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
|
Predtechenskaya EV, Rogachev AD, Melnikova PM. The Characteristics of the Metabolomic Profile in Patients with Parkinson's Disease and Vascular Parkinsonism. Acta Naturae 2024; 16:27-37. [PMID: 39877011 PMCID: PMC11771845 DOI: 10.32607/actanaturae.27511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/21/2024] [Indexed: 01/31/2025] Open
Abstract
The gradually increasing age of the world population implies that the prevalence of neurodegenerative diseases also continues to rise. These diseases are characterized by a progressive loss of cognitive and motor functions. Parkinson's disease, which involves the gradual death of specialized neural tissue, is a striking example of a neurodegenerative process. The pathomorphological analysis shows that chronic cerebral ischemia is accompanied by extensive complex neurodegeneration; parkinsonism is its clinical manifestation in 20-30% of cases. Although Parkinson's disease and vascular parkinsonism are similar, these two pathologies have fundamentally different etiopathogeneses. But their set of differential diagnosis traits is confined to some features of the neurological status. There currently exist no diagnostic markers for individual neurodegenerative pathologies or the neurodegeneration phenomenon in general. Metabolomic profiling can be a promising means for finding a unique "fingerprint" of the disease. Identifying the biomarkers of various neurodegenerative diseases will help shorten the time to the diagnosis, forecast the course of the disease, and personalize the therapeutic approach. This review summarizes and compares the current concepts of metabolomics research into Parkinson's disease and vascular parkinsonism, as well as the respective animal models.
Collapse
Affiliation(s)
| | - A. D. Rogachev
- Novosibirsk State University, Novosibirsk, 630090 Russian Federation
| | - P. M. Melnikova
- Novosibirsk State University, Novosibirsk, 630090 Russian Federation
| |
Collapse
|
3
|
Zedde M, Romani I, Scaravilli A, Cocozza S, Trojano L, Ragno M, Rifino N, Bersano A, Gerevini S, Pantoni L, Valzania F, Pascarella R. Expanding the Neurological Phenotype of Anderson-Fabry Disease: Proof of Concept for an Extrapyramidal Neurodegenerative Pattern and Comparison with Monogenic Vascular Parkinsonism. Cells 2024; 13:1131. [PMID: 38994983 PMCID: PMC11240674 DOI: 10.3390/cells13131131] [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: 06/02/2024] [Revised: 06/18/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
Anderson-Fabry disease (AFD) is a genetic sphingolipidosis involving virtually the entire body. Among its manifestation, the involvement of the central and peripheral nervous system is frequent. In recent decades, it has become evident that, besides cerebrovascular damage, a pure neuronal phenotype of AFD exists in the central nervous system, which is supported by clinical, pathological, and neuroimaging data. This neurodegenerative phenotype is often clinically characterized by an extrapyramidal component similar to the one seen in prodromal Parkinson's disease (PD). We analyzed the biological, clinical pathological, and neuroimaging data supporting this phenotype recently proposed in the literature. Moreover, we compared the neurodegenerative PD phenotype of AFD with a classical monogenic vascular disease responsible for vascular parkinsonism and cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). A substantial difference in the clinical and neuroimaging features of neurodegenerative and vascular parkinsonism phenotypes emerged, with AFD being potentially responsible for both forms of the extrapyramidal involvement, and CADASIL mainly associated with the vascular subtype. The available studies share some limitations regarding both patients' information and neurological and genetic investigations. Further studies are needed to clarify the potential association between AFD and extrapyramidal manifestations.
Collapse
Affiliation(s)
- Marialuisa Zedde
- Neurology Unit, Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy;
| | - Ilaria Romani
- Department of Neurosciences, Psychology, Pharmacology and Child Health, University of Florence, 50139 Firenze, Italy;
| | - Alessandra Scaravilli
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80133 Napoli, Italy; (A.S.); (S.C.)
| | - Sirio Cocozza
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80133 Napoli, Italy; (A.S.); (S.C.)
| | - Luigi Trojano
- Dipartimento di Psicologia, Università della Campania ‘Luigi Vanvitelli’, viale Ellittico 31, 81100 Caserta, Italy;
| | - Michele Ragno
- Centro Medico Salute 23, Via O. Licini 5, 63066 Grottammare (AP), Italy;
| | - Nicola Rifino
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milano, Italy; (N.R.); (A.B.)
| | - Anna Bersano
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milano, Italy; (N.R.); (A.B.)
| | - Simonetta Gerevini
- Head Diagnostic Dept and Neuroradiology Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy;
| | - Leonardo Pantoni
- Neuroscience Research Center, Department of Biomedical and Clinical Science, University of Milan, 20122 Milano, Italy;
| | - Franco Valzania
- Neurology Unit, Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy;
| | - Rosario Pascarella
- Neuroradiology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy;
| |
Collapse
|
4
|
Perosa V, Zanon Zotin MC, Schoemaker D, Sveikata L, Etherton MR, Charidimou A, Greenberg SM, Viswanathan A. Association Between Hippocampal Volumes and Cognition in Cerebral Amyloid Angiopathy. Neurology 2024; 102:e207854. [PMID: 38165326 PMCID: PMC10870737 DOI: 10.1212/wnl.0000000000207854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 10/03/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Accumulating evidence suggests that gray matter atrophy, often considered a marker of Alzheimer disease (AD), can also result from cerebral small vessel disease (CSVD). Cerebral amyloid angiopathy (CAA) is a form of sporadic CSVD, diagnosed through neuroimaging criteria, that often co-occurs with AD pathology and leads to cognitive impairment. We sought to identify the role of hippocampal integrity in the development of cognitive impairment in a cohort of patients with possible and probable CAA. METHODS Patients were recruited from an ongoing CAA study at Massachusetts General Hospital. Composite scores defined performance in the cognitive domains of memory, language, executive function, and processing speed. Hippocampal subfields' volumes were measured from 3T MRI, using an automated method, and multivariate linear regression models were used to estimate their association with each cognitive domain and relationship to CAA-related neuroimaging markers. RESULTS One hundred twenty patients, 36 with possible (age mean [range]: 75.6 [65.6-88.9]), 67 with probable CAA (75.9 [59.0-94.0]), and 17 controls without cognitive impairment and CSVD (72.4 [62.5-82.7]; 76.4% female patients), were included in this study. We found a positive association between all investigated hippocampal subfields and memory and language, whereas specific subfields accounted for executive function (CA4 [Estimate = 5.43; 95% CI 1.26-9.61; p = 0.020], subiculum [Estimate = 2.85; 95% CI 0.67-5.02; p = 0.022]), and processing speed (subiculum [Estimate = 1.99; 95% CI 0.13-3.85; p = 0.036]). These findings were independent of other CAA-related markers, which did not have an influence on cognition in this cohort. Peak width of skeletonized mean diffusivity (PSMD), a measure of white matter integrity, was negatively associated with hippocampal subfields' volumes (CA3 [Estimate = -0.012; 95% CI -0.020 to -0.004; p = 0.034], CA4 [Estimate = -0.010; 95% CI -0.020 to -0.0007; p = 0.037], subiculum [Estimate = -0.019; 95% CI -0.042 to -0.0001; p = 0.003]). DISCUSSION These results suggest that hippocampal integrity is an independent contributor to cognitive impairment in patients with CAA and that it might be related to loss of integrity in the white matter. Further studies exploring potential causes and directionality of the relationship between white matter and hippocampal integrity may be warranted.
Collapse
Affiliation(s)
- Valentina Perosa
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Maria Clara Zanon Zotin
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Dorothee Schoemaker
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Lukas Sveikata
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Mark R Etherton
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Andreas Charidimou
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Steven M Greenberg
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Anand Viswanathan
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
5
|
Wang W, Gong Z, Wang Y, Zhao Y, Lu Y, Sun R, Zhang H, Shang J, Zhang J. Mutant NOTCH3ECD Triggers Defects in Mitochondrial Function and Mitophagy in CADASIL Cell Models. J Alzheimers Dis 2024; 100:1299-1314. [PMID: 39031358 DOI: 10.3233/jad-240273] [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] [Indexed: 07/22/2024]
Abstract
Background Cerebral autosomal-dominant arteriopathy with subcortical infarction and leukoencephalopathy (CADASIL) is an inherited small-vessel disease that affects the white matter of the brain. Recent studies have confirmed that the deposition of NOTCH3ECD is the main pathological basis of CADASIL; however, whether different mutations present the same pathological characteristics remains to be further studied. Some studies have found that mitochondrial dysfunction is related to CADASIL; however, the specific effects of NOTCH3ECD on mitochondrial remain to be determined. Objective We aimed to explore the role of mitochondrial dysfunction in CADASIL. Methods We established transgenic human embryonic kidney-293T cell models (involving alterations in cysteine and non-cysteine residues) via lentiviral transfection. Mitochondrial function and structure were assessed using flow cytometry and transmission electron microscopy, respectively. Mitophagy was assessed using western blotting and immunofluorescence. Results We demonstrated that NOTCH3ECD deposition affects mitochondrial morphology and function, and that its protein levels are significantly correlated with mitochondrial quality and can directly bind to mitochondria. Moreover, NOTCH3ECD deposition promoted the induction of autophagy and mitophagy. However, these processes were impaired, leading to abnormal mitochondrial accumulation. Conclusions This study revealed a common pathological feature of NOTCH3ECD deposition caused by different NOTCH3 mutations and provided new insights into the role of NOTCH3ECD in mitochondrial dysfunction and mitophagy.
