a decade later compared to the p.R141C carriers. However, statistical analysis showed there were no overall differences between the phenotypic characteristics of the two common mutations, p.R141C and p.R544C. Geographically, China and Japan were the only two countries to report all the four common mutations vis a vis p.R75P, p.R141C, p.R182C and p.R544C. There is a possibility that this is due to a combination of a founder effect, but there also could be sampling biases.
Collapse
Affiliation(s)
- Georgina Boston
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Dan Jobson
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Toshiki Mizuno
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Centre, Osaka, Japan
| | - Raj N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
5
|
Malik R, Beaufort N, Li J, Tanaka K, Georgakis MK, He Y, Koido M, Terao C, Japan B, Anderson CD, Kamatani Y, Zand R, Dichgans M. Genetically proxied HTRA1 protease activity and circulating levels independently predict risk of ischemic stroke and coronary artery disease. NATURE CARDIOVASCULAR RESEARCH 2024; 3:701-713. [PMID: 39196222 DOI: 10.1038/s44161-024-00475-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/23/2024] [Indexed: 08/29/2024]
Abstract
Genetic variants in HTRA1 are associated with stroke risk. However, the mechanisms mediating this remain largely unknown, as does the full spectrum of phenotypes associated with genetic variation in HTRA1. Here we show that rare HTRA1 variants are linked to ischemic stroke in the UK Biobank and BioBank Japan. Integrating data from biochemical experiments, we next show that variants causing loss of protease function associated with ischemic stroke, coronary artery disease and skeletal traits in the UK Biobank and MyCode cohorts. Moreover, a common variant modulating circulating HTRA1 mRNA and protein levels enhances the risk of ischemic stroke and coronary artery disease while lowering the risk of migraine and macular dystrophy in genome-wide association study, UK Biobank, MyCode and BioBank Japan data. We found no interaction between proxied HTRA1 activity and levels. Our findings demonstrate the role of HTRA1 for cardiovascular diseases and identify two mechanisms as potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Nathalie Beaufort
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Jiang Li
- Department of Molecular and Functional Genomics, Geisinger Health System, Danville, PA, USA
| | - Koki Tanaka
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Marios K Georgakis
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Yunye He
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Masaru Koido
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - BioBank Japan
- Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Christopher D Anderson
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Boston, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Yoichiro Kamatani
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Ramin Zand
- Department of Neurology, Pennsylvania State University, Hershey, PA, USA
- Department of Neurology, Neuroscience Institute, Geisinger Health System, Danville, PA, USA
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University of Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- German Center for Cardiovascular Research (DZHK), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
6
|
Menéndez-Valladares P, Acevedo Aguilera R, Núñez-Jurado D, López Azcárate C, Domínguez Mayoral AM, Fernández-Vega A, Pérez-Sánchez S, Lamana Vallverdú M, García-Sánchez MI, Morales Bravo M, Busquier T, Montaner J. A Search for New Biological Pathways in Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy by Proteomic Research. J Clin Med 2024; 13:3138. [PMID: 38892848 PMCID: PMC11172732 DOI: 10.3390/jcm13113138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Background/Objectives: Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL) is a hereditary small vessel disease leading to significant morbidity and mortality. Despite advances in genetic diagnosis, the underlying pathophysiology remains incompletely understood. Proteomic studies offer insights into disease mechanisms by identifying altered protein expression patterns. Here, we conducted a proteomic analysis to elucidate molecular pathways associated with CADASIL. Methods: We enrolled genetically diagnosed CADASIL patients and healthy, genetically related controls. Plasma samples were subjected to proteomic analysis using the Olink platform, measuring 552 proteins across six panels. The data were analyzed from several approaches by using three different statistical methods: Exploratory Principal Component Analysis (PCA) and Partial Least Squares-Discriminant Analysis (PLS-DA), differential expression with moderated t-test, and gene set enrichment analysis (GSEA). In addition, bioinformatics analysis, including volcano plot, heatmap, and Variable Importance on Projection (VIP) scores from the PLS-DA model were drawn. Results: Significant differences in protein expression were observed between CADASIL patients and controls. RSPO1 and FGF-19 exhibited elevated levels (p < 0.05), while PPY showed downregulation (p < 0.05) in CADASIL patients, suggesting their involvement in disease pathogenesis. Furthermore, MIC-A/B expression varied significantly between patients with mutations in exon 4 versus exon 11 of the NOTCH3 gene (p < 0.05), highlighting potential immunological mechanisms underlying CADASIL. We identified altered pathways using GSEA, applied after ranking the study data. Conclusions: Our study provides novel insights into the proteomic profile of CADASIL, identifying dysregulated proteins associated with vascular pathology, metabolic dysregulation, and immune activation. These findings contribute to a deeper understanding of CADASIL pathophysiology and may inform the development of targeted therapeutic strategies. Further research is warranted to validate these biomarkers and elucidate their functional roles in disease progression.
Collapse
Affiliation(s)
- Paloma Menéndez-Valladares
- Department of Neurology, Virgen Macarena University Hospital, 41009 Seville, Spain; (P.M.-V.); (R.A.A.); (D.N.-J.); (C.L.A.); (S.P.-S.); (M.L.V.); (M.M.B.); (J.M.)
- Department of Neurology, Institute of Biomedicine of Seville (IBIS), 41013 Seville, Spain
- Department of Clinical Biochemistry, Virgen Macarena University Hospital, 41009 Seville, Spain
- Commission of Neurochemistry and Neurological Diseases, Spanish Society of Laboratory Medicine, 08025 Barcelona, Spain
| | - Rosa Acevedo Aguilera
- Department of Neurology, Virgen Macarena University Hospital, 41009 Seville, Spain; (P.M.-V.); (R.A.A.); (D.N.-J.); (C.L.A.); (S.P.-S.); (M.L.V.); (M.M.B.); (J.M.)
- Department of Neurology, Institute of Biomedicine of Seville (IBIS), 41013 Seville, Spain
| | - David Núñez-Jurado
- Department of Neurology, Virgen Macarena University Hospital, 41009 Seville, Spain; (P.M.-V.); (R.A.A.); (D.N.-J.); (C.L.A.); (S.P.-S.); (M.L.V.); (M.M.B.); (J.M.)
- Department of Neurology, Institute of Biomedicine of Seville (IBIS), 41013 Seville, Spain
- Department of Clinical Biochemistry, Virgen Macarena University Hospital, 41009 Seville, Spain
| | - Cristina López Azcárate
- Department of Neurology, Virgen Macarena University Hospital, 41009 Seville, Spain; (P.M.-V.); (R.A.A.); (D.N.-J.); (C.L.A.); (S.P.-S.); (M.L.V.); (M.M.B.); (J.M.)
- Department of Neurology, Institute of Biomedicine of Seville (IBIS), 41013 Seville, Spain
| | - Ana María Domínguez Mayoral
- Department of Neurology, Virgen Macarena University Hospital, 41009 Seville, Spain; (P.M.-V.); (R.A.A.); (D.N.-J.); (C.L.A.); (S.P.-S.); (M.L.V.); (M.M.B.); (J.M.)
- Department of Neurology, Institute of Biomedicine of Seville (IBIS), 41013 Seville, Spain
| | - Alejandro Fernández-Vega
- Department of Neurology, Virgen Macarena University Hospital, 41009 Seville, Spain; (P.M.-V.); (R.A.A.); (D.N.-J.); (C.L.A.); (S.P.-S.); (M.L.V.); (M.M.B.); (J.M.)
- Department of Neurology, Institute of Biomedicine of Seville (IBIS), 41013 Seville, Spain
| | - Soledad Pérez-Sánchez
- Department of Neurology, Virgen Macarena University Hospital, 41009 Seville, Spain; (P.M.-V.); (R.A.A.); (D.N.-J.); (C.L.A.); (S.P.-S.); (M.L.V.); (M.M.B.); (J.M.)
- Department of Neurology, Institute of Biomedicine of Seville (IBIS), 41013 Seville, Spain
| | - Marcel Lamana Vallverdú
- Department of Neurology, Virgen Macarena University Hospital, 41009 Seville, Spain; (P.M.-V.); (R.A.A.); (D.N.-J.); (C.L.A.); (S.P.-S.); (M.L.V.); (M.M.B.); (J.M.)
- Department of Neurology, Institute of Biomedicine of Seville (IBIS), 41013 Seville, Spain
| | | | - María Morales Bravo
- Department of Neurology, Virgen Macarena University Hospital, 41009 Seville, Spain; (P.M.-V.); (R.A.A.); (D.N.-J.); (C.L.A.); (S.P.-S.); (M.L.V.); (M.M.B.); (J.M.)
- Department of Neurology, Institute of Biomedicine of Seville (IBIS), 41013 Seville, Spain
| | - Teresa Busquier
- Department of Radiology, Virgen Macarena University Hospital, 41009 Seville, Spain;
| | - Joan Montaner
- Department of Neurology, Virgen Macarena University Hospital, 41009 Seville, Spain; (P.M.-V.); (R.A.A.); (D.N.-J.); (C.L.A.); (S.P.-S.); (M.L.V.); (M.M.B.); (J.M.)
- Department of Neurology, Institute of Biomedicine of Seville (IBIS), 41013 Seville, Spain
| |
Collapse
|
7
|
Song S, Li X, Xue X, Dong W, Li C. Progress in the Study of the Role and Mechanism of HTRA1 in Diseases Related to Vascular Abnormalities. Int J Gen Med 2024; 17:1479-1491. [PMID: 38650587 PMCID: PMC11034561 DOI: 10.2147/ijgm.s456912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
High temperature requirement A1 (HTRA1) is a member of the serine protease family, comprising four structural domains: IGFBP domain, Kazal domain, protease domain and PDZ domain. HTRA1 encodes a serine protease, a secreted protein that is widely expressed in the vasculature. HTRA1 regulates a wide range of physiological processes through its proteolytic activity, and is also involved in a variety of vascular abnormalities-related diseases. This article reviews the role of HTRA1 in the development of vascular abnormalities-related hereditary cerebral small vessel disease (CSVD), age-related macular degeneration (AMD), tumors and other diseases. Through relevant research advances to understand the role of HTRA1 in regulating signaling pathways or refolding, translocation, degradation of extracellular matrix (ECM) proteins, thus directly or indirectly regulating angiogenesis, vascular remodeling, and playing an important role in vascular homeostasis, further understanding the mechanism of HTRA1's role in vascular abnormality-related diseases is important for HTRA1 to be used as a therapeutic target in related diseases.
Collapse
Affiliation(s)
- Shina Song
- Department of Neurology, The First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
- Department of Geriatrics, General Hospital of TISCO, Taiyuan, People’s Republic of China
| | - Xiaofeng Li
- Department of Neurology, The First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Xuting Xue
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, The First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Wenping Dong
- Department of Geriatrics, General Hospital of TISCO, Taiyuan, People’s Republic of China
| | - Changxin Li
- Department of Neurology, The First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| |
Collapse
|
8
|
Todorov-Völgyi K, González-Gallego J, Müller SA, Beaufort N, Malik R, Schifferer M, Todorov MI, Crusius D, Robinson S, Schmidt A, Körbelin J, Bareyre F, Ertürk A, Haass C, Simons M, Paquet D, Lichtenthaler SF, Dichgans M. Proteomics of mouse brain endothelium uncovers dysregulation of vesicular transport pathways during aging. NATURE AGING 2024; 4:595-612. [PMID: 38519806 DOI: 10.1038/s43587-024-00598-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/20/2024] [Indexed: 03/25/2024]
Abstract
Age-related decline in brain endothelial cell (BEC) function contributes critically to neurological disease. Comprehensive atlases of the BEC transcriptome have become available, but results from proteomic profiling are lacking. To gain insights into endothelial pathways affected by aging, we developed a magnetic-activated cell sorting-based mouse BEC enrichment protocol compatible with proteomics and resolved the profiles of protein abundance changes during aging. Unsupervised cluster analysis revealed a segregation of age-related protein dynamics with biological functions, including a downregulation of vesicle-mediated transport. We found a dysregulation of key regulators of endocytosis and receptor recycling (most prominently Arf6), macropinocytosis and lysosomal degradation. In gene deletion and overexpression experiments, Arf6 affected endocytosis pathways in endothelial cells. Our approach uncovered changes not picked up by transcriptomic studies, such as accumulation of vesicle cargo and receptor ligands, including Apoe. Proteomic analysis of BECs from Apoe-deficient mice revealed a signature of accelerated aging. Our findings provide a resource for analysing BEC function during aging.
Collapse
Affiliation(s)
- Katalin Todorov-Völgyi
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
| | - Judit González-Gallego
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Graduate School of Systemic Neuroscience (GSN), University Hospital, LMU Munich, Munich, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Nathalie Beaufort
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mihail Ivilinov Todorov
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Zentrum München, Neuherberg, Germany
| | - Dennis Crusius
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Sophie Robinson
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Graduate School of Systemic Neuroscience (GSN), University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Andree Schmidt
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jakob Körbelin
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florence Bareyre
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Ali Ertürk
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Zentrum München, Neuherberg, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Division of Metabolic Biochemistry, Biomedical Center Munich (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Mikael Simons
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Dominik Paquet
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
9
|
Kastberger B, Winter S, Brandstätter H, Biller J, Wagner W, Plesnila N. Treatment with Cerebrolysin Prolongs Lifespan in a Mouse Model of Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy. Adv Biol (Weinh) 2024; 8:e2300439. [PMID: 38062874 DOI: 10.1002/adbi.202300439] [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: 08/22/2023] [Indexed: 02/15/2024]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is a rare familial neurological disorder caused by mutations in the NOTCH3 gene and characterized by migraine attacks, depressive episodes, lacunar strokes, dementia, and premature death. Since there is no therapy for CADASIL the authors investigate whether the multi-modal neuropeptide drug Cerebrolysin may improve outcome in a murine CADASIL model. Twelve-month-old NOTCH3R169C mutant mice (n=176) are treated for nine weeks with Cerebrolysin or Vehicle and histopathological and functional outcomes are evaluated within the subsequent ten months. Cerebrolysin treatment improves spatial memory and overall health, reduces epigenetic aging, and prolongs lifespan, however, CADASIL-specific white matter vacuolization is not affected. On the molecular level Cerebrolysin treatment increases expression of Calcitonin Gene-Related Peptide (CGRP) and Silent Information Regulator Two (Sir2)-like protein 6 (SIRT6), decreases expression of Insulin-like Growth Factor 1 (IGF-1), and normalizes the expression of neurovascular laminin. In summary, Cerebrolysin fosters longevity and healthy aging without specifically affecting CADASIL pathology. Hence, Cerebrolysin may serve a therapeutic option for CADASIL and other disorders characterized by accelerated aging.