Collapse
Affiliation(s)
- Wan Wang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Zhenping Gong
- Department of Neurology, Xinxiang medical university, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Yadan Wang
- Department of Neurology, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Ying Zhao
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Yaru Lu
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Ruihua Sun
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Haohan Zhang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Junkui Shang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Jiewen Zhang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| |
Collapse
|
6
|
Ding X, Chen Y, Guo C, Fu Y, Qin C, Zhu Q, Wang J, Zhang R, Tian H, Feng R, Liu H, Liang D, Wang G, Teng J, Li J, Tang B, Wang X. Mutations in ARHGEF15 cause autosomal dominant hereditary cerebral small vessel disease and osteoporotic fracture. Acta Neuropathol 2023; 145:681-705. [PMID: 36929019 DOI: 10.1007/s00401-023-02560-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/26/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023]
Abstract
Cerebral small vessel disease (CSVD) is a prominent cause of ischemic and hemorrhagic stroke and a leading cause of vascular dementia, affecting small penetrating vessels of the brain. Despite current advances in genetic susceptibility studies, challenges remain in defining the causative genes and the underlying pathophysiological mechanisms. Here, we reported that the ARHGEF15 gene was a causal gene linked to autosomal dominant inherited CSVD. We identified one heterozygous nonsynonymous mutation of the ARHGEF15 gene that cosegregated completely in two families with CSVD, and a heterozygous nonsynonymous mutation and a stop-gain mutation in two individuals with sporadic CSVD, respectively. Intriguingly, clinical imaging and pathological findings displayed severe osteoporosis and even osteoporotic fractures in all the ARHGEF15 mutation carriers. In vitro experiments indicated that ARHGEF15 mutations resulted in RhoA/ROCK2 inactivation-induced F-actin cytoskeleton disorganization in vascular smooth muscle cells and endothelial cells and osteoblast dysfunction by inhibiting the Wnt/β-catenin signaling pathway in osteoblast cells. Furthermore, Arhgef15-e(V368M)1 transgenic mice developed CSVD-like pathological and behavioral phenotypes, accompanied by severe osteoporosis. Taken together, our findings provide strong evidence that loss-of-function mutations of the ARHGEF15 gene cause CSVD accompanied by osteoporotic fracture.
Collapse
Affiliation(s)
- Xuebing Ding
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Yongkang Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Cancan Guo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Yu Fu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Chi Qin
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Qingyong Zhu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Jiuqi Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Rui Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Haiyan Tian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Renyi Feng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Han Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Dongxiao Liang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Guanghui Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases &, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Junfang Teng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Jinchen Li
- Bioinformatics Center, National Clinical Research Centre for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China.
| | - Beisha Tang
- The First Affiliated Hospital, Multi-Omics Research Center for Brain Disorders, Hengyang Medical School, University of South China, Hengyang, China.
| | - Xuejing Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
7
|
Prajjwal P, Marsool MDM, Inban P, Sharma B, Asharaf S, Aleti S, Gadam S, Al Sakini AS, Hadi DD. Vascular dementia subtypes, pathophysiology, genetics, neuroimaging, biomarkers, and treatment updates along with its association with Alzheimer's dementia and diabetes mellitus. Dis Mon 2023; 69:101557. [PMID: 37031059 DOI: 10.1016/j.disamonth.2023.101557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2023]
Abstract
Dementia is a chronic progressive cognitive decline illness that results in functional impairment. Vascular dementia (VaD), second only to Alzheimer's disease (AD), is one of the most prevalent forms of dementia in the elderly (aged over 65 years), with a varied presentation and unpredictable disease development caused by cerebrovascular or cardiovascular illness. To get a better understanding of the changes occurring in the brain and to drive therapy efforts, new biomarkers for early and precise diagnosis of AD and VaD are required. In this review, Firstly, we describe the subtypes of vascular dementia, their clinical features, pathogenesis, genetics implemented, and their associated neuroimaging and biomarkers, while describing extensively the recent biomarkers discovered in the literature. Secondly, we describe some of the well-documented and other less-defined risk factors and their association and pathophysiology in relation to vascular dementia. Finally, we follow recent updates in the management of vascular dementia along with its association and differentiation from Alzheimer's disease. The aim of this review is to gather the scattered updates and the most recent changes in blood, CSF, and neuroimaging biomarkers related to the multiple subtypes of vascular dementia along with its association with Alzheimer's dementia and diabetes mellitus.
Collapse
Affiliation(s)
| | | | - Pugazhendi Inban
- Internal Medicine, Government Medical College, Omandurar, Chennai, India
| | | | - Shahnaz Asharaf
- Internal Medicine, Travancore Medical College, Kollam, Kerala, India
| | - Soumya Aleti
- PGY-2, Internal Medicine, Berkshire Medical Center, Pittsfield, MA, USA
| | - Srikanth Gadam
- Internal Medicine, Postdoctoral Research Fellow, Mayo Clinic, USA
| | | | - Dalia Dhia Hadi
- University of Baghdad, Al-Kindy College of Medicine, Baghdad, Iraq
| |
Collapse
|
8
|
Mehan S, Bhalla S, Siddiqui EM, Sharma N, Shandilya A, Khan A. Potential Roles of Glucagon-Like Peptide-1 and Its Analogues in Dementia Targeting Impaired Insulin Secretion and Neurodegeneration. Degener Neurol Neuromuscul Dis 2022; 12:31-59. [PMID: 35300067 PMCID: PMC8921673 DOI: 10.2147/dnnd.s247153] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/16/2022] [Indexed: 12/20/2022] Open
Abstract
Dementia is a chronic, irreversible condition marked by memory loss, cognitive decline, and mental instability. It is clinically related to various progressive neurological diseases, including Parkinson’s disease, Alzheimer’s disease, and Huntington’s. The primary cause of neurological disorders is insulin desensitization, demyelination, oxidative stress, and neuroinflammation accompanied by various aberrant proteins such as amyloid-β deposits, Lewy bodies accumulation, tau formation leading to neurofibrillary tangles. Impaired insulin signaling is directly associated with amyloid-β and α-synuclein deposition, as well as specific signaling cascades involved in neurodegenerative diseases. Insulin dysfunction may initiate various intracellular signaling cascades, including phosphoinositide 3-kinase (PI3K), c-Jun N-terminal kinases (JNK), and mitogen-activated protein kinase (MAPK). Neuronal death, inflammation, neuronal excitation, mitochondrial malfunction, and protein deposition are all influenced by insulin. Recent research has focused on GLP-1 receptor agonists as a potential therapeutic target. They increase glucose-dependent insulin secretion and are beneficial in neurodegenerative diseases by reducing oxidative stress and cytokine production. They reduce the deposition of abnormal proteins by crossing the blood-brain barrier. The purpose of this article is to discuss the role of insulin dysfunction in the pathogenesis of neurological diseases, specifically dementia. Additionally, we reviewed the therapeutic target (GLP-1) and its receptor activators as a possible treatment of dementia.
Collapse
Affiliation(s)
- Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
- Correspondence: Sidharth Mehan, Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India, Tel +91 8059889909; +91 9461322911, Email ;
| | - Sonalika Bhalla
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Ehraz Mehmood Siddiqui
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Nidhi Sharma
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Ambika Shandilya
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Andleeb Khan
- Department of Pharmacology & Toxicology, College of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia
| |
Collapse
|
9
|
Tian D, Gao Q, Chang Z, Lin J, Ma D, Han Z. Network pharmacology and in vitro studies reveal the pharmacological effects and molecular mechanisms of Shenzhi Jiannao prescription against vascular dementia. BMC Complement Med Ther 2022; 22:33. [PMID: 35109845 PMCID: PMC8812053 DOI: 10.1186/s12906-021-03465-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 11/17/2021] [Indexed: 11/12/2022] Open
Abstract
Background Shenzhi Jiannao (SZJN) prescription is a type of herbal formula adopted in the management of cognitive impairment and related disorders. However, its effects and related regulatory mechanisms on vascular dementia (VD) are elusive. Herein, network pharmacology prediction was employed to explore the pharmacological effects and molecular mechanisms of SZJN prescription on VD using network pharmacology prediction, and validated the results through in vitro experiments. Methods Through a search in the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) database, chemical composition and targets for SZJN prescription were retrieved. The potential targets for VD were then obtained from the GeneCards and DisGeNET databases. The network was constructed that depicted the interactions between putative SZJN prescription and known therapeutic targets for VD using Cytoscape 3.7.1. Analysis of protein-protein interaction was achieved via STRING 11.0 software, followed by Gene Ontology (GO) functional enrichment and Kyoto Gene and Genome Encyclopedia (KEGG) pathway analyses. To validate the computer-predicted results, in vitro experiments based on an excitotoxic injury model were designed using glutamate-exposed PC12 cells, and treated with varying concentrations (low, 0.05; medium, 0.1 and high, 0.2 mg/mL) of SZJN prescription. Cell viability and cell death were detected using the IncuCyte imaging system. Moreover, the expression profiles of Caspase-3 were analyzed through qRT-PCR. Results Twenty-eight potentially active ingredients for SZJN prescription, including stigmasterol, beta-sitosterol, and kaempferol, plus 21 therapeutic targets for VD, including PTGS2, PTGS1, and PGR were revealed. The protein-protein interaction network was employed for the analysis of 20 target proteins, including CASP3, JUN, and AChE. The enrichment analysis demonstrated candidate targets of SZJN prescription were more frequently involved in neuroactive ligand-receptor interaction, calcium, apoptosis, and cholinergic synaptic signaling pathways. In vitro experiments revealed that SZJN prescription could significantly reverse glutamate-induced cell viability loss and cell death, and lower the levels of Caspase-3 mRNA in glutamate-induced PC12 cells. Conclusions Collectively, this study demonstrated that SZJN prescription exerted the effect of treating VD by regulating multi-targets and multi-channels with multi-components through the method of network pharmacology. Furthermore, in vitro results confirmed that SZJN prescription attenuated glutamate-induced neurotoxicity.
Collapse
Affiliation(s)
- Danfeng Tian
- Beijing University of Chinese Medicine, No.11 East road, North 3rd Ring Road, Beijing, 100029, China
| | - Qiang Gao
- Beijing University of Chinese Medicine, No.11 East road, North 3rd Ring Road, Beijing, 100029, China
| | - Ze Chang
- Beijing University of Chinese Medicine, No.11 East road, North 3rd Ring Road, Beijing, 100029, China
| | - Jingfeng Lin
- Beijing University of Chinese Medicine, No.11 East road, North 3rd Ring Road, Beijing, 100029, China
| | - Dayong Ma
- Neurology Department of Dongzhimen Hospital, Beijing University of Chinese Medicine, No.5 Haiyuncang, Dongcheng District, Beijing, 100700, China
| | - Zhenyun Han
- Shenzhen Hospital, Beijing University of Chinese Medicine (Longgang), No.1 Dayun road, Sports New City Road, Shenzhen, 518172, China.
| |
Collapse
|
10
|
Connectomic-genetic signatures in the cerebral small vessel disease. Neurobiol Dis 2022; 167:105671. [DOI: 10.1016/j.nbd.2022.105671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/31/2022] [Accepted: 02/21/2022] [Indexed: 11/19/2022] Open
|
11
|
Cognition, mood and behavior in CADASIL. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2022; 3:100043. [PMID: 36324403 PMCID: PMC9616390 DOI: 10.1016/j.cccb.2022.100043] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/06/2022] [Accepted: 02/06/2022] [Indexed: 01/02/2023]
Abstract
CADASIL is responsible for cognitive, mood or behavior disturbances. Cognitive disturbances range from moderate cognitive slowing to impairment of executive functions and may progress to a global decrease of cognitive efficiency up to severe dementia. Mood disturbances are extremely variable in intensity, depression is the most frequent symptom. Behavioral changes may occur at all stage of the disease, but are often associated with the onset of cognitive alterations. Apathy is the most prominent behavior alteration.
CADASIL is the most common familial cerebral small vessel disease (cSVD). Stereotyped mutations of the NOTCH3 gene are responsible for this archetypal ischemic cSVD that can lead, at the very end stage, to severe dementia. Variable cognitive alterations, mood, or behavior disturbances are frequently observed during the course of the disease. In this review, these clinical manifestations, their occurrence, severity and duration are analyzed in relation to the disease progression. Also, the potential relationships with cerebral lesions and treatment options are discussed.
Collapse
|
12
|
Zou D, Zhu X. Acute intracerebral haemorrhage and diffusion-weighted imaging lesions: A meta-analysis. Int J Clin Pract 2021; 75:e14265. [PMID: 33887093 DOI: 10.1111/ijcp.14265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 04/14/2021] [Indexed: 12/01/2022] Open
Abstract
INTRODUCTION Diffusion-weighted imaging lesions in intracerebral haemorrhage are related to a higher risk of recurrent intracerebral haemorrhage, cognitive damage, and mortality. However, it has been reported that the relationship between the risk of diffusion-weighted imaging lesions and intracerebral haemorrhage subtype or the risk factors for diffusion-weighted imaging lesions is variable. This meta-analysis was performed to evaluate this relationship. METHODS A systematic literature search up-to August 2020 was performed and 12 studies included 2815 subjects at the baseline with intracerebral haemorrhage. They were reporting relationships between the diffusion-weighted imaging lesions and intracerebral haemorrhage subtype or investigated the risk factors for diffusion-weighted imaging lesions. Odds ratio (OR) with 95% confidence intervals (CIs) was calculated to evaluate the prognostic role of diffusion-weighted imaging lesions and intracerebral haemorrhage subtype and investigated the risk factors for diffusion-weighted imaging lesions using the dichotomous and continuous method with a random or fixed-effect model. RESULTS Lobar intracerebral haemorrhage was not significantly related to a higher rate of diffusion-weighted imaging lesions (OR, 1.01; 95% CI, 0.75-1.36, P = .94) compared to the non-lobar intracerebral haemorrhage. Also, history of diabetes mellitus (OR, 1.15; 95% CI, 0.83-1.60, P = .39); history of smoking (OR, 0.95; 95% CI, 0.68-1.33, P = .76); history of hypercholesterolaemia (OR, 1.04; 95% CI, 0.73-1.48, P = .83) and history of ischaemic stroke (OR, 1.63; 95% CI, 0.57-4.66, P = .36) were not significantly related to higher rate of diffusion-weighted imaging lesions compared to no history of those factors. However, the history of hypertension was significantly related to a higher rate of diffusion-weighted imaging lesions (OR, 1.33; 95% CI, 1.04-1.70, P = .02) compared to no history of hypertension. Also, Subjects with diffusion-weighted imaging lesions had a greater decrease in systolic pressure in the acute phase of the intracerebral haemorrhage (OR, 10.23; 95% CI, 7.41-13.06, P < .001) compared to without diffusion-weighted imaging lesions. CONCLUSIONS Based on this meta-analysis, the history of hypertension may have an independent risk relationship with a higher rate of diffusion-weighted imaging lesions. Also, subjects with diffusion-weighted imaging lesions had a greater decrease in systolic pressure in the acute phase of the intracerebral haemorrhage compared to those without diffusion-weighted imaging lesions. This relationship forces us to recommend that identification of diffusion-weighted imaging lesions might add appreciated evidence to evaluate the progression of the underlying micro-angiopathy especially in subjects with a history of hypertension. Though further studies are needed to define the mechanisms by which these lesions may lead to cognitive damage and stroke reappearance.
Collapse
Affiliation(s)
- Dianjun Zou
- Department of Medical Imaging, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Xiaolong Zhu
- Department of Medical Imaging, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| |
Collapse
|
13
|
Ulivi L, Cosottini M, Migaleddu G, Orlandi G, Giannini N, Siciliano G, Mancuso M. Brain MRI in Monogenic Cerebral Small Vessel Diseases: A Practical Handbook. Curr Mol Med 2021; 22:300-311. [DOI: 10.2174/1566524021666210510164003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/28/2021] [Accepted: 02/09/2021] [Indexed: 11/22/2022]
Abstract
:
Monogenic cerebral small vessel diseases are a topic of growing interest, as several genes responsible have been recently described and new sequencing techniques such as Next generation sequencing are available. Brain imaging is a key exam in these diseases. First, since it is often the first exam performed, an MRI is key in selecting patients for genetic testing and for interpreting Next generation sequencing reports. In addition, neuroimaging can be helpful in describing the underlying pathological mechanisms involved in cerebral small vessel disease. With this review, we aim to provide Neurologists and Stroke physicians with an up-to date overview of the current neuroimaging knowledge on monogenic small vessel diseases.