Collapse
Affiliation(s)
| | - Stefan Winter
- Ever Pharma, Oberburgau 3, Unterach am Attersee, 4866, Austria
| | | | - Janina Biller
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377, Munich, Germany
| | - Wolfgang Wagner
- Institute for Stem Cell Biology, RWTH Aachen University Medical School, 52074, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany
- Cygenia GmbH, 52078, Aachen, Germany
| | - Nikolaus Plesnila
- Cluster of Systems Neurology (Synergy), 81377, Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377, Munich, Germany
| |
Collapse
|
10
|
Mizuta I, Nakao-Azuma Y, Yoshida H, Yamaguchi M, Mizuno T. Progress to Clarify How NOTCH3 Mutations Lead to CADASIL, a Hereditary Cerebral Small Vessel Disease. Biomolecules 2024; 14:127. [PMID: 38254727 PMCID: PMC10813265 DOI: 10.3390/biom14010127] [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: 12/08/2023] [Revised: 01/09/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
Notch signaling is conserved in C. elegans, Drosophila, and mammals. Among the four NOTCH genes in humans, NOTCH1, NOTCH2, and NOTCH3 are known to cause monogenic hereditary disorders. Most NOTCH-related disorders are congenital and caused by a gain or loss of Notch signaling activity. In contrast, cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) caused by NOTCH3 is adult-onset and considered to be caused by accumulation of the mutant NOTCH3 extracellular domain (N3ECD) and, possibly, by an impairment in Notch signaling. Pathophysiological processes following mutant N3ECD accumulation have been intensively investigated; however, the process leading to N3ECD accumulation and its association with canonical NOTCH3 signaling remain unknown. We reviewed the progress in clarifying the pathophysiological process involving mutant NOTCH3.
Collapse
Affiliation(s)
- Ikuko Mizuta
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan; (I.M.)
| | - Yumiko Nakao-Azuma
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan; (I.M.)
- Department of Rehabilitation Medicine, Gunma University Graduate School of Medicine, Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Hideki Yoshida
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Masamitsu Yamaguchi
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
- Kansai Gakken Laboratory, Kankyo Eisei Yakuhin Co., Ltd., 3-6-2 Hikaridai, Seika-cho, Kyoto 619-0237, Japan
| | - Toshiki Mizuno
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan; (I.M.)
| |
Collapse
|
11
|
Levites Y, Dammer EB, Ran Y, Tsering W, Duong D, Abreha M, Gadhavi J, Lolo K, Trejo-Lopez J, Phillips JL, Iturbe A, Erqiuzi A, Moore BD, Ryu D, Natu A, Dillon KD, Torrellas J, Moran C, Ladd TB, Afroz KF, Islam T, Jagirdar J, Funk CC, Robinson M, Borchelt DR, Ertekin-Taner N, Kelly JW, Heppner FL, Johnson EC, McFarland K, Levey AL, Prokop S, Seyfried NT, Golde TE. Aβ Amyloid Scaffolds the Accumulation of Matrisome and Additional Proteins in Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.29.568318. [PMID: 38076912 PMCID: PMC10705437 DOI: 10.1101/2023.11.29.568318] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
We report a highly significant correlation in brain proteome changes between Alzheimers disease (AD) and CRND8 APP695NL/F transgenic mice. However, integrating protein changes observed in the CRND8 mice with co-expression networks derived from human AD, reveals both conserved and divergent module changes. For the most highly conserved module (M42, matrisome) we find many proteins accumulate in plaques, cerebrovascular amyloid (CAA), dystrophic processes, or a combination thereof. Overexpression of two M42 proteins, midkine (Mdk) and pleiotrophin (PTN), in CRND8 mice brains leads to increased accumulation of A β ; in plaques and in CAA; further, recombinant MDK and PTN enhance A β ; aggregation into amyloid. Multiple M42 proteins, annotated as heparan sulfate binding proteins, bind to fibrillar A β 42 and a non-human amyloid fibril in vitro. Supporting this binding data, MDK and PTN co-accumulate with transthyretin (TTR) amyloid in the heart and islet amyloid polypeptide (IAPP) amyloid in the pancreas. Our findings establish several critical insights. Proteomic changes in modules observed in human AD brains define an A β ; amyloid responsome that is well conserved from mouse model to human. Further, distinct amyloid structures may serve as scaffolds, facilitating the co-accumulation of proteins with signaling functions. We hypothesize that this co-accumulation may contribute to downstream pathological sequalae. Overall, this contextualized understanding of proteomic changes and their interplay with amyloid deposition provides valuable insights into the complexity of AD pathogenesis and potential biomarkers and therapeutic targets.
Collapse
|
12
|
Haqqani AS, Mianoor Z, Star AT, Detcheverry FE, Delaney CE, Stanimirovic DB, Hamel E, Badhwar A. Proteome Profiling of Brain Vessels in a Mouse Model of Cerebrovascular Pathology. BIOLOGY 2023; 12:1500. [PMID: 38132326 PMCID: PMC10740654 DOI: 10.3390/biology12121500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023]
Abstract
Cerebrovascular pathology that involves altered protein levels (or signaling) of the transforming growth factor beta (TGFβ) family has been associated with various forms of age-related dementias, including Alzheimer disease (AD) and vascular cognitive impairment and dementia (VCID). Transgenic mice overexpressing TGFβ1 in the brain (TGF mice) recapitulate VCID-associated cerebrovascular pathology and develop cognitive deficits in old age or when submitted to comorbid cardiovascular risk factors for dementia. We characterized the cerebrovascular proteome of TGF mice using mass spectrometry (MS)-based quantitative proteomics. Cerebral arteries were surgically removed from 6-month-old-TGF and wild-type mice, and proteins were extracted and analyzed by gel-free nanoLC-MS/MS. We identified 3602 proteins in brain vessels, with 20 demonstrating significantly altered levels in TGF mice. For total and/or differentially expressed proteins (p ≤ 0.01, ≥ 2-fold change), using multiple databases, we (a) performed protein characterization, (b) demonstrated the presence of their RNA transcripts in both mouse and human cerebrovascular cells, and (c) demonstrated that several of these proteins were present in human extracellular vesicles (EVs) circulating in blood. Finally, using human plasma, we demonstrated the presence of several of these proteins in plasma and plasma EVs. Dysregulated proteins point to perturbed brain vessel vasomotricity, remodeling, and inflammation. Given that blood-isolated EVs are novel, attractive, and a minimally invasive biomarker discovery platform for age-related dementias, several proteins identified in this study can potentially serve as VCID markers in humans.
Collapse
Affiliation(s)
- Arsalan S. Haqqani
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada; (A.S.H.); (A.T.S.); (C.E.D.); (D.B.S.)
| | - Zainab Mianoor
- Multiomics Investigation of Neurodegenerative Diseases (MIND) Laboratory, 4545 Chemin Queen Mary, Montreal, QC H3W 1W4, Canada; (Z.M.); (F.E.D.)
- Département de Pharmacologie et Physiologie, Institut de Génie Biomédical, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
- Centre de Recherche de l’Institut Universitaire de Gériatrie (CRIUGM), 4545 Chemin Queen Mary, Montreal, QC H3W 1W4, Canada
| | - Alexandra T. Star
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada; (A.S.H.); (A.T.S.); (C.E.D.); (D.B.S.)
| | - Flavie E. Detcheverry
- Multiomics Investigation of Neurodegenerative Diseases (MIND) Laboratory, 4545 Chemin Queen Mary, Montreal, QC H3W 1W4, Canada; (Z.M.); (F.E.D.)
- Département de Pharmacologie et Physiologie, Institut de Génie Biomédical, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
- Centre de Recherche de l’Institut Universitaire de Gériatrie (CRIUGM), 4545 Chemin Queen Mary, Montreal, QC H3W 1W4, Canada
| | - Christie E. Delaney
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada; (A.S.H.); (A.T.S.); (C.E.D.); (D.B.S.)
| | - Danica B. Stanimirovic
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada; (A.S.H.); (A.T.S.); (C.E.D.); (D.B.S.)
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada;
| | - AmanPreet Badhwar
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada; (A.S.H.); (A.T.S.); (C.E.D.); (D.B.S.)
- Multiomics Investigation of Neurodegenerative Diseases (MIND) Laboratory, 4545 Chemin Queen Mary, Montreal, QC H3W 1W4, Canada; (Z.M.); (F.E.D.)
- Département de Pharmacologie et Physiologie, Institut de Génie Biomédical, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
- Centre de Recherche de l’Institut Universitaire de Gériatrie (CRIUGM), 4545 Chemin Queen Mary, Montreal, QC H3W 1W4, Canada
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada;
| |
Collapse
|
13
|
Dichgans M, Malik R, Beaufort N, Tanaka K, Georgakis M, He Y, Koido M, Terao C, Anderson C, Kamatani Y. Genetically proxied HTRA1 protease activity and circulating levels independently predict risk of ischemic stroke and coronary artery disease. RESEARCH SQUARE 2023:rs.3.rs-3523612. [PMID: 37986915 PMCID: PMC10659557 DOI: 10.21203/rs.3.rs-3523612/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
HTRA1 has emerged as a major risk gene for stroke and cerebral small vessel disease with both rare and common variants contributing to disease risk. However, the precise mechanisms mediating this risk remain largely unknown as does the full spectrum of phenotypes associated with genetic variation in HTRA1 in the general population. Using a family-history informed approach, we first show that rare variants in HTRA1 are linked to ischemic stroke in 425,338 European individuals from the UK Biobank with replication in 143,149 individuals from the Biobank Japan. Integrating data from biochemical experiments on 76 mutations occurring in the UK Biobank, we next show that rare variants causing loss of protease function in vitro associate with ischemic stroke, coronary artery disease, and skeletal traits. In addition, a common causal variant (rs2672592) modulating circulating HTRA1 mRNA and protein levels enhances the risk of ischemic stroke, small vessel stroke, and coronary artery disease while lowering the risk of migraine and age-related macular dystrophy in GWAS and UK Biobank data from > 2,000,000 individuals. There was no evidence of an interaction between genetically proxied HTRA1 activity and levels. Our findings demonstrate a central role of HTRA1 for human disease including stroke and coronary artery disease and identify two independent mechanisms that might qualify as targets for future therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Masaru Koido
- Institute of Medical Science, The University of Tokyo
| | | | | | | |
Collapse
|
14
|
Pan Y, Fu Y, Baird PN, Guymer RH, Das T, Iwata T. Exploring the contribution of ARMS2 and HTRA1 genetic risk factors in age-related macular degeneration. Prog Retin Eye Res 2023; 97:101159. [PMID: 36581531 DOI: 10.1016/j.preteyeres.2022.101159] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022]
Abstract
Age-related macular degeneration (AMD) is the leading cause of severe irreversible central vision loss in individuals over 65 years old. Genome-wide association studies (GWASs) have shown that the region at chromosome 10q26, where the age-related maculopathy susceptibility (ARMS2/LOC387715) and HtrA serine peptidase 1 (HTRA1) genes are located, represents one of the strongest associated loci for AMD. However, the underlying biological mechanism of this genetic association has remained elusive. In this article, we extensively review the literature by us and others regarding the ARMS2/HTRA1 risk alleles and their functional significance. We also review the literature regarding the presumed function of the ARMS2 protein and the molecular processes of the HTRA1 protein in AMD pathogenesis in vitro and in vivo, including those of transgenic mice overexpressing HtrA1/HTRA1 which developed Bruch's membrane (BM) damage, choroidal neovascularization (CNV), and polypoidal choroidal vasculopathy (PCV), similar to human AMD patients. The elucidation of the molecular mechanisms of the ARMS2 and HTRA1 susceptibility loci has begun to untangle the complex biological pathways underlying AMD pathophysiology, pointing to new testable paradigms for treatment.
Collapse
Affiliation(s)
- Yang Pan
- Division of Molecular and Cellular Biology, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, 2-5-1, Higashigaoka, Meguro-ku, Tokyo, 152-8902, Japan
| | - Yingbin Fu
- Department of Ophthalmology, Baylor College of Medicine, One Baylor Plaza, NC506, Houston, TX, 77030, USA
| | - Paul N Baird
- Department of Surgery, (Ophthalmology), Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Robyn H Guymer
- Department of Surgery, (Ophthalmology), Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia; Centre for Eye Research Australia, Royal Victorian Eye & Ear Hospital, East Melbourne, Victoria, 3002, Australia
| | - Taraprasad Das
- Anant Bajaj Retina Institute-Srimati Kanuri Santhamma Centre for Vitreoretinal Diseases, Kallam Anji Reddy Campus, L. V. Prasad Eye Institute, Hyderabad, 500034, India
| | - Takeshi Iwata
- Division of Molecular and Cellular Biology, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, 2-5-1, Higashigaoka, Meguro-ku, Tokyo, 152-8902, Japan.
| |
Collapse
|
15
|
Meschia JF, Worrall BB, Elahi FM, Ross OA, Wang MM, Goldstein ED, Rost NS, Majersik JJ, Gutierrez J. Management of Inherited CNS Small Vessel Diseases: The CADASIL Example: A Scientific Statement From the American Heart Association. Stroke 2023; 54:e452-e464. [PMID: 37602377 DOI: 10.1161/str.0000000000000444] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Lacunar infarcts and vascular dementia are important phenotypic characteristics of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy, the most common inherited cerebral small vessel disease. Individuals with the disease show variability in the nature and onset of symptoms and rates of progression, which are only partially explained by differences in pathogenic mutations in the NOTCH3 gene. Recognizing the disease early in its course and securing a molecular diagnosis are important clinical goals, despite the lack of proven disease-modifying treatments. The purposes of this scientific statement are to review the clinical, genetic, and imaging aspects of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy, contrasting it with other inherited small vessel diseases, and to provide key prevention, management, and therapeutic considerations with the intent of reducing practice variability and encouraging production of high-quality evidence to support future treatment recommendations.