Collapse
Affiliation(s)
- Leonardo Ulivi
- Department of Experimental and Clinical Medicine, Neurological Clinic, Pisa University, Via Roma 67, Pisa, Italy
| | - Mirco Cosottini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Gianmichele Migaleddu
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Giovanni Orlandi
- Department of Experimental and Clinical Medicine, Neurological Clinic, Pisa University, Via Roma 67, Pisa, Italy
| | - Nicola Giannini
- Department of Experimental and Clinical Medicine, Neurological Clinic, Pisa University, Via Roma 67, Pisa, Italy
| | - Gabriele Siciliano
- Department of Experimental and Clinical Medicine, Neurological Clinic, Pisa University, Via Roma 67, Pisa, Italy
| | - Michelangelo Mancuso
- Department of Experimental and Clinical Medicine, Neurological Clinic, Pisa University, Via Roma 67, Pisa, Italy
| |
Collapse
|
14
|
The relation between acute intracerebral hemorrhage and diffusion-weighted imaging lesions: a meta-analysis. J Thromb Thrombolysis 2021; 52:962-970. [PMID: 33783661 DOI: 10.1007/s11239-021-02430-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/13/2021] [Indexed: 10/21/2022]
Abstract
Diffusion-weighted imaging lesions in intracerebral hemorrhage are related to a higher risk of recurrent intracerebral hemorrhage, cognitive damage, and mortality. However, it has been reported that the relationship between the risk of diffusion-weighted imaging lesions and intracerebral hemorrhage subtype or the possible risk factors for diffusion-weighted imaging lesions is variable. This meta-analysis was performed to evaluate this relationship. A systematic literature search up-to August 2020 was performed and 12 studies included 2815 subjects at the baseline with intracerebral hemorrhage. Odds ratio (OR) or mean difference (MD) with 95% confidence intervals (CIs) was calculated to evaluate the prognostic role of diffusion-weighted imaging lesions and intracerebral hemorrhage subtype and investigated the possible risk factors for diffusion-weighted imaging lesions using the dichotomous and continuous methods with a random or fixed-effect model. Lobar intracerebral hemorrhage was not significantly related to a higher rate of diffusion-weighted imaging lesions (OR, 1.01; 95% CI, 0.75-1.36, p = 0.94) compared to the non-lobar intracerebral hemorrhage. Also, history of diabetes mellitus (OR, 1.15; 95% CI, 0.83-1.60, p = 0.39); history of smoking (OR, 0.95; 95% CI, 0.68-1.33, p = 0.76); history of hypercholesterolemia (OR, 1.04; 95% CI, 0.73-1.48, p = 0.83); and history of ischemic stroke (OR, 1.63; 95% CI, 0.57-4.66, p = 0.36) were not significantly related to higher rate of diffusion-weighted imaging lesions compared to no history of those factors. However, the history of hypertension was significantly related to a higher rate of diffusion-weighted imaging lesions (OR, 1.33; 95% CI, 1.04-1.70, p = 0.02) compared to no history of hypertension. Also, Subjects with diffusion-weighted imaging lesions had a greater decrease in systolic pressure in the acute phase of the intracerebral hemorrhage (MD, 10.23; 95% CI, 7.41-13.06, p < 0.001) compared to without diffusion-weighted imaging lesions. Based on this meta-analysis, the history of hypertension may have an independent risk relationship with a higher rate of diffusion-weighted imaging lesions. Also, subjects with diffusion-weighted imaging lesions had a greater decrease in systolic pressure in the acute phase of the intracerebral hemorrhage compared to those without diffusion-weighted imaging lesions. This relationship forces us to recommend that identification of diffusion-weighted imaging lesions might add appreciated evidence to evaluate the progression of the underlying micro-angiopathy especially in subjects with a history of hypertension. Though further studies are needed to define the mechanisms by which these lesions may lead to cognitive damage and stroke reappearance.
Collapse
|
15
|
Yamamoto Y, Hase Y, Ihara M, Khundakar A, Roeber S, Duering M, Kalaria RN. Neuronal densities and vascular pathology in the hippocampal formation in CADASIL. Neurobiol Aging 2020; 97:33-40. [PMID: 33130454 PMCID: PMC7758782 DOI: 10.1016/j.neurobiolaging.2020.09.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 09/21/2020] [Accepted: 09/21/2020] [Indexed: 11/17/2022]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common form of hereditary cerebral small vessel disease. Previous neuroimaging studies have suggested loss of hippocampal volume is a pathway for cognitive impairment in CADASIL. We used unbiased stereological methods to estimate SMI32-positive and total numbers and volumes of neurons in the hippocampal formation of 12 patients with CADASIL and similar age controls (young controls) and older controls. We found densities of SMI32-positive neurons in the entorhinal cortex, layer V, and cornu ammonis CA2 regions were reduced by 26%–50% in patients with CADASIL compared with young controls (p < 0.01), with a decreasing trend observed in older controls in the order of young controls> older controls ≥ CADASIL. These changes were not explained by any hippocampal infarct or vascular pathology or glial changes. Our results suggest notable loss of subsets of projection neurons within the hippocampal formation that may contribute to certain memory deficits in CADASIL, which is purely a vascular disease. It is likely that the severe arteriopathy leads to white matter damage which disconnects cortico-cortical and subcortical-cortical networks including the hippocampal formation. Hippocampal volume loss was associated with cognitive dysfunction in CADASIL. SMI32+ neurons in entorhinal cortex (V) and CA2 regions were reduced in CADASIL. Hippocampal cellular changes were not explained by any infarct pathology. Neuronal volumes or glial cell numbers per neuron were not changed. Selective loss of projection neurons within the hippocampal formation in CADASIL.
Collapse
Affiliation(s)
- Yumi Yamamoto
- Translational and Clinical Research Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK; Department of Molecular Innovation in Lipidemiology, Suita, Osaka, Japan; Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Yoshiki Hase
- Translational and Clinical Research Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - Masafumi Ihara
- Department of Molecular Innovation in Lipidemiology, Suita, Osaka, Japan; Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Ahmad Khundakar
- School of Health and Life Sciences, Teesside University, Middlesbrough, Tees Valley, UK
| | | | - Marco Duering
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich & Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Raj N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK.
| |
Collapse
|
16
|
Pan H, Huang Q, Ban S, Du X, Su J, Liu J. Brain structural changes in CADASIL patients: A morphometric magnetic resonance imaging study. Neurosci Lett 2020; 738:135388. [PMID: 32949660 DOI: 10.1016/j.neulet.2020.135388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 09/07/2020] [Accepted: 09/11/2020] [Indexed: 01/23/2023]
Abstract
BACKGROUND Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is a subcortical, inherited, cerebral small vessel disease. Several studies have revealed the involvement of specific cortical regions. However, the structural brain alterations and their clinical correlations remain largely undetermined. METHODS We evaluated 22 CADASIL patients and 22 age- and sex-matched healthy controls. We used surface- and voxel-based morphometric data derived from 3.0-T magnetic resonance imaging (MRI) to explore structural changes in gray and white matter. We calculated Pearson correlations between such data and clinical and MRI metrics. RESULTS Compared with controls, CADASIL patients exhibited significantly decreased cortical thickness in the left supramarginal gyrus, superior temporal gyrus, transverse temporal gyrus, insula, lateral orbitofrontal gyrus, isthmus cingulate gyrus and precentral gyrus. An extensive decrease in the white (but not gray) matter volume was also evident, predominantly in the frontal, parietal, temporal, and occipital lobes. The number of previous strokes or transient ischemic attacks was negatively associated with the cortical thickness of the left pars opercularis and right posterior cingulate gyrus. CONCLUSION Reductions in cortical thickness and white matter volume were evident in CADASIL patients compared with controls, and higher numbers of strokes and transient ischemic attacks were associated with regional cortical thinning.
Collapse
Affiliation(s)
- Hui Pan
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Qi Huang
- PET Center, Huashan Hospital, Fudan University, 518 East Wuzhong Road, Shanghai, 200235, People's Republic of China
| | - Shiyu Ban
- Shanghai Key Laboratory of Magnetic Resonance and Department of Physics, School of Physics and Materials Science, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, People's Republic of China
| | - Xiaoxia Du
- Shanghai Key Laboratory of Magnetic Resonance and Department of Physics, School of Physics and Materials Science, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, People's Republic of China.
| | - Jingjing Su
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| | - Jianren Liu
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
17
|
Chabriat H, Joutel A, Tournier‐Lasserve E, Bousser MG. CADASIL: yesterday, today, tomorrow. Eur J Neurol 2020; 27:1588-1595. [DOI: 10.1111/ene.14293] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 04/28/2020] [Indexed: 12/27/2022]
Affiliation(s)
- H. Chabriat
- Department of Neurology and CERVCO Reference Center for Rare Vascular Diseases of the Eye and Brain Hôpital Lariboisiére, APHP Paris France
- INSERM U 1141 Paris France
- University of Paris Paris France
| | - A. Joutel
- University of Paris Paris France
- Institute of Psychiatry and Neurosciences of Paris INSERM U1266 Paris France
| | - E. Tournier‐Lasserve
- INSERM U 1141 Paris France
- University of Paris Paris France
- Molecular Genetics Department and CERVCO Reference Center for Rare Vascular Diseases of the Eye and Brain Hopital Lariboisiére, APHP Paris France
| | - M. G. Bousser
- Department of Neurology and CERVCO Reference Center for Rare Vascular Diseases of the Eye and Brain Hôpital Lariboisiére, APHP Paris France
- University of Paris Paris France
| |
Collapse
|
18
|
Chabriat H, Jouvent E. Imaging of the aging brain and development of MRI signal abnormalities. Rev Neurol (Paris) 2020; 176:661-669. [PMID: 32229042 DOI: 10.1016/j.neurol.2019.12.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 12/14/2019] [Accepted: 12/16/2019] [Indexed: 02/04/2023]
Abstract
Major changes occur at the cerebral level with aging. Cerebral atrophy develops progressively. Multiple lesions related to small-vessel diseases are detected in association with cerebral atrophy including white-matter hyperintensities, lacunes, microbleeds, dilated perivascular spaces and cerebral, including cortex, atrophy. The clinical impact and predictive value of these Imaging makers were examined.
Collapse
Affiliation(s)
- H Chabriat
- Inserm U1161 and DHU NeuroVasc, department of neurology, Paris University, Lariboisiere Hospital,Assistance Publique-Hopitaux de Paris, Paris, France.
| | - E Jouvent
- Inserm U1161 and DHU NeuroVasc, department of neurology, Paris University, Lariboisiere Hospital,Assistance Publique-Hopitaux de Paris, Paris, France
| |
Collapse
|
19
|
Dunn PJ, Maksemous N, Smith RA, Sutherland HG, Haupt LM, Griffiths LR. Investigating diagnostic sequencing techniques for CADASIL diagnosis. Hum Genomics 2020; 14:2. [PMID: 31915071 PMCID: PMC6950909 DOI: 10.1186/s40246-019-0255-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/30/2019] [Indexed: 11/18/2022] Open
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is a cerebral small vessel disease caused by mutations in the NOTCH3 gene. Our laboratory has been undertaking genetic diagnostic testing for CADASIL since 1997. Work originally utilised Sanger sequencing methods targeting specific NOTCH3 exons. More recently, next-generation sequencing (NGS)-based technologies such as a targeted gene panel and whole exome sequencing (WES) have been used for improved genetic diagnostic testing. In this study, data from 680 patient samples was analysed for 764 tests utilising 3 different sequencing technologies. Sanger sequencing was performed for 407 tests, a targeted NGS gene panel which includes NOTCH3 exonic regions accounted for 354 tests, and WES with targeted analysis was performed for 3 tests. In total, 14.7% of patient samples (n = 100/680) were determined to have a mutation. Testing efficacy varied by method, with 10.8% (n = 44/407) of tests using Sanger sequencing able to identify mutations, with 15.8% (n = 56/354) of tests performed using the NGS custom panel successfully identifying mutations and a likely non-NOTCH3 pathogenic variant (n = 1/3) identified through WES. Further analysis was then performed through stratification of the number of mutations detected at our facility based on the number of exons, level of pathogenicity and the classification of mutations as known or novel. A systematic review of NOTCH3 mutation testing data from 1997 to 2017 determined the diagnostic rate of pathogenic findings and found the NGS-customised panel increases our ability to identify disease-causing mutations in NOTCH3.
Collapse
Affiliation(s)
- P J Dunn
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - N Maksemous
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - R A Smith
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - H G Sutherland
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - L M Haupt
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.
| | - L R Griffiths
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
20
|
Jouvent E, Duering M, Chabriat H. Cerebral Autosomal Dominant Arteriopathy With Subcortical Infarcts and Leukoencephalopathy: Lessons From Neuroimaging. Stroke 2019; 51:21-28. [PMID: 31752612 DOI: 10.1161/strokeaha.119.024152] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Eric Jouvent
- From the Department of Neurology and Referral Center for Rare Vascular Diseases of the Brain and Retina (CERVCO), APHP, Lariboisière Hospital, F-75475 Paris, France (E.J., H.C.).,DHU NeuroVasc, University Paris Diderot (E.J., H.C.).,U1141 INSERM, Paris, France (E.J., H.C.)
| | - Marco Duering
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Germany (M.D.).,Munich Cluster for Systems Neurology (SyNergy), Germany (M.D.)
| | - Hugues Chabriat
- From the Department of Neurology and Referral Center for Rare Vascular Diseases of the Brain and Retina (CERVCO), APHP, Lariboisière Hospital, F-75475 Paris, France (E.J., H.C.).,DHU NeuroVasc, University Paris Diderot (E.J., H.C.).,U1141 INSERM, Paris, France (E.J., H.C.)
| |
Collapse
|
21
|
Kelly DM, Rothwell PM. Blood pressure and the brain: the neurology of hypertension. Pract Neurol 2019; 20:100-108. [DOI: 10.1136/practneurol-2019-002269] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2019] [Indexed: 12/11/2022]
Abstract
Hypertension affects more than one in four adults. The brain is an early target of hypertension-induced organ damage, and may manifest as stroke, subclinical cerebrovascular abnormalities and dementia. Hypertension-related small vessel disease can cause vascular dementia and can potentiate Alzheimer’s pathology, lowering the threshold at which signs and symptoms manifest. Many hypertensive emergencies may also have a neurological presentation, such as hypertensive encephalopathy, haemorrhagic stroke or pre-eclampsia. Here we highlight the importance of blood pressure in maintaining brain health and the brain’s role in controlling blood pressure.
Collapse
|
22
|
Ling Y, De Guio F, Jouvent E, Duering M, Hervé D, Guichard JP, Godin O, Dichgans M, Chabriat H. Clinical correlates of longitudinal MRI changes in CADASIL. J Cereb Blood Flow Metab 2019; 39:1299-1305. [PMID: 29400120 PMCID: PMC6668524 DOI: 10.1177/0271678x18757875] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Previous studies showed that various types of cerebral lesions, as assessed on MRI, largely contribute to the clinical severity of CADASIL. However, the clinical impact of longitudinal changes of classical markers of small vessel disease on conventional MRI has been only poorly investigated. One hundred sixty NOTCH3 mutation carriers (mean age ± SD, 49.8 ± 10.9 years) were followed over three years. Validated methods were used to determine the percent brain volume change (PBVC), number of incident lacunes, change of volume of white matter hyperintensities and change of number of cerebral microbleeds. Multivariable logistic regression analyses were performed to assess the independent association between changes of these MRI markers and incident clinical events. Mixed-effect multiple linear regression analyses were used to assess their association with changes of clinical scales. Over a mean period of 3.1 ± 0.2 years, incident lacunes are found independently associated with incident stroke and change of Trail Making Test Part B. PBVC is independently associated with all incident events and clinical scale changes except the modified Rankin Scale at three years. Our results suggest that, on conventional MRI, PBVC and the number of incident lacunes are the most sensitive and independent correlates of clinical worsening over three years in CADASIL.
Collapse
Affiliation(s)
- Yifeng Ling
- 1 INSERM, U1161 Paris, France.,2 Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - François De Guio
- 1 INSERM, U1161 Paris, France.,3 Department of Neurology, Groupe Hospitalier Saint-Louis-Lariboisière, Assistance Publique des Hôpitaux de Paris (APHP), Université Paris Denis Diderot and DHU NeuroVasc Sorbonne Paris-Cité, Paris, France
| | - Eric Jouvent
- 1 INSERM, U1161 Paris, France.,3 Department of Neurology, Groupe Hospitalier Saint-Louis-Lariboisière, Assistance Publique des Hôpitaux de Paris (APHP), Université Paris Denis Diderot and DHU NeuroVasc Sorbonne Paris-Cité, Paris, France
| | - Marco Duering
- 4 Institute for Stroke and Dementia Research, Klinikum der Universitaüt Muünchen, Ludwig-Maximilians-University, Munich, Germany
| | - Dominique Hervé
- 1 INSERM, U1161 Paris, France.,3 Department of Neurology, Groupe Hospitalier Saint-Louis-Lariboisière, Assistance Publique des Hôpitaux de Paris (APHP), Université Paris Denis Diderot and DHU NeuroVasc Sorbonne Paris-Cité, Paris, France
| | | | - Ophélia Godin
- 3 Department of Neurology, Groupe Hospitalier Saint-Louis-Lariboisière, Assistance Publique des Hôpitaux de Paris (APHP), Université Paris Denis Diderot and DHU NeuroVasc Sorbonne Paris-Cité, Paris, France
| | - Martin Dichgans
- 4 Institute for Stroke and Dementia Research, Klinikum der Universitaüt Muünchen, Ludwig-Maximilians-University, Munich, Germany.,5 Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Hugues Chabriat
- 1 INSERM, U1161 Paris, France.,3 Department of Neurology, Groupe Hospitalier Saint-Louis-Lariboisière, Assistance Publique des Hôpitaux de Paris (APHP), Université Paris Denis Diderot and DHU NeuroVasc Sorbonne Paris-Cité, Paris, France
| |
Collapse
|
23
|
Chen X, Deng S, Xu H, Hou D, Hu P, Yang Y, Wen J, Deng H, Yuan L. Novel and Recurring NOTCH3 Mutations in Two Chinese Patients with CADASIL. NEURODEGENER DIS 2019; 19:35-42. [PMID: 31212292 DOI: 10.1159/000500166] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/05/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is an autosomal-dominant, inherited, systemic, vascular disorder primarily involving the small arteries. It is characterized by migraine, recurrent ischemic strokes, cognitive decline, and dementia. Mutations in the Notch receptor 3 gene (NOTCH3) and the HtrA serine peptidase 1 gene (HTRA1) are 2 genetic causes for CADASIL. The NOTCH3 gene, located on chromosome 19p13.12, is the most common disease-causing gene in CADASIL. OBJECTIVE To investigate genetic causes in 2 unrelated Han-Chinese patients with presentations strongly suggestive of CADASIL. METHODS Exome sequencing was performed on both patients and potential pathogenic mutations were validated by Sanger sequencing. RESULTS This study reports on 2 unrelated Han-Chinese patients with presentations strongly suggestive of CADASIL, identifying that NOTCH3 mutations were the genetic cause. A common mutation, c.268C>T (p.Arg90Cys), and a novel mutation, c.331G>T (p.Gly111Cys) in the NOTCH3 gene, were detected and confirmed in the patients, respectively, and were predicted to be deleterious based on bioinformation analyses. CONCLUSIONS We identified 2 NOTCH3 mutations as likely genetic causes for CADASIL in these 2 patients. Our findings broaden the mutational spectrum of the NOTCH3 gene accountable for CADASIL. Clinical manifestations supplemented with molecular genetic analyses are critical for accurate diagnosis, the provision of genetic counseling, and the development of therapies for CADASIL.