Collapse
|
16
|
Hack RJ, Gravesteijn G, Cerfontaine MN, Santcroos MA, Gatti L, Kopczak A, Bersano A, Duering M, Rutten JW, Lesnik Oberstein SAJ. Three-tiered EGFr domain risk stratification for individualized NOTCH3-small vessel disease prediction. Brain 2023; 146:2913-2927. [PMID: 36535904 PMCID: PMC10316769 DOI: 10.1093/brain/awac486] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 11/23/2022] [Accepted: 11/27/2022] [Indexed: 02/10/2024] Open
Abstract
Cysteine-altering missense variants (NOTCH3cys) in one of the 34 epidermal growth-factor-like repeat (EGFr) domains of the NOTCH3 protein are the cause of NOTCH3-associated small vessel disease (NOTCH3-SVD). NOTCH3-SVD is highly variable, ranging from cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) at the severe end of the spectrum to non-penetrance. The strongest known NOTCH3-SVD modifier is NOTCH3cys variant position: NOTCH3cys variants located in EGFr domains 1-6 are associated with a more severe phenotype than NOTCH3cys variants located in EGFr domains 7-34. The objective of this study was to further improve NOTCH3-SVD genotype-based risk prediction by using relative differences in NOTCH3cys variant frequencies between large CADASIL and population cohorts as a starting point. Scientific CADASIL literature, cohorts and population databases were queried for NOTCH3cys variants. For each EGFr domain, the relative difference in NOTCH3cys variant frequency (NVFOR) was calculated using genotypes of 2574 CADASIL patients and 1647 individuals from population databases. Based on NVFOR cut-off values, EGFr domains were classified as either low (LR-EGFr), medium (MR-EGFr) or high risk (HR-EGFr). The clinical relevance of this new three-tiered EGFr risk classification was cross-sectionally validated by comparing SVD imaging markers and clinical outcomes between EGFr risk categories using a genotype-phenotype data set of 434 CADASIL patients and 1003 NOTCH3cys positive community-dwelling individuals. CADASIL patients and community-dwelling individuals harboured 379 unique NOTCH3cys variants. Nine EGFr domains were classified as an HR-EGFr, which included EGFr domains 1-6, but additionally also EGFr domains 8, 11 and 26. Ten EGFr domains were classified as MR-EGFr and 11 as LR-EGFr. In the population genotype-phenotype data set, HR-EGFr individuals had the highest risk of stroke [odds ratio (OR) = 10.81, 95% confidence interval (CI): 5.46-21.37], followed by MR-EGFr individuals (OR = 1.81, 95% CI: 0.84-3.88) and LR-EGFr individuals (OR = 1 [reference]). MR-EGFr individuals had a significantly higher normalized white matter hyperintensity volume (nWMHv; P = 0.005) and peak width of skeletonized mean diffusivity (PSMD; P = 0.035) than LR-EGFr individuals. In the CADASIL genotype-phenotype data set, HR-EGFr domains 8, 11 and 26 patients had a significantly higher risk of stroke (P = 0.002), disability (P = 0.041), nWMHv (P = 1.8 × 10-8), PSMD (P = 2.6 × 10-8) and lacune volume (P = 0.006) than MR-EGFr patients. SVD imaging marker load and clinical outcomes were similar between HR-EGFr 1-6 patients and HR-EGFr 8, 11 and 26 patients. NVFOR was significantly associated with vascular NOTCH3 aggregation load (P = 0.006), but not with NOTCH3 signalling activity (P = 0.88). In conclusion, we identified three clinically distinct NOTCH3-SVD EGFr risk categories based on NFVOR cut-off values, and identified three additional HR-EGFr domains located outside of EGFr domains 1-6. This EGFr risk classification will provide an important key to individualized NOTCH3-SVD disease prediction.
Collapse
Affiliation(s)
- Remco J Hack
- Department of Clinical Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Gido Gravesteijn
- Department of Clinical Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Minne N Cerfontaine
- Department of Clinical Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Mark A Santcroos
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Laura Gatti
- Laboratory of Neurobiology, Fondazione IRCSS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Anna Kopczak
- Institute for Stroke and Dementia Research, LMU University Hospital Munich, 81377 Munich, Germany
| | - Anna Bersano
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Marco Duering
- Institute for Stroke and Dementia Research, LMU University Hospital Munich, 81377 Munich, Germany
- Medical Image Analysis Center (MIAC) and Department of Biomedical Engineering, University of Basel, 4051 Basel, Switzerland
| | - Julie W Rutten
- Department of Clinical Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | |
Collapse
|
17
|
Wen JH, He XH, Feng ZS, Li DY, Tang JX, Liu HF. Cellular Protein Aggregates: Formation, Biological Effects, and Ways of Elimination. Int J Mol Sci 2023; 24:ijms24108593. [PMID: 37239937 DOI: 10.3390/ijms24108593] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
The accumulation of protein aggregates is the hallmark of many neurodegenerative diseases. The dysregulation of protein homeostasis (or proteostasis) caused by acute proteotoxic stresses or chronic expression of mutant proteins can lead to protein aggregation. Protein aggregates can interfere with a variety of cellular biological processes and consume factors essential for maintaining proteostasis, leading to a further imbalance of proteostasis and further accumulation of protein aggregates, creating a vicious cycle that ultimately leads to aging and the progression of age-related neurodegenerative diseases. Over the long course of evolution, eukaryotic cells have evolved a variety of mechanisms to rescue or eliminate aggregated proteins. Here, we will briefly review the composition and causes of protein aggregation in mammalian cells, systematically summarize the role of protein aggregates in the organisms, and further highlight some of the clearance mechanisms of protein aggregates. Finally, we will discuss potential therapeutic strategies that target protein aggregates in the treatment of aging and age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Jun-Hao Wen
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Xiang-Hong He
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Ze-Sen Feng
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Dong-Yi Li
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Ji-Xin Tang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Hua-Feng Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| |
Collapse
|
18
|
Panahi M, Hase Y, Gallart-Palau X, Mitra S, Watanabe A, Low RC, Yamamoto Y, Sepulveda-Falla D, Hainsworth AH, Ihara M, Sze SK, Viitanen M, Behbahani H, Kalaria RN. ER stress induced immunopathology involving complement in CADASIL: implications for therapeutics. Acta Neuropathol Commun 2023; 11:76. [PMID: 37158955 PMCID: PMC10169505 DOI: 10.1186/s40478-023-01558-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/26/2023] [Indexed: 05/10/2023] Open
Abstract
Cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is caused by NOTCH3 mutations. Typical CADASIL is characterised by subcortical ischemic strokes due to severe arteriopathy and fibrotic thickening of small arteries. Arteriolar vascular smooth muscle cells (VSMCs) are the key target in CADASIL, but the potential mechanisms involved in their degeneration are still unclear. Focusing on cerebral microvessels in the frontal and anterior temporal lobes and the basal ganglia, we used advanced proteomic and immunohistochemical methods to explore the extent of inflammatory and immune responses in CADASIL subjects compared to similar age normal and other disease controls. There was variable loss of VSMC in medial layers of arteries in white matter as well as the cortex, that could not be distinguished whether NOTCH3 mutations were in the epidermal growth factor (EGFr) domains 1-6 or EGFr7-34. Proteomics of isolated cerebral microvessels showed alterations in several proteins, many associated with endoplasmic reticulum (ER) stress including heat shock proteins. Cerebral vessels with sparsely populated VSMCs also attracted robust accrual of perivascular microglia/macrophages in order CD45+ > CD163+ > CD68+cells, with > 60% of vessel walls exhibiting intercellular adhesion molecule-1 (ICAM-1) immunoreactivity. Functional VSMC cultures bearing the NOTCH3 Arg133Cys mutation showed increased gene expression of the pro-inflammatory cytokine interleukin 6 and ICAM-1 by 16- and 50-fold, respectively. We further found evidence for activation of the alternative pathway of complement. Immunolocalisation of complement Factor B, C3d and C5-9 terminal complex but not C1q was apparent in ~ 70% of cerebral vessels. Increased complement expression was corroborated in > 70% of cultured VSMCs bearing the Arg133Cys mutation independent of N3ECD immunoreactivity. Our observations suggest that ER stress and other cellular features associated with arteriolar VSMC damage instigate robust localized inflammatory and immune responses in CADASIL. Our study has important implications for immunomodulation approaches to counter the characteristic arteriopathy of CADASIL.
Collapse
Affiliation(s)
- Mahmod Panahi
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Clinical Geriatrics, Karolinska Institutet, BioClinicum J9:20 Visionsgatan 4, Solna, 171 64, Sweden
| | - Yoshiki Hase
- Translational and Clinical Research Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Xavier Gallart-Palau
- Biomedical Research Institute of Lleida (IRBLLEIDA) - +Pec Proteomics Research Group (+PPRG) - Neuroscience Area, University Hospital Arnau de Vilanova (HUAV) - Department of Psychology, University of Lleida (UdL), Lleida, Spain
| | - Sumonto Mitra
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Clinical Geriatrics, Karolinska Institutet, BioClinicum J9:20 Visionsgatan 4, Solna, 171 64, Sweden
| | - Atsushi Watanabe
- Equipment Management Division, Center for Core Facility Administration, Research Institute, National Center for Geriatrics and Gerontology (NCGG), 7-430, Morioka-cho, Obu-shi, 474-8511, Aichi, Japan
| | - Roger C Low
- Translational and Clinical Research Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Yumi Yamamoto
- Department of Molecular Innovation in Lipidemiology and Department of Neurology, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita, 564-8565, Osaka, Japan
| | - Diego Sepulveda-Falla
- Molecular Neuropathology of Alzheimer's Disease, Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Atticus H Hainsworth
- Molecular and Clinical Sciences, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Masafumi Ihara
- Department of Molecular Innovation in Lipidemiology and Department of Neurology, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita, 564-8565, Osaka, Japan
| | - Siu Kwan Sze
- Faculty of Applied Health Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Matti Viitanen
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Clinical Geriatrics, Karolinska Institutet, BioClinicum J9:20 Visionsgatan 4, Solna, 171 64, Sweden
- Department of Geriatrics, University of Turku, Turku City Hospital, Kunnallissairaalantie 20, Turku, 20700, Finland
| | - Homira Behbahani
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Clinical Geriatrics, Karolinska Institutet, BioClinicum J9:20 Visionsgatan 4, Solna, 171 64, Sweden
| | - Raj N Kalaria
- Translational and Clinical Research Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK.
| |
Collapse
|
19
|
Haffner C. The emerging role of the HTRA1 protease in brain microvascular disease. FRONTIERS IN DEMENTIA 2023; 2:1146055. [PMID: 39081996 PMCID: PMC11285548 DOI: 10.3389/frdem.2023.1146055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/10/2023] [Indexed: 08/02/2024]
Abstract
Pathologies of the brain microvasculature, often referred to as cerebral small-vessel disease, are important contributors to vascular dementia, the second most common form of dementia in aging societies. In addition to their role in acute ischemic and hemorrhagic stroke, they have emerged as major cause of age-related cognitive decline in asymptomatic individuals. A central histological finding in these pathologies is the disruption of the vessel architecture including thickening of the vessel wall, narrowing of the vessel lumen and massive expansion of the mural extracellular matrix. The underlying molecular mechanisms are largely unknown, but from the investigation of several disease forms with defined etiology, high temperature requirement protein A1 (HTRA1), a secreted serine protease degrading primarily matrisomal substrates, has emerged as critical factor and potential therapeutic target. A genetically induced loss of HTRA1 function in humans is associated with cerebral autosomal-recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL), a rare, hereditary form of brain microvascular disease. Recently, proteomic studies on cerebral amyloid angiopathy (CAA), a common cause of age-related dementia, and cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), the most prevalent monogenic small-vessel disease, have provided evidence for an impairment of HTRA1 activity through sequestration into pathological protein deposits, suggesting an alternative mechanism of HTRA1 inactivation and expanding the range of diseases with HTRA1 involvement. Further investigations of the mechanisms of HTRA1 regulation in the brain microvasculature might spawn novel strategies for the treatment of small-vessel pathologies.
Collapse
Affiliation(s)
- Christof Haffner
- Department of Psychiatry and Psychotherapy, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
20
|
Qian E, Uemura M, Kobayashi H, Nakamura S, Ozawa F, Yoshimatsu S, Ishikawa M, Onodera O, Morimoto S, Okano H. A human induced pluripotent stem cell model from a patient with hereditary cerebral small vessel disease carrying a heterozygous R302Q mutation in HTRA1. Inflamm Regen 2023; 43:23. [PMID: 37009886 PMCID: PMC10069112 DOI: 10.1186/s41232-023-00273-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 03/13/2023] [Indexed: 04/04/2023] Open
Abstract
Cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL) is an inherited cerebral small vessel disease (CSVD) caused by biallelic mutations in the high-temperature requirement serine peptidase A1 (HTRA1) gene. Even heterozygous mutations in HTRA1 are recently revealed to cause cardinal clinical features of CSVD. Here, we report the first establishment of a human induced pluripotent stem cell (hiPSC) line from a patient with heterozygous HTRA1-related CSVD. Peripheral blood mononuclear cells (PBMCs) were reprogrammed by the transfection of episomal vectors encoding human OCT3/4 (POU5F1), SOX2, KLF4, L-MYC, LIN28, and a murine dominant-negative mutant of p53 (mp53DD). The established iPSCs had normal morphology as human pluripotent stem cells and normal karyotype (46XX). Moreover, we found that the HTRA1 missense mutation (c.905G>A, p.R302Q) was heterozygous. These iPSCs expressed pluripotency-related markers and had the potential to differentiate into all three germ layers in vitro. HTRA1 and the supposed disease-associated gene NOG were differentially expressed in the patient iPSCs at mRNA levels compared to those of control lines. The iPSC line would facilitate in vitro research for understanding the cellular pathomechanisms caused by the HTRA1 mutation including its dominant-negative effect.
Collapse
Affiliation(s)
- Emi Qian
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Masahiro Uemura
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hiroya Kobayashi
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Shiho Nakamura
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Fumiko Ozawa
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Sho Yoshimatsu
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Mitsuru Ishikawa
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Satoru Morimoto
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
21
|
Yamamoto Y, Liao YC, Lee YC, Ihara M, Choi JC. Update on the Epidemiology, Pathogenesis, and Biomarkers of Cerebral Autosomal Dominant Arteriopathy With Subcortical Infarcts and Leukoencephalopathy. J Clin Neurol 2023; 19:12-27. [PMID: 36606642 PMCID: PMC9833879 DOI: 10.3988/jcn.2023.19.1.12] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/06/2022] [Accepted: 11/09/2022] [Indexed: 01/04/2023] Open
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common monogenic disorder of the cerebral small blood vessels. It is caused by mutations in the NOTCH3 gene on chromosome 19, and more than 280 distinct pathogenic mutations have been reported to date. CADASIL was once considered a very rare disease with an estimated prevalence of 1.3-4.1 per 100,000 adults. However, recent large-scale genomic studies have revealed a high prevalence of pathogenic NOTCH3 variants among the general population, with the highest risk being among Asians. The disease severity and age at onset vary significantly even among individuals who carry the same NOTCH3 mutations. It is still unclear whether a significant genotype-phenotype correlation is present in CADASIL. The accumulation of granular osmiophilic material in the vasculature is a characteristic feature of CADASIL. However, the exact pathogenesis of CADASIL remains largely unclear despite various laboratory and clinical observations being made. Major hypotheses proposed so far have included aberrant NOTCH3 signaling, toxic aggregation, and abnormal matrisomes. Several characteristic features have been observed in the brain magnetic resonance images of patients with CADASIL, including subcortical lacunar lesions and white matter hyperintensities in the anterior temporal lobe or external capsule, which were useful in differentiating CADASIL from sporadic stroke in patients. The number of lacunes and the degree of brain atrophy were useful in predicting the clinical outcomes of patients with CADASIL. Several promising blood biomarkers have also recently been discovered for CADASIL, which require further research for validation.
Collapse
Affiliation(s)
- Yumi Yamamoto
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yi-Chu Liao
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Chung Lee
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Jay Chol Choi
- Department of Neurology, Jeju National University, Jeju, Korea.,Institute for Medical Science, Jeju National University, Jeju, Korea
| |
Collapse
|
22
|
Yao T, Zhu J, Wu X, Li X, Fu Y, Wang Y, Wang Z, Xu F, Lai H, He A, Teng L, Wang C, Song H. Heterozygous HTRA1Mutations Cause Cerebral Small Vessel Diseases. Neurol Genet 2022; 8:e200044. [DOI: 10.1212/nxg.0000000000200044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 09/28/2022] [Indexed: 12/12/2022]
Abstract
Background and ObjectivesCerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL) is a rare hereditary cerebrovascular disease caused by homozygous or compound heterozygous variations in the high-temperature requirement A serine peptidase 1 (HTRA1) gene. However, several studies in recent years have found that some heterozygousHTRA1mutations also cause cerebral small vessel disease (CSVD). The current study aims to report the novel genotypes, phenotypes, and histopathologic results of 3 pedigrees of CSVD with heterozygousHTRA1mutation.MethodsThree pedigrees of familiar CSVD, including 11 symptomatic patients and 3 asymptomatic carriers, were enrolled. Whole-exome sequencing was conducted in the probands for identifying rare variants, which were then evaluated for pathogenicity according to the American College of Medical Genetics and Genomics guidelines. Sanger sequencing was performed for validation of mutations in the probands and other family members. The protease activity was assayed for the novel mutations. All the participants received detailed clinical and imaging examinations and the corresponding results were concluded. Hematoma evacuation was performed for an intracerebral hemorrhage patient with the p.Q318H mutation, and the postoperative pathology including hematoma and cerebral small vessels were examined.ResultsThree novel heterozygousHTRA1mutations (p.Q318H, p.V279M, and p.R274W) were detected in the 3 pedigrees. The protease activity was largely lost for all the mutations, confirming that they were loss-of-function mutations. The patients in each pedigree presented with typical clinical and imaging features of CVSD, and some of them displayed several new phenotypes including color blindness, hydrocephalus, and multiple arachnoid cysts. In addition, family 1 is the largest pedigree with heterozygousHTRA1mutation so far and includes homozygous twins, displaying some variation in clinical phenotypes. More importantly, pathologic study of a patient with p.Q318H mutation showed hyalinization, luminal stenosis, loss of smooth muscle cells, splitting of the internal elastic lamina, and intramural hemorrhage/dissection-like structures.DiscussionThese findings broaden the mutational and clinical spectrum of heterozygousHTRA1-related CSVD. Pathologic features were similar with the previous heterozygous and homozygous cases. Moreover, clinical heterogeneity was revealed within the largest single family, and the mechanisms of the phenotypic heterogenetic remain unclear. Overall, heterozygous HTRA1-related CSVD should not be simply taken as a mild type of CARASIL as previously considered.
Collapse
|
23
|
Cheng YW, Chao CC, Chen CH, Yeh TY, Jeng JS, Tang SC, Hsieh ST. Small Fiber Pathology in CADASIL: Clinical Correlation With Cognitive Impairment. Neurology 2022; 99:e583-e593. [PMID: 35584924 PMCID: PMC9442619 DOI: 10.1212/wnl.0000000000200672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/16/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES This study investigated the cutaneous small fiber pathology of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and its clinical significance, that is, the NOTCH3 deposition in cutaneous vasculatures and CNS neurodegeneration focusing on cognitive impairment. METHODS Thirty-seven patients with CADASIL and 59 age-matched healthy controls were enrolled to evaluate cutaneous small fiber pathology by quantitative measures of intraepidermal nerve fiber density (IENFD), sweat gland innervation, and vascular innervation. Cognitive performance of patients with CADASIL was evaluated by a comprehensive neuropsychological assessment, and its association with small fiber pathology was tested using multivariable linear regression analysis adjusted for age and diabetes mellitus. We further assessed the relationships of IENFD with cutaneous vascular NOTCH3 ectodomain (NOTCH3ECD) deposition and biomarkers of neurodegeneration including structural brain MRI measures, serum neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), tau, and ubiquitin carboxy-terminal hydrolase L1. RESULTS Patients with CADASIL showed reduced IENFD (5.22 ± 2.42 vs 7.88 ± 2.89 fibers/mm, p = 0.0001) and reduced sweat gland (p < 0.0001) and vascular (p < 0.0001) innervations compared with age-matched controls. Reduced IENFD was associated with impaired global cognition measured by Mini-Mental State Examination (B = 1.062, 95% CI = 0.370-1.753, p = 0.004), and this association remained after adjustment for age and diabetes mellitus (p = 0.043). In addition, IENFD in patients with CADASIL was associated with mean cortical thickness (Pearson r = 0.565, p = 0.0023) but not white matter hyperintensity volume, total lacune count, or total microbleed count. Reduced IENFD was associated with cutaneous vascular NOTCH3ECD deposition amount among patients harboring pathogenic variants in exon 11 (mainly p.R544C) (B = -0.092, 95% CI = -0.175 to -0.009, p = 0.031). Compared with those with normal cognition, patients with CADASIL with cognitive impairment had an elevated plasma NfL level regardless of concurrent small fiber denervation, whereas only patients with both cognitive impairment and small fiber denervation showed an elevated plasma GFAP level. DISCUSSION Cutaneous small fiber pathology correlates with cognitive impairment and CNS neurodegeneration in patients with CADASIL, indicating a peripheral neurodegenerative process related to NOTCH3ECD aggregation.
Collapse
Affiliation(s)
- Yu-Wen Cheng
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Chi-Chao Chao
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Chih-Hao Chen
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Ti-Yen Yeh
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Jiann-Shing Jeng
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Sung-Chun Tang
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Sung-Tsang Hsieh
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei.
| |
Collapse
|
24
|
Hack RJ, Gravesteijn G, Cerfontaine MN, Hegeman IM, Mulder AA, Lesnik Oberstein SA, Rutten JW. Cerebral Autosomal Dominant Arteriopathy With Subcortical Infarcts and Leukoencephalopathy Family Members With a Pathogenic NOTCH3 Variant Can Have a Normal Brain Magnetic Resonance Imaging and Skin Biopsy Beyond Age 50 Years. Stroke 2022; 53:1964-1974. [PMID: 35300531 PMCID: PMC9126263 DOI: 10.1161/strokeaha.121.036307] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/16/2021] [Accepted: 12/15/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND To determine whether extremely mild small vessel disease (SVD) phenotypes can occur in NOTCH3 variant carriers from Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL) pedigrees using clinical, genetic, neuroimaging, and skin biopsy findings. METHODS Individuals from CADASIL pedigrees fulfilling criteria for extremely mild NOTCH3-associated SVD (mSVDNOTCH3) were selected from the cross-sectional Dutch CADASIL cohort (n=200), enrolled between 2017 and 2020. Brain magnetic resonance imaging were quantitatively assessed for SVD imaging markers. Immunohistochemistry and electron microscopy was used to quantitatively assess and compare NOTCH3 ectodomain (NOTCH3ECD) aggregation and granular osmiophilic material deposits in the skin vasculature of mSVDNOTCH3 cases and symptomatic CADASIL patients. RESULTS Seven cases were identified that fulfilled the mSVDNOTCH3 criteria, with a mean age of 56.6 years (range, 50-72). All of these individuals harbored a NOTCH3 variant located in one of EGFr domains 7-34 and had a normal brain magnetic resonance imaging, except the oldest individual, aged 72, who had beginning confluence of WMH (Fazekas score 2) and 1 cerebral microbleed. mSVDNOTCH3 cases had very low levels of NOTCH3ECD aggregation in skin vasculature, which was significantly less than in symptomatic EGFr 7-34 CADASIL patients (P=0.01). Six mSVDNOTCH3 cases had absence of granular osmiophilic material deposits. CONCLUSIONS Our findings demonstrate that extremely mild SVD phenotypes can occur in individuals from CADASIL pedigrees harboring NOTCH3 EGFr 7-34 variants with normal brain magnetic resonance imaging up to age 58 years. Our study has important implications for CADASIL diagnosis, disease prediction, and the counseling of individuals from EGFr 7-34 CADASIL pedigrees.
Collapse
Affiliation(s)
- Remco J. Hack
- Department of Clinical Genetics (R.J.H., G.G., M.N.C., S.A.J.L.O., J.W.R.), Leiden University Medical Center, the Netherlands
| | - Gido Gravesteijn
- Department of Clinical Genetics (R.J.H., G.G., M.N.C., S.A.J.L.O., J.W.R.), Leiden University Medical Center, the Netherlands
| | - Minne N. Cerfontaine
- Department of Clinical Genetics (R.J.H., G.G., M.N.C., S.A.J.L.O., J.W.R.), Leiden University Medical Center, the Netherlands
| | - Ingrid M. Hegeman
- Department of Pathology (I.M.H.), Leiden University Medical Center, the Netherlands
| | - Aat A. Mulder
- Department of Cell and Chemical Biology (A.A.M.), Leiden University Medical Center, the Netherlands
| | - Saskia A.J. Lesnik Oberstein
- Department of Clinical Genetics (R.J.H., G.G., M.N.C., S.A.J.L.O., J.W.R.), Leiden University Medical Center, the Netherlands
| | - Julie W. Rutten
- Department of Clinical Genetics (R.J.H., G.G., M.N.C., S.A.J.L.O., J.W.R.), Leiden University Medical Center, the Netherlands
| |
Collapse
|
25
|
Park JH, Cho SJ, Jo C, Park MH, Han C, Kim EJ, Huh GY, Koh YH. Altered TIMP-3 Levels in the Cerebrospinal Fluid and Plasma of Patients with Alzheimer’s Disease. J Pers Med 2022; 12:jpm12050827. [PMID: 35629249 PMCID: PMC9144624 DOI: 10.3390/jpm12050827] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/07/2022] [Accepted: 05/14/2022] [Indexed: 02/04/2023] Open
Abstract
Tissue inhibitor of metalloproteinase-3 (TIMP-3) is a component of the extracellular environment and is suggested to play an indirect role in regulating Aβ production and the pathophysiology of Aβ deposition in brains. However, studies on the amount of TIMP-3 in bodily fluids of Alzheimer’s disease (AD) patients have not been conducted. Here, we investigated the relationship between fluid TIMP-3 levels and AD pathology. We first showed that the fluid levels of TIMP-3 were lower in AD dementia patients compared with in non-AD patients. ELISA results revealed that plasma levels of TIMP-3 in 65 patients with AD were significantly lower than those in 115 healthy control subjects and 71 mild cognitive impairment (MCI) subjects. Furthermore, we found that cerebrospinal fluid (CSF) level of TIMP-3 was decreased in AD compared with that in healthy control. These data suggest that fluid TIMP-3 levels negatively correlated with progress of cognitive decline. Collectively, our study suggests that alterations of fluid TIMP-3 levels might be associated with AD pathology.
Collapse
Affiliation(s)
- Jung Hyun Park
- Department of Chronic Disease Convergence Research, Division of Brain Disease Research, Korea National Institute of Health, 187 Osongsaengmyeong2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28159, Korea; (J.H.P.); (S.-J.C.); (C.J.)
| | - Sun-Jung Cho
- Department of Chronic Disease Convergence Research, Division of Brain Disease Research, Korea National Institute of Health, 187 Osongsaengmyeong2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28159, Korea; (J.H.P.); (S.-J.C.); (C.J.)
| | - Chulman Jo
- Department of Chronic Disease Convergence Research, Division of Brain Disease Research, Korea National Institute of Health, 187 Osongsaengmyeong2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28159, Korea; (J.H.P.); (S.-J.C.); (C.J.)
| | - Moon Ho Park
- Departments of Neurology, Korea University Medical College, Ansan Hospital, 123 Jeokgeum-ro, Danwon-gu, Ansan-si 15355, Korea;
| | - Changsu Han
- Departments of Psychiatry, Korea University Medical College, Ansan Hospital, 123 Jeokgeum-ro, Danwon-gu, Ansan-si 15355, Korea;
| | - Eun-Joo Kim
- Department of Neurology, Pusan National University Hospital, 179 Gudeok-ro, Seo-gu, Busan 49241, Korea;
| | - Gi Yeong Huh
- Department of Forensic Medicine, Pusan National University School of Medicine, 49 Busandaehak-ro, Mulgeum-eup, Yangsan-si 50612, Korea;
| | - Young Ho Koh
- Department of Chronic Disease Convergence Research, Division of Brain Disease Research, Korea National Institute of Health, 187 Osongsaengmyeong2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28159, Korea; (J.H.P.); (S.-J.C.); (C.J.)
- Correspondence:
| |
Collapse
|
26
|
Guo H, Guo X, Jiang S. Long non-coding RNA lincRNA-erythroid prosurvival (EPS) alleviates cerebral ischemia/reperfusion injury by maintaining high-temperature requirement protein A1 (Htra1) stability through recruiting heterogeneous nuclear ribonucleoprotein L (HNRNPL). Bioengineered 2022; 13:12248-12260. [PMID: 35549989 PMCID: PMC9275866 DOI: 10.1080/21655979.2022.2074738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
This study aimed at investigating the role and mechanism of lincRNA-EPS (erythroid prosurvival) in cerebral ischemia/reperfusion (CIR) injury. The results showed that the overexpression of lincRNA-EPS was able to reduce the levels of interleukin-6, tumor necrosis factor-alpha and interleukin-1β stimulated in the OGD-treated Neuro-2a (N-2a) cells. The levels of reactive oxygen species and malondialdehyde were enhanced while the superoxide dismutase levels were reduced by oxygen and glucose deprivation (OGD) treatment, in which the lincRNA-EPS overexpression could reverse this effect in the cells. LincRNA-EPS interacted with high-temperature requirement protein A1 (Htra1) and heterogeneous nuclear ribonucleoprotein L (HNRNPL), and their depletion inhibited the Htra1 mRNA stability in N-2a cells. HNRNPL knockdown blocked lincRNA-EPS overexpression-induced Htra1 expression in the cells. The depletion of Htra1 could rescue lincRNA-EPS overexpression-mediated N-2a cell injury, inflammation, and oxidative stress induced by OGD. Functionally, lincRNA-EPS alleviates CIR injury of the middle cerebral artery occlusion/reperfusion mice in vivo. In conclusion, lincRNA-EPS attenuates CIR injury by maintaining Htra1 stability through recruiting HNRNPL.