Collapse
Affiliation(s)
- Xiangyu Chen
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Sheng Deng
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, China.,Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Hongbo Xu
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Deren Hou
- Department of Neurology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Pengzhi Hu
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Yan Yang
- Department of Neurology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Jie Wen
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Hao Deng
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, China.,Department of Neurology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Lamei Yuan
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, China,
| |
Collapse
|
24
|
Su J, Wang M, Ban S, Wang L, Cheng X, Hua F, Tang Y, Zhou H, Zhai Y, Du X, Liu J. Relationship between changes in resting-state spontaneous brain activity and cognitive impairment in patients with CADASIL. J Headache Pain 2019; 20:36. [PMID: 30995925 PMCID: PMC6734224 DOI: 10.1186/s10194-019-0982-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/14/2019] [Indexed: 01/15/2023] Open
Abstract
Background Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) mainly manifests with cognitive impairment. Cognitive deficits in patients with CADASIL are correlated with structural brain changes such as lacunar lesion burden, normalized brain volume, and anterior thalamic radiation lesions, but changes in resting-state functional brain activity in patients with CADASIL have not been reported. Methods This study used resting-state functional magnetic resonance imaging (fMRI) to measure the amplitude of low-frequency fluctuation (ALFF) in 22 patients with CADASIL and 44 healthy matched controls. A seed-based functional connectivity (FC) analysis was used to investigate whether the dysfunctional areas identified by ALFF analysis exhibited abnormal FC with other brain areas. Pearson’s correlation analysis was used to detect correlations between the ALFF z-score of abnormal brain areas and clinical scores in patients with CADASIL. Results Patients with CADASIL exhibited significantly lower ALFF values in the right precuneus and cuneus (Pcu/CU) and higher ALFF values in the bilateral superior frontal gyrus (SFG) and left cerebellar anterior and posterior lobes compared with controls. Patients with CADASIL showed weaker FC between the areas with abnormal ALFF (using peaks in the left and right SFG and the right Pcu/CU) and other brain areas. Importantly, the ALFF z-scores for the left and right SFG were negatively associated with cognitive performance, including Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment scores (MoCA), respectively, whereas those of the right Pcu/CU were positively correlated with the MMSE score. Conclusions This preliminary study provides evidence for changes in ALFF of the right Pcu/CU, bilateral SFG and left cerebellar anterior and posterior lobes, and associations between ALFF values for abnormal brain areas and cognitive performance in patients with CADASIL. Therefore, spontaneous brain activity may be a novel imaging biomarker of cognitive impairment in this population.
Collapse
Affiliation(s)
- Jingjing Su
- Department of Neurology and Jiuyuan Municipal Stroke Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Mengxing Wang
- Shanghai Key Laboratory of Magnetic Resonance and Department of Physics, School of Physics and Materials Science, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, People's Republic of China.,College of Medical Imaging, Shanghai University of Medicine & Health Sciences, 279 Zhouzhu Highway, Shanghai, 201318, People's Republic of China
| | - Shiyu Ban
- Shanghai Key Laboratory of Magnetic Resonance and Department of Physics, School of Physics and Materials Science, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, People's Republic of China
| | - Liang Wang
- Department of Neurology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai, 200040, People's Republic of China
| | - Xin Cheng
- Department of Neurology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai, 200040, People's Republic of China
| | - Fengchun Hua
- PET Center, Huashan Hospital, Fudan University, 518 East Wuzhong Road, Shanghai, 200235, People's Republic of China
| | - Yuping Tang
- Department of Neurology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai, 200040, People's Republic of China
| | - Houguang Zhou
- Department of Geriatrics Neurology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai, 200040, People's Republic of China
| | - Yu Zhai
- Department of Neurology and Jiuyuan Municipal Stroke Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| | - Xiaoxia Du
- Shanghai Key Laboratory of Magnetic Resonance and Department of Physics, School of Physics and Materials Science, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, People's Republic of China.
| | - Jianren Liu
- Department of Neurology and Jiuyuan Municipal Stroke Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
25
|
Schoemaker D, Quiroz YT, Torrico-Teave H, Arboleda-Velasquez JF. Clinical and research applications of magnetic resonance imaging in the study of CADASIL. Neurosci Lett 2019; 698:173-179. [PMID: 30634011 PMCID: PMC6661177 DOI: 10.1016/j.neulet.2019.01.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 12/19/2022]
Abstract
Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL) is an inherited small vessel disease that leads to early cerebrovascular events and functional disability. It is the most common single-gene disorder leading to stroke. Magnetic resonance imaging (MRI) is a central component of the diagnosis and monitoring of CADASIL. Here we provide a descriptive review of the literature on three important aspects pertaining to the use of MRI in CADASIL. First, we review past research exploring MRI markers for this disease. Secondly, we describe results from studies investigating associations between neuroimaging abnormalities and neuropathology in CADASIL. Finally, we discuss previous findings relating MRI markers to clinical symptoms. This review thus provides a summary of the current state of knowledge regarding the use of MRI in CADASIL as well as suggestions for future research.
Collapse
Affiliation(s)
- Dorothee Schoemaker
- Schepens Eye Research Institute of Massachusetts Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA, United States; Massachusetts General Hospital and Department of Psychiatry, Harvard Medical School, Boston, MA, United States.
| | - Yakeel T Quiroz
- Massachusetts General Hospital and Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| | - Heirangi Torrico-Teave
- Massachusetts General Hospital and Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| | - Joseph F Arboleda-Velasquez
- Schepens Eye Research Institute of Massachusetts Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
26
|
Boulanger M, Schneckenburger R, Join-Lambert C, Werring DJ, Wilson D, Hodel J, Zuber M, Touzé E. Diffusion-Weighted Imaging Hyperintensities in Subtypes of Acute Intracerebral Hemorrhage: Meta-Analysis. Stroke 2019; 50:135-142. [PMID: 30580720 DOI: 10.1161/strokeaha.118.021407] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose- Diffusion-weighted imaging (DWI) hyperintensities in intracerebral hemorrhage (ICH) are associated with increased risk of recurrent ICH, cognitive impairment, and death, but whether these lesions are specific to a subtype of ICH remains uncertain. We investigated the association between DWI lesions and ICH subtype and explored the risk factors for DWI lesions. Methods- In a systematic review of ICH studies, we identified those reporting prevalence of DWI lesions. Two reviewers independently assessed study eligibility and risk of bias and collected data. We determined the pooled prevalence of DWI lesions within 90 days after ICH onset for cerebral amyloid angiopathy- and hypertensive angiopathy-related ICH using random-effects meta-analysis. We calculated odds ratios to compare prevalence of DWI lesions by ICH subtype and to assess risk factors for DWI lesions. Results- Eleven studies (1910 patients) were included. The pooled prevalence of DWI lesions was 18.9% (95% CI, 11.1-26.7) in cerebral amyloid angiopathy- and 21.0% (95% CI, 15.3-26.6) in hypertensive angiopathy-related ICH. There was no difference in the prevalence of DWI lesions between cerebral amyloid angiopathy- (64/292 [21.9%]) and hypertensive angiopathy-related ICH (79/370 [21.4%]; odds ratio, 1.25; 95% CI, 0.73-2.15) in the 5 studies reporting data on both ICH pathogeneses. In all ICH, presence of DWI lesions was associated with neuroimaging features of microangiopathy (leukoaraiosis extension, previous ICH, and presence, and number of microbleeds) but not with vascular risk factors or the use of antithrombotic therapies. Conclusions- Prevalence of DWI lesions in acute ICH averages 20%, with no difference between cerebral amyloid angiopathy- and hypertensive angiopathy-related ICH. Detection of DWI lesions may add valuable information to assess the progression of the underlying microangiopathy.
Collapse
Affiliation(s)
- Marion Boulanger
- From the Normandie University, UNICAEN, Inserm U1237, Caen, France (M.B., M.Z., E.T.)
- CHU Côte de Nacre, Neurology Department, Caen, France (M.B., R.S., E.T.)
| | | | - Claire Join-Lambert
- Neurology Department, Hôpital Saint Joseph, Université Paris Descartes, France (C.J.-L., M.Z.)
| | - David J Werring
- Department of Brain Repair and Rehabilitation, Stroke Research Centre, Institute of Neurology, Queen Square, London, United Kingdom (D.J.W., D.W.)
| | - Duncan Wilson
- Department of Brain Repair and Rehabilitation, Stroke Research Centre, Institute of Neurology, Queen Square, London, United Kingdom (D.J.W., D.W.)
| | - Jérome Hodel
- Neuroradiology Department, Hôpital Henri Mondor, Université Paris-Est Créteil, France (J.H.)
| | - Mathieu Zuber
- From the Normandie University, UNICAEN, Inserm U1237, Caen, France (M.B., M.Z., E.T.)
- Neurology Department, Hôpital Saint Joseph, Université Paris Descartes, France (C.J.-L., M.Z.)
| | - Emmanuel Touzé
- From the Normandie University, UNICAEN, Inserm U1237, Caen, France (M.B., M.Z., E.T.)