Collapse
Affiliation(s)
- Haifeng Guo
- Department of encephalopathy, Jinan Municipal Hospital of Traditional Chinese Medicine, Jinan, Shandong, P.R.China
| | - Xia Guo
- Department of Obstetrics, Dongying People's Hospital, Dongying, Shandong, P.R.China
| | - Shiting Jiang
- Department of Internal Medicine-Neurology, Dongping People's Hospital, Taian, Shandong, P.R.China
| |
Collapse
|
27
|
Interplay between HTRA1 and classical signalling pathways in organogenesis and diseases. Saudi J Biol Sci 2022; 29:1919-1927. [PMID: 35531175 PMCID: PMC9072889 DOI: 10.1016/j.sjbs.2021.11.056] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/05/2021] [Accepted: 11/17/2021] [Indexed: 11/20/2022] Open
Abstract
The high temperature requirement factor A1 (HTRA1) is a serine protease which modulates an array of signalling pathways driving basal biological processes. HTRA1 plays a significant role in cell proliferation, migration and fate determination, in addition to controlling protein aggregates through refolding, translocation or degradation. The mutation of HTRA1 has been implicated in a plethora of disorders and this has also led to its growing interest as drug therapy target. This review details the involvement of HTRA1 in certain signalling pathways, namely the transforming growth factor beta (TGF-β), canonical Wingless/Integrated (WNT) and NOTCH signalling pathways during organogenesis and various disease pathogenesis such as preeclampsia, age-related macular degeneration (AMD), small vessel disease and cancer. We have also explored possible avenues of exploiting the serine proteases for therapeutic management of these disorders.
Collapse
|
28
|
Zellner A, Müller SA, Lindner B, Beaufort N, Rozemuller AJM, Arzberger T, Gassen NC, Lichtenthaler SF, Kuster B, Haffner C, Dichgans M. Proteomic profiling in cerebral amyloid angiopathy reveals an overlap with CADASIL highlighting accumulation of HTRA1 and its substrates. Acta Neuropathol Commun 2022; 10:6. [PMID: 35074002 PMCID: PMC8785498 DOI: 10.1186/s40478-021-01303-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) is an age-related condition and a major cause of intracerebral hemorrhage and cognitive decline that shows close links with Alzheimer's disease (AD). CAA is characterized by the aggregation of amyloid-β (Aβ) peptides and formation of Aβ deposits in the brain vasculature resulting in a disruption of the angioarchitecture. Capillaries are a critical site of Aβ pathology in CAA type 1 and become dysfunctional during disease progression. Here, applying an advanced protocol for the isolation of parenchymal microvessels from post-mortem brain tissue combined with liquid chromatography tandem mass spectrometry (LC-MS/MS), we determined the proteomes of CAA type 1 cases (n = 12) including a patient with hereditary cerebral hemorrhage with amyloidosis-Dutch type (HCHWA-D), and of AD cases without microvascular amyloid pathology (n = 13) in comparison to neurologically healthy controls (n = 12). ELISA measurements revealed microvascular Aβ1-40 levels to be exclusively enriched in CAA samples (mean: > 3000-fold compared to controls). The proteomic profile of CAA type 1 was characterized by massive enrichment of multiple predominantly secreted proteins and showed significant overlap with the recently reported brain microvascular proteome of patients with cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), a hereditary cerebral small vessel disease (SVD) characterized by the aggregation of the Notch3 extracellular domain. We found this overlap to be largely attributable to the accumulation of high-temperature requirement protein A1 (HTRA1), a serine protease with an established role in the brain vasculature, and several of its substrates. Notably, this signature was not present in AD cases. We further show that HTRA1 co-localizes with Aβ deposits in brain capillaries from CAA type 1 patients indicating a pathologic recruitment process. Together, these findings suggest a central role of HTRA1-dependent protein homeostasis in the CAA microvasculature and a molecular connection between multiple types of brain microvascular disease.
Collapse
Affiliation(s)
- Andreas Zellner
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Barbara Lindner
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Nathalie Beaufort
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Annemieke J M Rozemuller
- Department of Pathology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Thomas Arzberger
- Department of Psychiatry and Psychotherapy, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Nils C Gassen
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
| | - Christof Haffner
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany.
- Department of Psychiatry and Psychotherapy, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany.
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
29
|
Yan X, Shang J, Wang R, Wang F, Zhang J. Mechanisms regulating cerebral hypoperfusion in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. J Biomed Res 2022; 36:353-357. [PMID: 36165325 PMCID: PMC9548441 DOI: 10.7555/jbr.36.20220208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Cerebral small vessel disease (CSVD) is a leading cause of stroke and dementia. As the most common type of inherited CSVD, cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is characterized by the NOTCH3 gene mutation which leads to Notch3 ectodomain deposition and extracellular matrix aggregation around the small vessels. It further causes smooth muscle cell degeneration and small vessel arteriopathy in the central nervous system. Compromised cerebral blood flow occurs in the early stage of CADASIL and is associated with white matter hyperintensity, the typical neuroimaging pathology of CADASIL. This suggests that cerebral hypoperfusion may play an important role in the pathogenesis of CADASIL. However, the mechanistic linkage between NOTCH3 mutation and cerebral hypoperfusion remains unknown. Therefore, in this mini-review, it examines the cellular and molecular mechanisms contributing to cerebral hypoperfusion in CADASIL.
Collapse
Affiliation(s)
- Xi Yan
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan 450003, China
| | - Junkui Shang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan 450003, China
| | - Runrun Wang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan 450003, China
| | - Fengyu Wang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan 450003, China
| | - Jiewen Zhang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan 450003, China
- Jiewen Zhang, Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, No. 7 Weiwu Road, Zhengzhou, Henan 450003, China. Tel: +86-371-65580782, E-mail:
| |
Collapse
|
30
|
Kato T, Manabe RI, Igarashi H, Kametani F, Hirokawa S, Sekine Y, Fujita N, Saito S, Kawashima Y, Hatano Y, Ando S, Nozaki H, Sugai A, Uemura M, Fukunaga M, Sato T, Koyama A, Saito R, Sugie A, Toyoshima Y, Kawata H, Murayama S, Matsumoto M, Kakita A, Hasegawa M, Ihara M, Kanazawa M, Nishizawa M, Tsuji S, Onodera O. Candesartan prevents arteriopathy progression in cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy model. J Clin Invest 2021; 131:140555. [PMID: 34779414 DOI: 10.1172/jci140555] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 10/01/2021] [Indexed: 01/15/2023] Open
Abstract
Cerebral small vessel disease (CSVD) causes dementia and gait disturbance due to arteriopathy. Cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL) is a hereditary form of CSVD caused by loss of high-temperature requirement A1 (HTRA1) serine protease activity. In CARASIL, arteriopathy causes intimal thickening, smooth muscle cell (SMC) degeneration, elastic lamina splitting, and vasodilation. The molecular mechanisms were proposed to involve the accumulation of matrisome proteins as substrates or abnormalities in transforming growth factor β (TGF-β) signaling. Here, we show that HTRA1-/- mice exhibited features of CARASIL-associated arteriopathy: intimal thickening, abnormal elastic lamina, and vasodilation. In addition, the mice exhibited reduced distensibility of the cerebral arteries and blood flow in the cerebral cortex. In the thickened intima, matrisome proteins, including the hub protein fibronectin (FN) and latent TGF-β binding protein 4 (LTBP-4), which are substrates of HTRA1, accumulated. Candesartan treatment alleviated matrisome protein accumulation and normalized the vascular distensibility and cerebral blood flow. Furthermore, candesartan reduced the mRNA expression of Fn1, Ltbp-4, and Adamtsl2, which are involved in forming the extracellular matrix network. Our results indicate that these accumulated matrisome proteins may be potential therapeutic targets for arteriopathy in CARASIL.
Collapse
Affiliation(s)
- Taisuke Kato
- Department of System Pathology for Neurological Disorders, Brain Science Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Ri-Ichiroh Manabe
- Laboratory for Comprehensive Genomic Analysis, Center for Integrative Medical Sciences, RIKEN, Kanagawa, Japan
| | - Hironaka Igarashi
- Center for Integrated Human Brain Science, Brain Research Institute, Niigata University, Niigata, Japan
| | - Fuyuki Kametani
- Department of Brain and Neuroscience, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Sachiko Hirokawa
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yumi Sekine
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Natsumi Fujita
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Satoshi Saito
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Yusuke Kawashima
- Department of Applied Genomics, Kazusa DNA Research Institute, Chiba, Japan
| | - Yuya Hatano
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Shoichiro Ando
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hiroaki Nozaki
- Department of Medical Technology, Graduate School of Health Sciences, Niigata University, Niigata, Japan
| | - Akihiro Sugai
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masahiro Uemura
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masaki Fukunaga
- Division of Cerebral Integration, Department of System Neuroscience, National Institute for Physiological Sciences, Aichi, Japan
| | - Toshiya Sato
- Department of Laboratory Animal Science, Kitasato University School of Medicine, Kanagawa, Japan
| | - Akihide Koyama
- Department of Legal Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Rie Saito
- Department of Pathology, Clinical Neuroscience Branch and
| | - Atsushi Sugie
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, Japan
| | | | - Hirotoshi Kawata
- Department of Pathology, Jichi Medical University, Tochigi, Japan
| | - Shigeo Murayama
- Brain Bank for Aging Research, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan.,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, University of Osaka, Osaka, Japan
| | - Masaki Matsumoto
- Department of Omics and Systems Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | | | - Masato Hasegawa
- Department of Brain and Neuroscience, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Masato Kanazawa
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | | | - Shoji Tsuji
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Osamu Onodera
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
31
|
Malik R, Beaufort N, Frerich S, Gesierich B, Georgakis MK, Rannikmäe K, Ferguson AC, Haffner C, Traylor M, Ehrmann M, Sudlow CLM, Dichgans M. Whole-exome sequencing reveals a role of HTRA1 and EGFL8 in brain white matter hyperintensities. Brain 2021; 144:2670-2682. [PMID: 34626176 PMCID: PMC8557338 DOI: 10.1093/brain/awab253] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/01/2021] [Accepted: 06/19/2021] [Indexed: 11/13/2022] Open
Abstract
White matter hyperintensities (WMH) are among the most common radiological abnormalities in the ageing population and an established risk factor for stroke and dementia. While common variant association studies have revealed multiple genetic loci with an influence on their volume, the contribution of rare variants to the WMH burden in the general population remains largely unexplored. We conducted a comprehensive analysis of this burden in the UK Biobank using publicly available whole-exome sequencing data (n up to 17 830) and found a splice-site variant in GBE1, encoding 1,4-alpha-glucan branching enzyme 1, to be associated with lower white matter burden on an exome-wide level [c.691+2T>C, β = -0.74, standard error (SE) = 0.13, P = 9.7 × 10-9]. Applying whole-exome gene-based burden tests, we found damaging missense and loss-of-function variants in HTRA1 (frequency of 1 in 275 in the UK Biobank population) to associate with an increased WMH volume (P = 5.5 × 10-6, false discovery rate = 0.04). HTRA1 encodes a secreted serine protease implicated in familial forms of small vessel disease. Domain-specific burden tests revealed that the association with WMH volume was restricted to rare variants in the protease domain (amino acids 204-364; β = 0.79, SE = 0.14, P = 9.4 × 10-8). The frequency of such variants in the UK Biobank population was 1 in 450. The WMH volume was brought forward by ∼11 years in carriers of a rare protease domain variant. A comparison with the effect size of established risk factors for WMH burden revealed that the presence of a rare variant in the HTRA1 protease domain corresponded to a larger effect than meeting the criteria for hypertension (β = 0.26, SE = 0.02, P = 2.9 × 10-59) or being in the upper 99.8% percentile of the distribution of a polygenic risk score based on common genetic variants (β = 0.44, SE = 0.14, P = 0.002). In biochemical experiments, most (6/9) of the identified protease domain variants resulted in markedly reduced protease activity. We further found EGFL8, which showed suggestive evidence for association with WMH volume (P = 1.5 × 10-4, false discovery rate = 0.22) in gene burden tests, to be a direct substrate of HTRA1 and to be preferentially expressed in cerebral arterioles and arteries. In a phenome-wide association study mapping ICD-10 diagnoses to 741 standardized Phecodes, rare variants in the HTRA1 protease domain were associated with multiple neurological and non-neurological conditions including migraine with aura (odds ratio = 12.24, 95%CI: 2.54-35.25; P = 8.3 × 10-5]. Collectively, these findings highlight an important role of rare genetic variation and the HTRA1 protease in determining WMH burden in the general population.
Collapse
Affiliation(s)
- Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Nathalie Beaufort
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Simon Frerich
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Benno Gesierich
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Marios K Georgakis
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Kristiina Rannikmäe
- Centre for Medical Informatics, Usher Institute, University of Edinburgh, Edinburgh EH16 4TL, UK
| | - Amy C Ferguson
- Centre for Medical Informatics, Usher Institute, University of Edinburgh, Edinburgh EH16 4TL, UK
| | - Christof Haffner
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Matthew Traylor
- Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
- The Barts Heart Centre and NIHR Barts Biomedical Research Centre - Barts Health NHS Trust, The William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Michael Ehrmann
- Center of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen 45141, Germany
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Cathie L M Sudlow
- Centre for Medical Informatics, Usher Institute, University of Edinburgh, Edinburgh EH16 4TL, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4TL, UK
- Health Data Research UK Scotland, University of Edinburgh, Edinburgh EH16 4TL, UK
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology, Munich 81377, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich 81377, Germany
| |
Collapse
|
32
|
Tüshaus J, Müller SA, Shrouder J, Arends M, Simons M, Plesnila N, Blobel CP, Lichtenthaler SF. The pseudoprotease iRhom1 controls ectodomain shedding of membrane proteins in the nervous system. FASEB J 2021; 35:e21962. [PMID: 34613632 DOI: 10.1096/fj.202100936r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/31/2021] [Accepted: 09/15/2021] [Indexed: 12/22/2022]
Abstract
Proteolytic ectodomain shedding of membrane proteins is a fundamental mechanism to control the communication between cells and their environment. A key protease for membrane protein shedding is ADAM17, which requires a non-proteolytic subunit, either inactive Rhomboid 1 (iRhom1) or iRhom2 for its activity. While iRhom1 and iRhom2 are co-expressed in most tissues and appear to have largely redundant functions, the brain is an organ with predominant expression of iRhom1. Yet, little is known about the spatio-temporal expression of iRhom1 in mammalian brain and about its function in controlling membrane protein shedding in the nervous system. Here, we demonstrate that iRhom1 is expressed in mouse brain from the prenatal stage to adulthood with a peak in early postnatal development. In the adult mouse brain iRhom1 was widely expressed, including in cortex, hippocampus, olfactory bulb, and cerebellum. Proteomic analysis of the secretome of primary neurons using the hiSPECS method and of cerebrospinal fluid, obtained from iRhom1-deficient and control mice, identified several membrane proteins that require iRhom1 for their shedding in vitro or in vivo. One of these proteins was 'multiple-EGF-like-domains protein 10' (MEGF10), a phagocytic receptor in the brain that is linked to the removal of amyloid β and apoptotic neurons. MEGF10 was further validated as an ADAM17 substrate using ADAM17-deficient mouse embryonic fibroblasts. Taken together, this study discovers a role for iRhom1 in controlling membrane protein shedding in the mouse brain, establishes MEGF10 as an iRhom1-dependent ADAM17 substrate and demonstrates that iRhom1 is widely expressed in murine brain.
Collapse
Affiliation(s)
- Johanna Tüshaus
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Joshua Shrouder
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Martina Arends
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Mikael Simons
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Carl P Blobel
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, New York, USA.,Department of Medicine, Weill Cornell Medicine, New York, New York, USA.,Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York, USA
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
33
|
Williams BL, Seager NA, Gardiner JD, Pappas CM, Cronin MC, Amat di San Filippo C, Anstadt RA, Liu J, Toso MA, Nichols L, Parnell TJ, Eve JR, Bartel PL, Zouache MA, Richards BT, Hageman GS. Chromosome 10q26-driven age-related macular degeneration is associated with reduced levels of HTRA1 in human retinal pigment epithelium. Proc Natl Acad Sci U S A 2021; 118:e2103617118. [PMID: 34301870 PMCID: PMC8325339 DOI: 10.1073/pnas.2103617118] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/07/2021] [Indexed: 11/18/2022] Open
Abstract
Genome-wide association studies have identified the chromosome 10q26 (Chr10) locus, which contains the age-related maculopathy susceptibility 2 (ARMS2) and high temperature requirement A serine peptidase 1 (HTRA1) genes, as the strongest genetic risk factor for age-related macular degeneration (AMD) [L.G. Fritsche et al., Annu. Rev. Genomics Hum. Genet. 15, 151-171, (2014)]. To date, it has been difficult to assign causality to any specific single nucleotide polymorphism (SNP), haplotype, or gene within this region because of high linkage disequilibrium among the disease-associated variants [J. Jakobsdottir et al. Am. J. Hum. Genet. 77, 389-407 (2005); A. Rivera et al. Hum. Mol. Genet. 14, 3227-3236 (2005)]. Here, we show that HTRA1 messenger RNA (mRNA) is reduced in retinal pigment epithelium (RPE) but not in neural retina or choroid tissues derived from human donors with homozygous risk at the 10q26 locus. This tissue-specific decrease is mediated by the presence of a noncoding, cis-regulatory element overlapping the ARMS2 intron, which contains a potential Lhx2 transcription factor binding site that is disrupted by risk variant rs36212733. HtrA1 protein increases with age in the RPE-Bruch's membrane (BM) interface in Chr10 nonrisk donors but fails to increase in donors with homozygous risk at the 10q26 locus. We propose that HtrA1, an extracellular chaperone and serine protease, functions to maintain the optimal integrity of the RPE-BM interface during the aging process and that reduced expression of HTRA1 mRNA and protein in Chr10 risk donors impairs this protective function, leading to increased risk of AMD pathogenesis. HtrA1 augmentation, not inhibition, in high-risk patients should be considered as a potential therapy for AMD.
Collapse
Affiliation(s)
- Brandi L. Williams
- Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132
| | - Nathan A. Seager
- Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132
| | - Jamie D. Gardiner
- Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132
| | - Chris M. Pappas
- Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132
| | - Monica C. Cronin
- Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132
| | - Cristina Amat di San Filippo
- Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132
| | - Robert A. Anstadt
- Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132
| | - Jin Liu
- Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132
| | - Marc A. Toso
- Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132
| | - Lisa Nichols
- Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132
| | - Timothy J. Parnell
- Bioinformatics Analysis, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84132
| | - Jacqueline R. Eve
- Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132
| | - Paul L. Bartel
- Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132
| | - Moussa A. Zouache
- Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132
| | - Burt T. Richards
- Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132
| | - Gregory S. Hageman
- Steele Center for Translational Medicine, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132
| |
Collapse
|
34
|
Gravesteijn G, Hack RJ, Mulder AA, Cerfontaine MN, van Doorn R, Hegeman IM, Jost CR, Rutten JW, Lesnik Oberstein SAJ. NOTCH3 variant position is associated with NOTCH3 aggregation load in CADASIL vasculature. Neuropathol Appl Neurobiol 2021; 48:e12751. [PMID: 34297860 PMCID: PMC9291091 DOI: 10.1111/nan.12751] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/15/2021] [Accepted: 07/17/2021] [Indexed: 12/02/2022]
Abstract
Aims CADASIL, the most prevalent hereditary cerebral small vessel disease, is caused by cysteine‐altering NOTCH3 variants (NOTCH3cys) leading to vascular NOTCH3 protein aggregation. It has recently been shown that variants located in one of NOTCH3 protein epidermal growth‐factor like repeat (EGFr) domains 1–6, are associated with a more severe phenotype than variants located in one of the EGFr domains 7–34. The underlying mechanism for this genotype–phenotype correlation is unknown. The aim of this study was to analyse whether NOTCH3cys variant position is associated with NOTCH3 protein aggregation load. Methods We quantified vascular NOTCH3 aggregation in skin biopsies (n = 25) and brain tissue (n = 7) of CADASIL patients with a NOTCH3cys EGFr 1–6 variant or a EGFr 7–34 variant, using NOTCH3 immunohistochemistry (NOTCH3 score) and ultrastructural analysis of granular osmiophilic material (GOM count). Disease severity was assessed by neuroimaging (lacune count and white matter hyperintensity volume) and disability (modified Rankin scale). Results Patients with NOTCH3cys EGFr 7–34 variants had lower NOTCH3 scores (P = 1.3·10−5) and lower GOM counts (P = 8.2·10−5) than patients with NOTCH3cys EGFr 1–6 variants in skin vessels. A similar trend was observed in brain vasculature. In the EGFr 7–34 group, NOTCH3 aggregation levels were associated with lacune count (P = 0.03) and white matter hyperintensity volume (P = 0.02), but not with disability. Conclusions CADASIL patients with an EGFr 7–34 variant have significantly less vascular NOTCH3 aggregation than patients with an EGFr 1–6 variant. This may be one of the factors underlying the difference in disease severity between NOTCH3cys EGFr 7–34 and EGFr 1–6 variants.
Collapse
Affiliation(s)
- Gido Gravesteijn
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Remco J Hack
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Aat A Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Minne N Cerfontaine
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Remco van Doorn
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ingrid M Hegeman
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Carolina R Jost
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Julie W Rutten
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
35
|
Muiño E, Fernández-Cadenas I, Arboix A. Contribution of "Omic" Studies to the Understanding of Cadasil. A Systematic Review. Int J Mol Sci 2021; 22:7357. [PMID: 34298974 PMCID: PMC8304933 DOI: 10.3390/ijms22147357] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/04/2021] [Accepted: 07/05/2021] [Indexed: 12/21/2022] Open
Abstract
CADASIL (Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy) is a small vessel disease caused by mutations in NOTCH3 that lead to an odd number of cysteines in the epidermal growth factor (EGF)-like repeat domain, causing protein misfolding and aggregation. The main symptoms are migraines, psychiatric disorders, recurrent strokes, and dementia. Omic technologies allow the massive study of different molecules for understanding diseases in a non-biased manner or even for discovering targets and their possible treatments. We analyzed the progress in understanding CADASIL that has been made possible by omics sciences. For this purpose, we included studies that focused on CADASIL and used omics techniques, searching bibliographic resources, such as PubMed. We excluded studies with other phenotypes, such as migraine or leukodystrophies. A total of 18 articles were reviewed. Due to the high prevalence of NOTCH3 mutations considered pathogenic to date in genomic repositories, one can ask whether all of them produce CADASIL, different degrees of the disease, or whether they are just a risk factor for small vessel disease. Besides, proteomics and transcriptomics studies found that the molecules that are significantly altered in CADASIL are mainly related to cell adhesion, the cytoskeleton or extracellular matrix components, misfolding control, autophagia, angiogenesis, or the transforming growth factor β (TGFβ) signaling pathway. The omics studies performed on CADASIL have been useful for understanding the biological mechanisms and could be key factors for finding potential drug targets.
Collapse
Affiliation(s)
- Elena Muiño
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
| | - Israel Fernández-Cadenas
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
| | - Adrià Arboix
- Cerebrovascular Division, Department of Neurology, Hospital Universitari del Sagrat Cor, Universitat de Barcelona, 08007 Barcelona, Spain
| |
Collapse
|
36
|
Zhang G, Liu M, Chen H, Wu Z, Gao Y, Ma Z, He X, Kang X. NF-κB signalling pathways in nucleus pulposus cell function and intervertebral disc degeneration. Cell Prolif 2021; 54:e13057. [PMID: 34028920 PMCID: PMC8249791 DOI: 10.1111/cpr.13057] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/25/2021] [Accepted: 05/01/2021] [Indexed: 02/06/2023] Open
Abstract
Intervertebral disc degeneration (IDD) is a common clinical degenerative disease of the spine. A series of factors, such as inflammation, oxidative stress and mechanical stress, promote degradation of the extracellular matrix (ECM) of the intervertebral discs (IVD), leading to dysfunction and structural destruction of the IVD. Nuclear factor-κB (NF-κB) transcription factor has long been regarded as a pathogenic factor of IDD. Therefore, NF-κB may be an ideal therapeutic target for IDD. As NF-κB is a multifunctional functional transcription factor with roles in a variety of biological processes, a comprehensive understanding of the function and regulatory mechanism of NF-κB in IDD pathology will be useful for the development of targeted therapeutic strategies for IDD, which can prevent the progression of IDD and reduce potential risks. This review discusses the role of the NF-κB signalling pathway in the nucleus pulposus (NP) in the process of IDD to understand pathological NP degeneration further and provide potential therapeutic targets that may interfere with NF-κB signalling for IDD therapy.
Collapse
Affiliation(s)
- Guang‐Zhi Zhang
- Department of OrthopedicsLanzhou University Second HospitalLanzhouChina
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
- Key Laboratory of Orthopedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouChina
| | - Ming‐Qiang Liu
- Department of OrthopedicsLanzhou University Second HospitalLanzhouChina
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
- Key Laboratory of Orthopedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouChina
| | - Hai‐Wei Chen
- Department of OrthopedicsLanzhou University Second HospitalLanzhouChina
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
- Key Laboratory of Orthopedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouChina
| | - Zuo‐Long Wu
- Department of OrthopedicsLanzhou University Second HospitalLanzhouChina
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
- Key Laboratory of Orthopedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouChina
| | - Yi‐Cheng Gao
- Department of OrthopedicsLanzhou University Second HospitalLanzhouChina
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
- Key Laboratory of Orthopedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouChina
| | - Zhan‐Jun Ma
- Department of OrthopedicsLanzhou University Second HospitalLanzhouChina
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
- Key Laboratory of Orthopedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouChina
| | - Xue‐Gang He
- Department of OrthopedicsLanzhou University Second HospitalLanzhouChina
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
- Key Laboratory of Orthopedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouChina
| | - Xue‐Wen Kang
- Department of OrthopedicsLanzhou University Second HospitalLanzhouChina
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
- Key Laboratory of Orthopedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouChina
- The International Cooperation Base of Gansu Province for the Pain Research in Spinal DisordersLanzhouChina
| |
Collapse
|
37
|
Cerebral microbleeds in vascular dementia from clinical aspects to host-microbial interaction. Neurochem Int 2021; 148:105073. [PMID: 34048844 DOI: 10.1016/j.neuint.2021.105073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 05/15/2021] [Accepted: 05/16/2021] [Indexed: 12/30/2022]
Abstract
Vascular dementia is the second leading cause of dementia after Alzheimer's disease in the elderly population worldwide. Cerebral microbleeds (CMBs) are frequently observed in MRI of elderly subjects and considered as a possible surrogate marker. The number and location of CMBs reflect the severity of diseases and the underlying pathologies may involve cerebral amyloid angiopathy or hypertensive vasculopathy. Accumulating evidence demonstrated the clinicopathological discrepancies of CMBs, the clinical significance of CMBs associated with other MRI markers of cerebral small vessel disease, cognitive impairments, serum, and cerebrospinal fluid biomarkers. Moreover, emerging evidence has shown that genetic factors and gene-environmental interactions might shed light on the underlying etiologies of CMBs, focusing on blood-brain-barrier and inflammation. In this review, we introduce recent genetic and microbiome studies as a cutting-edge approach to figure out the etiology of CMBs through the "microbe-brain-oral axis" and "microbiome-brain-gut axis." Finally, we propose novel concepts, "microvascular matrisome" and "imbalanced proteostasis," which may provide better perspectives for elucidating the pathophysiology of CMBs and future development of therapeutics for vascular dementia using CMBs as a surrogate marker.