- CHU Côte de Nacre, Neurology Department, Caen, France (M.B., R.S., E.T.)
| |
Collapse
|
27
|
De Guio F, Vignaud A, Chabriat H, Jouvent E. Different types of white matter hyperintensities in CADASIL: Insights from 7-Tesla MRI. J Cereb Blood Flow Metab 2018; 38:1654-1663. [PMID: 28128022 PMCID: PMC6125962 DOI: 10.1177/0271678x17690164] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
In Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL), by contrast to sporadic cerebral small vessel disease related to age and hypertension, white matter hyperintensities (WMH) are frequently observed in the white matter of anterior temporal poles, external capsules, and superior frontal regions. Whether these WMH (specific WMH) differ from those observed in other white matter areas (nonspecific WMH) remains unknown. Twenty patients were scanned to compare specific and nonspecific WMH using high-resolution images and analyses of relaxation times (T1R: longitudinal relaxation time and T2*R: effective transversal relaxation time). Specific WMH were characterized by significantly longer T1R and T2*R (T1R: 2309 ± 120 ms versus 2145 ± 138 ms; T2*R: 40 ± 5 ms versus 35 ± 5 ms, p < 0.001). These results were not explained by the presence of dilated perivascular spaces found in the close vicinity of specific WMH. They were not either explained by the normal regional variability of T1R and T2*R in the white matter nor by systematic imaging artifacts as shown by the study of 17 age- and sex-matched healthy controls. Our results suggest large differences in water content between specific and nonspecific WMH in CADASIL, supporting that mechanisms underlying WMH may differ according to their location.
Collapse
Affiliation(s)
- François De Guio
- 1 University Paris Diderot, Sorbonne Paris Cité, UMR-S 1161 INSERM, Paris, France.,2 DHU NeuroVasc Sorbonne Paris Cité, Paris, France
| | | | - Hugues Chabriat
- 1 University Paris Diderot, Sorbonne Paris Cité, UMR-S 1161 INSERM, Paris, France.,2 DHU NeuroVasc Sorbonne Paris Cité, Paris, France.,4 AP-PH, Lariboisière Hosp., Department of neurology, F-75475, Paris, France
| | - Eric Jouvent
- 1 University Paris Diderot, Sorbonne Paris Cité, UMR-S 1161 INSERM, Paris, France.,2 DHU NeuroVasc Sorbonne Paris Cité, Paris, France.,4 AP-PH, Lariboisière Hosp., Department of neurology, F-75475, Paris, France.,5 UNIACT, NeuroSpin, I2BM/DSV, CEA, Gif-sur-Yvette, France
| |
Collapse
|
28
|
Increased PKR level in human CADASIL brains. Virchows Arch 2018; 473:771-774. [PMID: 30073405 DOI: 10.1007/s00428-018-2425-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 07/22/2018] [Indexed: 10/28/2022]
Abstract
Cerebral autosomal dominant arteriolopathy with subcortical infarcts and leucoencephalopathy (CADASIL) is the most common form of hereditary small vessel disease (SVD) of the brain. Neuronal apoptosis has been demonstrated in the cortex of patients. Whether it is associated with an activation of the pro-apoptotic protein PKR pathway is unknown. Similarly, activation of autophagy in CADASIL has never been explored. Immunostaining of four CADASIL brains previously analyzed for cortical neuronal apoptosis and five control brains for PKR (phosphoPKR) and autophagy (ATG5, LC3II) activation markers. Significant nuclear pPKR staining was observed in CADASIL neurons comparatively to controls (p = 0.001). No difference was observed between patients and controls with autophagy markers. We demonstrated the activation of PKR pathway in CADASIL. This was not associated with a detectable modulation of autophagy. These results open a new field to explore in order to better understand the mechanisms underlying cortical neurons apoptosis.
Collapse
|
29
|
Wilker EH, Martinez-Ramirez S, Kloog I, Schwartz J, Mostofsky E, Koutrakis P, Mittleman MA, Viswanathan A. Fine Particulate Matter, Residential Proximity to Major Roads, and Markers of Small Vessel Disease in a Memory Study Population. J Alzheimers Dis 2018; 53:1315-23. [PMID: 27372639 DOI: 10.3233/jad-151143] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Long-term exposure to ambient air pollution has been associated with impaired cognitive function and vascular disease in older adults, but little is known about these associations among people with concerns about memory loss. OBJECTIVE To examine associations between exposures to fine particulate matter and residential proximity to major roads and markers of small vessel disease. METHODS From 2004-2010, 236 participants in the Massachusetts Alzheimer's Disease Research Center Longitudinal Cohort participated in neuroimaging studies. Residential proximity to major roads and estimated 2003 residential annual average of fine particulate air pollution (PM2.5) were linked to measures of brain parenchymal fraction (BPF), white matter hyperintensities (WMH), and cerebral microbleeds. Associations were modeled using linear and logistic regression and adjusted for clinical and lifestyle factors. RESULTS In this population (median age [interquartile range] = 74 [12], 57% female) living in a region with median 2003 PM2.5 annual average below the current Environmental Protection Agency (EPA) standard, there were no associations between living closer to a major roadway or for a 2μg/m3 increment in PM2.5 and smaller BPF, greater WMH volume, or a higher odds of microbleeds. However, a 2μg/m3 increment in PM2.5 was associated with -0.19 (95% Confidence Interval (CI): -0.37, -0.005) lower natural log-transformed WMH volume. Other associations had wide confidence intervals. CONCLUSIONS In this population, where median 2003 estimated PM2.5 levels were below the current EPA standard, we observed no pattern of association between residential proximity to major roads or 2003 average PM2.5 and greater burden of small vessel disease or neurodegeneration.
Collapse
Affiliation(s)
- Elissa H Wilker
- Cardiovascular Division, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Sergi Martinez-Ramirez
- Hemorrhagic Stroke Research Group, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Itai Kloog
- Department of Geography and Environmental Development, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Joel Schwartz
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Elizabeth Mostofsky
- Cardiovascular Division, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Petros Koutrakis
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Murray A Mittleman
- Cardiovascular Division, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Anand Viswanathan
- Hemorrhagic Stroke Research Group, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
30
|
Tang M, Shi C, Song B, Yang J, Yang T, Mao C, Li Y, Liu X, Zhang S, Wang H, Luo H, Xu Y. CADASIL mutant NOTCH3(R90C) decreases the viability of HS683 oligodendrocytes via apoptosis. Mol Biol Rep 2017; 44:273-280. [PMID: 28601945 DOI: 10.1007/s11033-017-4107-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 05/16/2017] [Indexed: 12/12/2022]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common hereditary cerebral small vessel disease caused by mutations in NOTCH3. Prevailing models suggest that demyelination occurs secondary to vascular pathology. However, in zebrafish, NOTCH3 is also expressed in mature oligodendrocytes. Thus, we hypothesized that in addition to vascular defects, mutant NOTCH3 may alter glial function in individuals with CADASIL. The aim of this study was to characterize the direct effects of a mutant NOTCH3 protein in HS683 oligodendrocytes. HS683 oligodendrocytes transfected with wild-type NOTCH3, mutant NOTCH3(R90C), and empty control vector were used to study the impact of the NOTCH3(R90C) mutant on its protein hydrolytic processing, cell viability, apoptosis, autophagy, oxidative stress, and the related upstream events using immunoblotting, immunofluorescence, RT-PCR, and flow cytometry. We determined that HS683 oligodendrocytes transfected with mutant NOTCH3(R90C), which is the hotspot mutation site-associated with CADASIL, exhibited aberrant NOTCH3 proteolytic processing. Compared to cells overexpressing wild-type NOTCH3, cells overexpressing NOTCH3(R90C) were less viable and had a higher rate of apoptosis. Immunoblotting revealed that cells transfected with NOTCH3(R90C) had higher levels of intrinsic mitochondrial apoptosis, extrinsic death receptor path-related apoptosis, and autophagy compared with cells transfected with wild-type NOTCH3. This study suggests that in patients with CADASIL, early defects in glia influenced by NOTCH3(R90C) may directly contribute to white matter pathology in addition to secondary vascular defects. This study provides a potential therapeutic target for the future treatment of CADASIL.
Collapse
Affiliation(s)
- Mibo Tang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.,Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Bo Song
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Jing Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Ting Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Chengyuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.,Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yusheng Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Xinjing Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Shuyu Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.,Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Hui Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.,Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Haiyang Luo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.,Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
31
|
Lyoubi-Idrissi A, De Guio F, Chabriat H, Jouvent E. Focal Macroscopic Cortical Lesions in Cerebral Autosomal-Dominant Arteriopathy With Subcortical Infarcts and Leukoencephalopathy. Stroke 2017; 48:1408-1411. [DOI: 10.1161/strokeaha.116.015724] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/26/2017] [Accepted: 02/02/2017] [Indexed: 11/16/2022]
Abstract
Background and Purpose—
Cortical microinfarcts and secondary cortical degeneration have been demonstrated in cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), a severe monogenic cerebral small vessel disease. The aim of this study was to determine whether focal macroscopic cortical lesions can be detected using a specific in vivo magnetic resonance imaging approach.
Methods—
Three-dimensional T1 magnetic resonance imaging scans were obtained in 28 nondemented nondisabled CADASIL patients and 29 age- and sex-matched controls. The cortical mantle of patients and controls were extracted using Brainvisa by an experienced user and then evaluated during a dedicated reading session by a second reader after removing the white matter to stay blind to the clinical status. Thereafter, confirmed focal macroscopic cortical lesions were characterized using all available imaging data, including 7-T magnetic resonance imaging in some patients.