Collapse
|
38
|
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: 54] [Impact Index Per Article: 13.5] [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
|
39
|
Arnardottir S, Del Gaudio F, Klironomos S, Braune EB, Lombraña AA, Oliveira DV, Jin S, Karlström H, Lendahl U, Sjöstrand C. Novel Cysteine-Sparing Hypomorphic NOTCH3 A1604T Mutation Observed in a Family With Migraine and White Matter Lesions. NEUROLOGY-GENETICS 2021; 7:e584. [PMID: 33898742 PMCID: PMC8063633 DOI: 10.1212/nxg.0000000000000584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 02/09/2021] [Indexed: 12/22/2022]
Abstract
Objective To conduct a clinical study of a family with neurologic symptoms and findings carrying a novel NOTCH3 mutation and to analyze the molecular consequences of the mutation. Methods We analyzed a family with complex neurologic symptoms by MRI and neurologic examinations. Exome sequencing of the NOTCH3 locus was conducted, and whole-genome sequencing was performed to identify COL4A1, COL4A2, and HTRA1 mutations. Cell lines expressing the normal or NOTCH3A1604T receptors were analyzed to assess proteolytic processing, cell morphology, receptor routing, and receptor signaling. Results Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common hereditary form of cerebral small vessel disease (SVD) and caused by mutations in the NOTCH3 gene. Most CADASIL mutations alter the number of cysteine residues in the extracellular domain of the NOTCH3 receptor, but in this article, we describe a family in which some members carry a novel cysteine-sparing NOTCH3 mutation (c.4810 G>A, p.Ala1604Thr). Two of 3 siblings heterozygous for the NOTCH3A1604T mutation presented with migraine and white matter lesions (WMLs), the latter of a type related to but distinct from what is normally observed in CADASIL. Two other members instead carried a novel COL4A1 missense mutation (c.4795 G>A; p.(Ala1599Thr)). The NOTCH3A1604T receptor was aberrantly processed, showed reduced presence at the cell surface, and less efficiently activated Notch downstream target genes. Conclusions We identify a family with migraine and WML in which some members carry a cysteine-sparing hypomorphic NOTCH3 mutation. Although a causal relationship is not established, we believe that the observations contribute to the discussion on dysregulated Notch signaling in cerebral SVDs.
Collapse
Affiliation(s)
- Snjolaug Arnardottir
- Division of Neurology (S.A., S.K., C.S.), Department of Clinical Neuroscience, Karolinska Institutet; Department of Cell and Molecular Biology (F.D.G., E.-B.B., A.A.L., S.J., U.L.), Karolinska Institutet; and Department of Neurobiology, Care Sciences and Society (D.O., H.K., U.L.), Karolinska Institutet, Stockholm, Sweden
| | - Francesca Del Gaudio
- Division of Neurology (S.A., S.K., C.S.), Department of Clinical Neuroscience, Karolinska Institutet; Department of Cell and Molecular Biology (F.D.G., E.-B.B., A.A.L., S.J., U.L.), Karolinska Institutet; and Department of Neurobiology, Care Sciences and Society (D.O., H.K., U.L.), Karolinska Institutet, Stockholm, Sweden
| | - Stefanos Klironomos
- Division of Neurology (S.A., S.K., C.S.), Department of Clinical Neuroscience, Karolinska Institutet; Department of Cell and Molecular Biology (F.D.G., E.-B.B., A.A.L., S.J., U.L.), Karolinska Institutet; and Department of Neurobiology, Care Sciences and Society (D.O., H.K., U.L.), Karolinska Institutet, Stockholm, Sweden
| | - Eike-Benjamin Braune
- Division of Neurology (S.A., S.K., C.S.), Department of Clinical Neuroscience, Karolinska Institutet; Department of Cell and Molecular Biology (F.D.G., E.-B.B., A.A.L., S.J., U.L.), Karolinska Institutet; and Department of Neurobiology, Care Sciences and Society (D.O., H.K., U.L.), Karolinska Institutet, Stockholm, Sweden
| | - Ariane Araujo Lombraña
- Division of Neurology (S.A., S.K., C.S.), Department of Clinical Neuroscience, Karolinska Institutet; Department of Cell and Molecular Biology (F.D.G., E.-B.B., A.A.L., S.J., U.L.), Karolinska Institutet; and Department of Neurobiology, Care Sciences and Society (D.O., H.K., U.L.), Karolinska Institutet, Stockholm, Sweden
| | - Daniel V Oliveira
- Division of Neurology (S.A., S.K., C.S.), Department of Clinical Neuroscience, Karolinska Institutet; Department of Cell and Molecular Biology (F.D.G., E.-B.B., A.A.L., S.J., U.L.), Karolinska Institutet; and Department of Neurobiology, Care Sciences and Society (D.O., H.K., U.L.), Karolinska Institutet, Stockholm, Sweden
| | - Shaobo Jin
- Division of Neurology (S.A., S.K., C.S.), Department of Clinical Neuroscience, Karolinska Institutet; Department of Cell and Molecular Biology (F.D.G., E.-B.B., A.A.L., S.J., U.L.), Karolinska Institutet; and Department of Neurobiology, Care Sciences and Society (D.O., H.K., U.L.), Karolinska Institutet, Stockholm, Sweden
| | - Helena Karlström
- Division of Neurology (S.A., S.K., C.S.), Department of Clinical Neuroscience, Karolinska Institutet; Department of Cell and Molecular Biology (F.D.G., E.-B.B., A.A.L., S.J., U.L.), Karolinska Institutet; and Department of Neurobiology, Care Sciences and Society (D.O., H.K., U.L.), Karolinska Institutet, Stockholm, Sweden
| | - Urban Lendahl
- Division of Neurology (S.A., S.K., C.S.), Department of Clinical Neuroscience, Karolinska Institutet; Department of Cell and Molecular Biology (F.D.G., E.-B.B., A.A.L., S.J., U.L.), Karolinska Institutet; and Department of Neurobiology, Care Sciences and Society (D.O., H.K., U.L.), Karolinska Institutet, Stockholm, Sweden
| | - Christina Sjöstrand
- Division of Neurology (S.A., S.K., C.S.), Department of Clinical Neuroscience, Karolinska Institutet; Department of Cell and Molecular Biology (F.D.G., E.-B.B., A.A.L., S.J., U.L.), Karolinska Institutet; and Department of Neurobiology, Care Sciences and Society (D.O., H.K., U.L.), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
40
|
Muiño E, Maisterra O, Jiménez-Balado J, Cullell N, Carrera C, Torres-Aguila NP, Cárcel-Márquez J, Gallego-Fabrega C, Lledós M, González-Sánchez J, Olmos-Alpiste F, Espejo E, March Á, Pujol R, Rodríguez-Campello A, Romeral G, Krupinski J, Martí-Fàbregas J, Montaner J, Roquer J, Fernández-Cadenas I. Genome-wide transcriptome study in skin biopsies reveals an association of E2F4 with cadasil and cognitive impairment. Sci Rep 2021; 11:6846. [PMID: 33767277 PMCID: PMC7994794 DOI: 10.1038/s41598-021-86349-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 03/11/2021] [Indexed: 01/31/2023] Open
Abstract
CADASIL is a small vessel disease caused by mutations in NOTCH3 that lead to an odd number of cysteines in the EGF-like repeat domain, causing protein misfolding and aggregation. The main symptoms are migraine, psychiatric disturbances, recurrent strokes and dementia, being executive function characteristically impaired. The molecular pathways altered by this receptor aggregation need to be studied further. A genome-wide transcriptome study (four cases paired with three healthy siblings) was carried out, in addition to a qRT-PCR for validation purposes (ten new cases and eight new controls). To study the expression profile by cell type of the significant mRNAs found, we performed an in situ hybridization (ISH) (nine cases and eight controls) and a research in the Single-nuclei Brain RNA-seq expression browser (SNBREB). Pathway analysis enrichment was carried out with Gene Ontology and Reactome. Neuropsychological tests were performed in five of the qRT-PCR cases. The two most significant differentially expressed mRNAs (BANP, p-value = 7.23 × 10-4 and PDCD6IP, p-value = 8.36 × 10-4) were selected for the validation study by qRT-PCR. Additionally, we selected two more mRNAs (CAMK2G, p-value = 4.52 × 10-3 and E2F4, p-value = 4.77 × 10-3) due to their association with ischemic neuronal death. E2F4 showed differential expression in the genome-wide transcriptome study and in the qRT-PCR (p = 1.23 × 10-3), and it was upregulated in CADASIL cases. Furthermore, higher E2F4 expression was associated with worse executive function (p = 2.04 × 10-2) and attention and information processing speed (IPS) (p = 8.73 × 10-2). In situ hibridization showed E2F4 expression in endothelial and vascular smooth vessel cells. In silico studies indicated that E2F4 is also expressed in brain endothelial cells. Among the most significant pathways analyzed, there was an enrichment of vascular development, cell adhesion and vesicular machinery terms and autophagy process. E2F4 is more highly expressed in the skin biopsy of CADASIL patients compared to controls, and its expression is present in endothelial cells and VSMCs. Further studies are needed to understand whether E2F4 could be useful as a biomarker, to monitor the disease or be used as a therapeutic target.
Collapse
Affiliation(s)
- Elena Muiño
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l`Hospital de la Santa Creu i Sant Pau, C/Sant Antoni María Claret 167, Barcelona, Spain
| | - Olga Maisterra
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joan Jiménez-Balado
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Natalia Cullell
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l`Hospital de la Santa Creu i Sant Pau, C/Sant Antoni María Claret 167, Barcelona, Spain
- Stroke Pharmacogenomics and Genetics, Fundació MútuaTerrassa per la Docència i la Recerca, Terrassa, Spain
| | - Caty Carrera
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l`Hospital de la Santa Creu i Sant Pau, C/Sant Antoni María Claret 167, Barcelona, Spain
| | - Nuria P Torres-Aguila
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l`Hospital de la Santa Creu i Sant Pau, C/Sant Antoni María Claret 167, Barcelona, Spain
| | - Jara Cárcel-Márquez
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l`Hospital de la Santa Creu i Sant Pau, C/Sant Antoni María Claret 167, Barcelona, Spain
| | - Cristina Gallego-Fabrega
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l`Hospital de la Santa Creu i Sant Pau, C/Sant Antoni María Claret 167, Barcelona, Spain
- Stroke Pharmacogenomics and Genetics, Fundació MútuaTerrassa per la Docència i la Recerca, Terrassa, Spain
| | - Miquel Lledós
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l`Hospital de la Santa Creu i Sant Pau, C/Sant Antoni María Claret 167, Barcelona, Spain
| | - Jonathan González-Sánchez
- Stroke Pharmacogenomics and Genetics, Fundació MútuaTerrassa per la Docència i la Recerca, Terrassa, Spain
- The Manchester Metropolitan University of All Saints, Manchester, UK
| | | | - Eva Espejo
- Dermatology Department, Hospital del Mar-Parc de Salut Mar, Barcelona, Spain
| | - Álvaro March
- Dermatology Department, Hospital del Mar-Parc de Salut Mar, Barcelona, Spain
| | - Ramón Pujol
- Dermatology Department, Hospital del Mar-Parc de Salut Mar, Barcelona, Spain
| | | | - Gemma Romeral
- Neurology Department, IMIM-Hospital del Mar, Barcelona, Spain
| | - Jurek Krupinski
- Neurology Department, Hospital Mútua Terrassa, Terrassa, Spain
| | - Joan Martí-Fàbregas
- Neurology Department, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Joan Montaner
- The Manchester Metropolitan University of All Saints, Manchester, UK
- Biomedicine Institute of Seville, IBiS/Hospital Universitario Virgen del Rocío/CSIC, University of Seville, Seville, Spain
- Department of Neurology, Hospital Universitario Virgen Macarena, Seville, Spain
| | - Jaume Roquer
- Neurology Department, IMIM-Hospital del Mar, Barcelona, Spain
| | - Israel Fernández-Cadenas
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l`Hospital de la Santa Creu i Sant Pau, C/Sant Antoni María Claret 167, Barcelona, Spain.
| |
Collapse
|
41
|
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: 18] [Impact Index Per Article: 4.5] [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
|
42
|
Solovyeva EM, Moshkovskii SA, Gorshkov MV. Identification-Free Control over the Precursor Isotopic Mass Misassignment in Orbitrap-Based Proteomics. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:218-224. [PMID: 33119294 DOI: 10.1021/jasms.0c00281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Selection of a precursor ion from a peptide isotopic cluster to obtain a fragmentation mass spectrum is a crucial step in data-dependent proteome analysis. However, the monoisotopic mass assignment performed in this step is often an issue confronted by the data acquisition software of hybrid Orbitrap FTMS that is most widely used in proteomics. To address the problem, many data processing tools, such as raw data converters and search engines, have optional accounting for the precursor mass shift due to the isotopic error. These solutions require additional data preprocessing steps and lead to an increase in the search space, thus making the analysis longer and/or less reliable. In this work, we processed 100 Orbitrap-based LC-MS/MS runs from 10 publicly available data sets to examine the rate of precursor isotope misassignment. The effect from taking the isotope error into account during the search on the number of identified peptides varied in a wide range from 0 to 33%. Thus, it may be tempting to spend extra time before or during a search to account for the mass assignment issue. Alternatively, this effect can be predicted a priori using an identification-free metric, which can be a part of data quality control software. Based on the results obtained in this work, we propose such a metric be further added into the visual and intuitive quality control software, viQC, developed previously and available at https://github.com/lisavetasol/viQC. It takes about a minute to calculate and plot nine quality metrics, including the proposed one for typical proteome analysis.
Collapse
Affiliation(s)
- Elizaveta M Solovyeva
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Moscow Region 141701, Russia
- V.L. Talrose Institute for Energy Problems of Chemical Physics, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | - Sergei A Moshkovskii
- Pirogov Russian National Research Medical University, Moscow 117997, Russia
- Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow 119435, Russia
| | - Mikhail V Gorshkov
- V.L. Talrose Institute for Energy Problems of Chemical Physics, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| |
Collapse
|
43
|
Young KZ, Xu G, Keep SG, Borjigin J, Wang MM. Overlapping Protein Accumulation Profiles of CADASIL and CAA: Is There a Common Mechanism Driving Cerebral Small-Vessel Disease? THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:1871-1887. [PMID: 33387456 DOI: 10.1016/j.ajpath.2020.11.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/04/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and cerebral amyloid angiopathy (CAA) are two distinct vascular angiopathies that share several similarities in clinical presentation and vascular pathology. Given the clinical and pathologic overlap, the molecular overlap between CADASIL and CAA was explored. CADASIL and CAA protein profiles from recently published proteomics-based and immuno-based studies were compared to investigate the potential for shared disease mechanisms. A comparison of affected proteins in each disease highlighted 19 proteins that are regulated in both CADASIL and CAA. Functional analysis of the shared proteins predicts significant interaction between them and suggests that most enriched proteins play roles in extracellular matrix structure and remodeling. Proposed models to explain the observed enrichment of extracellular matrix proteins include both increased protein secretion and decreased protein turnover by sequestration of chaperones and proteases or formation of stable protein complexes. Single-cell RNA sequencing of vascular cells in mice suggested that the vast majority of the genes accounting for the overlapped proteins between CADASIL and CAA are expressed by fibroblasts. Thus, our current understanding of the molecular profiles of CADASIL and CAA appears to support potential for common mechanisms underlying the two disorders.
Collapse
Affiliation(s)
- Kelly Z Young
- Departments of Neurology, University of Michigan, Ann Arbor, Michigan; Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Gang Xu
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Simon G Keep
- Departments of Neurology, University of Michigan, Ann Arbor, Michigan
| | - Jimo Borjigin
- Departments of Neurology, University of Michigan, Ann Arbor, Michigan; Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Michael M Wang
- Departments of Neurology, University of Michigan, Ann Arbor, Michigan; Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan; Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan.
| |
Collapse
|
44
|
PIP 2: A critical regulator of vascular ion channels hiding in plain sight. Proc Natl Acad Sci U S A 2020; 117:20378-20389. [PMID: 32764146 PMCID: PMC7456132 DOI: 10.1073/pnas.2006737117] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The phosphoinositide, phosphatidylinositol 4,5-bisphosphate (PIP2), has long been established as a major contributor to intracellular signaling, primarily by virtue of its role as a substrate for phospholipase C (PLC). Signaling by Gq-protein-coupled receptors triggers PLC-mediated hydrolysis of PIP2 into inositol 1,4,5-trisphosphate and diacylglycerol, which are well known to modulate vascular ion channel activity. Often overlooked, however, is the role PIP2 itself plays in this regulation. Although numerous reports have demonstrated that PIP2 is critical for ion channel regulation, how it impacts vascular function has received scant attention. In this review, we focus on PIP2 as a regulator of ion channels in smooth muscle cells and endothelial cells-the two major classes of vascular cells. We further address the concerted effects of such regulation on vascular function and blood flow control. We close with a consideration of current knowledge regarding disruption of PIP2 regulation of vascular ion channels in disease.
Collapse
|
45
|
Andjelkovic AV, Stamatovic SM, Phillips CM, Martinez-Revollar G, Keep RF. Modeling blood-brain barrier pathology in cerebrovascular disease in vitro: current and future paradigms. Fluids Barriers CNS 2020; 17:44. [PMID: 32677965 PMCID: PMC7367394 DOI: 10.1186/s12987-020-00202-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022] Open
Abstract
The complexity of the blood-brain barrier (BBB) and neurovascular unit (NVU) was and still is a challenge to bridge. A highly selective, restrictive and dynamic barrier, formed at the interface of blood and brain, the BBB is a "gatekeeper" and guardian of brain homeostasis and it also acts as a "sensor" of pathological events in blood and brain. The majority of brain and cerebrovascular pathologies are associated with BBB dysfunction, where changes at the BBB can lead to or support disease development. Thus, an ultimate goal of BBB research is to develop competent and highly translational models to understand mechanisms of BBB/NVU pathology and enable discovery and development of therapeutic strategies to improve vascular health and for the efficient delivery of drugs. This review article focuses on the progress being made to model BBB injury in cerebrovascular diseases in vitro.
Collapse
Affiliation(s)
- Anuska V Andjelkovic
- Department of Pathology, University of Michigan Medical School, 7520 MSRB I, 1150 West Medical Center Dr, Ann Arbor, MI, 48109-5602, USA.
| | - Svetlana M Stamatovic
- Department of Pathology, University of Michigan Medical School, 7520 MSRB I, 1150 West Medical Center Dr, Ann Arbor, MI, 48109-5602, USA
| | - Chelsea M Phillips
- Graduate Program in Neuroscience, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gabriela Martinez-Revollar
- Department of Pathology, University of Michigan Medical School, 7520 MSRB I, 1150 West Medical Center Dr, Ann Arbor, MI, 48109-5602, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Molecular Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
46
|
Kato T, Sekine Y, Nozaki H, Uemura M, Ando S, Hirokawa S, Onodera O. Excessive Production of Transforming Growth Factor β1 Causes Mural Cell Depletion From Cerebral Small Vessels. Front Aging Neurosci 2020; 12:151. [PMID: 32581764 PMCID: PMC7283554 DOI: 10.3389/fnagi.2020.00151] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 05/05/2020] [Indexed: 12/14/2022] Open
Abstract
It is increasingly becoming apparent that cerebrovascular dysfunction contributes to the pathogenic processes involved in vascular dementia, Alzheimer’s disease, and other neurodegenerative disorders. Under these pathologic conditions, the degeneration of cerebral blood vessels is frequently accompanied by a loss of mural cells from the vascular walls. Vascular mural cells play pivotal roles in cerebrovascular functions, such as regulation of cerebral blood flow and maintenance of the blood-brain barrier (BBB). Therefore, cerebrovascular mural cell impairment is involved in the pathophysiology of vascular-related encephalopathies, and protecting these cells is essential for maintaining brain health. However, our understanding of the molecular mechanism underlying mural cell abnormalities is incomplete. Several reports have indicated that dysregulated transforming growth factor β (TGFβ) signaling is involved in the development of cerebral arteriopathies. These studies have specifically suggested the involvement of TGFβ overproduction. Although cerebrovascular toxicity via vascular fibrosis by extracellular matrix accumulation or amyloid deposition is known to occur with enhanced TGFβ production, whether increased TGFβ results in the degeneration of vascular mural cells in vivo remains unknown. Here, we demonstrated that chronic TGFβ1 overproduction causes a dropout of mural cells and reduces their coverage on cerebral vessels in both smooth muscle cells and pericytes. Mural cell degeneration was also accompanied by vascular luminal dilation. TGFβ1 overproduction in astrocytes significantly increased TGFβ1 content in the cerebrospinal fluid (CSF) and increased TGFβ signaling-regulated gene expression in both pial arteries and brain capillaries. These results indicate that TGFβ is an important effector that mediates mural cell abnormalities under pathological conditions related to cerebral arteriopathies.
Collapse
Affiliation(s)
- Taisuke Kato
- Department of System Pathology for Neurological Disorders, Brain Science Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yumi Sekine
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hiroaki Nozaki
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masahiro Uemura
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Shoichiro Ando
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Sachiko Hirokawa
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| | - Osamu Onodera
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
47
|
Ma Y, Ueda M, Ueda A, Shinriki S, Nagatoshi A, Isoguchi A, Okada M, Tasaki M, Nomura T, Inoue Y, Masuda T, Misumi Y, Yamashita T, Matsui H, Ando Y. Novel dot-blot assay for detection of vascular Notch3 aggregates in patients with CADASIL. J Neurol Sci 2020; 415:116931. [PMID: 32470649 DOI: 10.1016/j.jns.2020.116931] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 05/05/2020] [Accepted: 05/17/2020] [Indexed: 11/19/2022]
Abstract
To detect vascular Notch3 extracellular domain aggregates in CADASIL, we developed a novel dot-blot assay with both autopsy and biopsy skin samples. We obtained samples from 11 patients with CADASIL and 12 control patients, and we performed dot-blot analyses by using sequential biochemical tissue extractions with three different antibodies against specific regions of the Notch3 extracellular domain. We also analyzed clinical features and vascular accumulations of Notch3 by immunohistochemistry. Via the dot-blot assay with the antibody against the C-terminal region of the Notch3 extracellular domain, we successfully detected Notch3 extracellular domain aggregates in skin tissue homogenates obtained from patients with CADASIL. Our novel method may therefore aid the diagnosis of CADASIL.
Collapse
Affiliation(s)
- Yihong Ma
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Mitsuharu Ueda
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan.
| | - Akihiko Ueda
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Satoru Shinriki
- Department of Molecular Laboratory Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Akihito Nagatoshi
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan; Department of Neurology, Kumamoto General Hospital, Yatsushiro 866-8660, Japan
| | - Aito Isoguchi
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Masamitsu Okada
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Masayoshi Tasaki
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan; Department of Morphological and Physiological Sciences, Graduate School of Health Sciences, Kumamoto University, Kumamoto 862-0976, Japan
| | - Toshiya Nomura
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Yasuteru Inoue
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Teruaki Masuda
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Yohei Misumi
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Taro Yamashita
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Hirotaka Matsui
- Department of Molecular Laboratory Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Yukio Ando
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan; Department of Amyloidosis Research, Nagasaki International University, Sasebo 859-3298, Japan
| |
Collapse
|
48
|
Yamamoto Y, Kojima K, Taura D, Sone M, Washida K, Egawa N, Kondo T, Minakawa EN, Tsukita K, Enami T, Tomimoto H, Mizuno T, Kalaria RN, Inagaki N, Takahashi R, Harada-Shiba M, Ihara M, Inoue H. Human iPS cell-derived mural cells as an in vitro model of hereditary cerebral small vessel disease. Mol Brain 2020; 13:38. [PMID: 32188464 PMCID: PMC7081541 DOI: 10.1186/s13041-020-00573-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023] Open
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is one of the most common forms of hereditary cerebral small vessel diseases and is caused by mutations in NOTCH3. Our group has previously reported incorporation of NOTCH3 extracellular domain (N3ECD) in the CADASIL-specific granular osmiophilic materials and increase of PDGFRβ immunoreactivity in CADASIL postmortem brains. Here, we aimed to establish an in vitro model of CADASIL, which can recapitulate those CADASIL phenotypes, using induced pluripotent stem cells (iPSCs). We have refined a differentiation protocol of endothelial cells to obtain mature mural cells (MCs) with their characteristic properties. iPSCs from three CADASIL patients with p.Arg182Cys, p.Arg141Cys and p.Cys106Arg mutations were differentiated into MCs and their functional and molecular profiles were compared. The differentiated CADASIL MCs recapitulated pathogenic changes reported previously: increased PDGFRβ and abnormal structure/distribution of filamentous actin network, as well as N3ECD/LTBP-1/HtrA1-immunopositive deposits. Migration rate of CADASIL MCs was enhanced but suppressed by knockdown of NOTCH3 or PDGFRB. CADASIL MCs showed altered reactivity to PDGF-BB. Patient-derived MCs can recapitulate CADASIL pathology and are therefore useful in understanding the pathogenesis and developing potential treatment strategies.
Collapse
Affiliation(s)
- Yumi Yamamoto
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan.,Department of Molecular Innovation in Lipidemiology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibeshinmachi, Suita-shi, Osaka, 564-0018, Japan
| | - Katsutoshi Kojima
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Daisuke Taura
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Masakatsu Sone
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Kazuo Washida
- Department of Stroke and Cerebrovascular Diseases, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita-shi, Osaka, 564-0018, Japan
| | - Naohiro Egawa
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan.,Department of Neurology, Kyoto University Graduate School of Medicine, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takayuki Kondo
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan.,Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Eiko N Minakawa
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan
| | - Kayoko Tsukita
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan
| | - Takako Enami
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.,Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Hidekazu Tomimoto
- Department of Dementia Prevention and Therapeutics, Graduate School of Medicine, Mie University, 2-174 Edobashi Tsu, Mie, 514-8507, Japan
| | - Toshiki Mizuno
- Department of Neurology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Raj N Kalaria
- Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing & Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Mariko Harada-Shiba
- Department of Molecular Innovation in Lipidemiology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibeshinmachi, Suita-shi, Osaka, 564-0018, Japan
| | - Masafumi Ihara
- Department of Stroke and Cerebrovascular Diseases, National Cerebral and Cardiovascular Center, 6-1 Kishibeshinmachi, Suita-shi, Osaka, 564-0018, Japan.
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan. .,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan. .,Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan.
| |
Collapse
|
49
|
Haffner C. Proteostasis in Cerebral Small Vessel Disease. Front Neurosci 2019; 13:1142. [PMID: 31798396 PMCID: PMC6874119 DOI: 10.3389/fnins.2019.01142] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/10/2019] [Indexed: 01/02/2023] Open
Abstract
Maintaining the homeostasis of proteins (proteostasis) by controlling their synthesis, folding and degradation is a central task of cells and tissues. The gradual decline of the capacity of the various proteostasis machineries, frequently in combination with their overload through mutated, aggregation-prone proteins, is increasingly recognized as an important catalyst of age-dependent pathologies in the brain, most prominently neurodegenerative disorders. A dysfunctional proteostasis might also contribute to neurovascular disease as indicated by the occurrence of excessive protein accumulation or massive extracellular matrix expansion within vessel walls in conditions such as cerebral small vessel disease (SVD), a major cause of ischemic stroke, and cerebral amyloid angiopathy. Recent advances in brain vessel isolation techniques and mass spectrometry methodology have facilitated the analysis of cerebrovascular proteomes and fueled efforts to determine the proteomic signatures associated with neurovascular disease. In several studies in humans and mice considerable differences between healthy and diseased vessel proteomes were observed, emphasizing the critical contribution of an impaired proteostasis to disease pathogenesis. These findings highlight the important role of a balanced proteostasis for cerebrovascular health.
Collapse
Affiliation(s)
- Christof Haffner
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
50
|
Lendahl U, Nilsson P, Betsholtz C. Emerging links between cerebrovascular and neurodegenerative diseases-a special role for pericytes. EMBO Rep 2019; 20:e48070. [PMID: 31617312 PMCID: PMC6831996 DOI: 10.15252/embr.201948070] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/11/2019] [Accepted: 06/24/2019] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative and cerebrovascular diseases cause considerable human suffering, and therapy options for these two disease categories are limited or non-existing. It is an emerging notion that neurodegenerative and cerebrovascular diseases are linked in several ways, and in this review, we discuss the current status regarding vascular dysregulation in neurodegenerative disease, and conversely, how cerebrovascular diseases are associated with central nervous system (CNS) degeneration and dysfunction. The emerging links between neurodegenerative and cerebrovascular diseases are reviewed with a particular focus on pericytes-important cells that ensheath the endothelium in the microvasculature and which are pivotal for blood-brain barrier function and cerebral blood flow. Finally, we address how novel molecular and cellular insights into pericytes and other vascular cell types may open new avenues for diagnosis and therapy development for these important diseases.
Collapse
Affiliation(s)
- Urban Lendahl
- Department of Cell and Molecular BiologyKarolinska InstitutetStockholmSweden
- Department of Neurobiology, Care Sciences and SocietyDivision of NeurogeriatricsCenter for Alzheimer ResearchKarolinska InstitutetSolnaSweden
- Integrated Cardio Metabolic Centre (ICMC)HuddingeSweden
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and SocietyDivision of NeurogeriatricsCenter for Alzheimer ResearchKarolinska InstitutetSolnaSweden
| | - Christer Betsholtz
- Integrated Cardio Metabolic Centre (ICMC)HuddingeSweden
- Department of Immunology, Genetics and PathologyRudbeck LaboratoryUppsala UniversityUppsalaSweden
- Department of MedicineKarolinska InstitutetHuddingeSweden
| |
Collapse
|