Results—
Three focal macroscopic cortical lesions were confirmed in 3 of 28 patients (11%) but none in controls. All lesions were observed in the close vicinity of severe signal changes in the underlying white matter.
Conclusions—
Focal macroscopic cortical lesions can be detected using specific magnetic resonance imaging approaches in CADASIL patients long before the end stage of the disorder. The underlying mechanisms and precise clinical consequences of these cortical changes still need to be determined.
Collapse
Affiliation(s)
- Aicha Lyoubi-Idrissi
- From the Department of Neurology, AP-HP, Lariboisière Hospital, Paris, France (A.L.-I., H.C., E.J.); DHU NeuroVasc Sorbonne Paris Cité, France (A.L.I., F.D.G., H.C., E.J.); UNIACT, NeuroSpin, Gif-sur-Yvette, France (A.L.-I., E.J.); and University Paris Diderot, Sorbonne Paris Cité, UMR-S 1161 INSERM, France (F.D.G., H.C., E.J.)
| | - François De Guio
- From the Department of Neurology, AP-HP, Lariboisière Hospital, Paris, France (A.L.-I., H.C., E.J.); DHU NeuroVasc Sorbonne Paris Cité, France (A.L.I., F.D.G., H.C., E.J.); UNIACT, NeuroSpin, Gif-sur-Yvette, France (A.L.-I., E.J.); and University Paris Diderot, Sorbonne Paris Cité, UMR-S 1161 INSERM, France (F.D.G., H.C., E.J.)
| | - Hugues Chabriat
- From the Department of Neurology, AP-HP, Lariboisière Hospital, Paris, France (A.L.-I., H.C., E.J.); DHU NeuroVasc Sorbonne Paris Cité, France (A.L.I., F.D.G., H.C., E.J.); UNIACT, NeuroSpin, Gif-sur-Yvette, France (A.L.-I., E.J.); and University Paris Diderot, Sorbonne Paris Cité, UMR-S 1161 INSERM, France (F.D.G., H.C., E.J.)
| | - Eric Jouvent
- From the Department of Neurology, AP-HP, Lariboisière Hospital, Paris, France (A.L.-I., H.C., E.J.); DHU NeuroVasc Sorbonne Paris Cité, France (A.L.I., F.D.G., H.C., E.J.); UNIACT, NeuroSpin, Gif-sur-Yvette, France (A.L.-I., E.J.); and University Paris Diderot, Sorbonne Paris Cité, UMR-S 1161 INSERM, France (F.D.G., H.C., E.J.)
| |
Collapse
|
32
|
Pescini F, Donnini I, Cesari F, Nannucci S, Valenti R, Rinnoci V, Poggesi A, Gori AM, Giusti B, Rogolino A, Carluccio A, Bianchi S, Dotti MT, Federico A, Balestrino M, Adriano E, Abbate R, Inzitari D, Pantoni L. Circulating Biomarkers in Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy Patients. J Stroke Cerebrovasc Dis 2016; 26:823-833. [PMID: 27876311 DOI: 10.1016/j.jstrokecerebrovasdis.2016.10.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 07/28/2016] [Accepted: 10/23/2016] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is an inherited cerebral microangiopathy presenting with variable features, including migraine, psychiatric disorders, stroke, and cognitive decline and variable disability. On neuroimaging, CADASIL is characterized by leukoencephalopathy, multiple lacunar infarcts, and microbleeds. Previous studies suggest a possible role of endothelial impairment in the pathogenesis of the disease. METHODS We assessed plasma levels of von Willebrand factor (vWF) and thrombomodulin (TM) and the blood levels of endothelial progenitor cells (EPCs) and circulating progenitor cells (CPCs) in 49 CADASIL patients and 49 age-matched controls and their association with clinical/functional and neuroimaging features. RESULTS In multivariate analysis, CADASIL patients had significantly higher vWF and lower EPC levels. TM levels were similar in the 2 groups. CADASIL patients with a more severe clinical phenotype (history of stroke or dementia) presented lower CPC levels in comparison with patients with a milder phenotype. On correlation analysis, lower CPC levels were associated with worse performances on neuropsychological, motor and functional tests, and with higher lesion load on brain magnetic resonance imaging (degree of leukoencephalopathy and number of lacunar infarcts). CONCLUSIONS This is the first CADASIL series in which multiple circulating biomarkers have been studied. Our findings support previous studies on the presence and the possible modulating effect of endothelial impairment in the disease. Furthermore, our research data suggest that blood CPCs may be markers of disease severity.
Collapse
Affiliation(s)
- Francesca Pescini
- Emergency Department, Stroke Unit, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Ida Donnini
- NEUROFARBA Department, Neuroscience Section, University of Florence, Florence, Italy
| | - Francesca Cesari
- Cardio-Thorax and Vascular Department, Atherothrombotic Diseases Unit, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Serena Nannucci
- NEUROFARBA Department, Neuroscience Section, University of Florence, Florence, Italy; Department of Emergency Neurology, C. Mondino National Neurological Institute, Pavia, Italy
| | - Raffaella Valenti
- NEUROFARBA Department, Neuroscience Section, University of Florence, Florence, Italy
| | - Valentina Rinnoci
- NEUROFARBA Department, Neuroscience Section, University of Florence, Florence, Italy
| | - Anna Poggesi
- NEUROFARBA Department, Neuroscience Section, University of Florence, Florence, Italy
| | - Anna Maria Gori
- Cardio-Thorax and Vascular Department, Atherothrombotic Diseases Unit, Azienda Ospedaliero Universitaria Careggi, Florence, Italy; Don Carlo Gnocchi IRCCS Foundation, Florence, Italy
| | - Betti Giusti
- Cardio-Thorax and Vascular Department, Atherothrombotic Diseases Unit, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Angela Rogolino
- Cardio-Thorax and Vascular Department, Atherothrombotic Diseases Unit, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Alessandra Carluccio
- Department of Medical Surgical and Neurological Sciences, University of Siena, Siena, Italy
| | - Silvia Bianchi
- Department of Medical Surgical and Neurological Sciences, University of Siena, Siena, Italy
| | - Maria Teresa Dotti
- Department of Medical Surgical and Neurological Sciences, University of Siena, Siena, Italy
| | - Antonio Federico
- Department of Medical Surgical and Neurological Sciences, University of Siena, Siena, Italy
| | - Maurizio Balestrino
- Department of Neuroscience, Ophthalmology and Genetics, University of Genoa, Genoa, Italy
| | - Enrico Adriano
- Department of Neuroscience, Ophthalmology and Genetics, University of Genoa, Genoa, Italy
| | - Rosanna Abbate
- Cardio-Thorax and Vascular Department, Atherothrombotic Diseases Unit, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Domenico Inzitari
- NEUROFARBA Department, Neuroscience Section, University of Florence, Florence, Italy; Institute of Neuroscience, Italian National Research Council, Florence, Italy
| | - Leonardo Pantoni
- NEUROFARBA Department, Neuroscience Section, University of Florence, Florence, Italy.
| |
Collapse
|
33
|
Cai Z, Zhao B, Deng Y, Shangguan S, Zhou F, Zhou W, Li X, Li Y, Chen G. Notch signaling in cerebrovascular diseases (Review). Mol Med Rep 2016; 14:2883-98. [PMID: 27574001 PMCID: PMC5042775 DOI: 10.3892/mmr.2016.5641] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 07/22/2016] [Indexed: 12/30/2022] Open
Abstract
The Notch signaling pathway is a crucial regulator of numerous fundamental cellular processes. Increasing evidence suggests that Notch signaling is involved in inflammation and oxidative stress, and thus in the progress of cerebrovascular diseases. In addition, Notch signaling in cerebrovascular diseases is associated with apoptosis, angiogenesis and the function of blood-brain barrier. Despite the contradictory results obtained to date as to whether Notch signaling is harmful or beneficial, the regulation of Notch signaling may provide a novel strategy for the treatment of cerebrovascular diseases.
Collapse
Affiliation(s)
- Zhiyou Cai
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Bin Zhao
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yanqing Deng
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Shouqin Shangguan
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Faming Zhou
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Wenqing Zhou
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Xiaoli Li
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yanfeng Li
- Department of Neurology, Peking Union Medical College Hospital, Beijing 100730, P.R. China
| | - Guanghui Chen
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| |
Collapse
|
34
|
Abou Al-Shaar H, Qadi N, Al-Hamed MH, Meyer BF, Bohlega S. Phenotypic comparison of individuals with homozygous or heterozygous mutation of NOTCH3 in a large CADASIL family. J Neurol Sci 2016; 367:239-43. [DOI: 10.1016/j.jns.2016.05.061] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 05/10/2016] [Accepted: 05/31/2016] [Indexed: 12/19/2022]
|
35
|
Peres R, De Guio F, Chabriat H, Jouvent E. Alterations of the cerebral cortex in sporadic small vessel disease: A systematic review of in vivo MRI data. J Cereb Blood Flow Metab 2016; 36:681-95. [PMID: 26787108 PMCID: PMC4821027 DOI: 10.1177/0271678x15625352] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 11/30/2015] [Accepted: 12/10/2015] [Indexed: 11/16/2022]
Abstract
Cerebral small vessel diseases of the brain are a major determinant of cognitive impairment in the elderly. In small vessel diseases, the most easily identifiable lesions, both at post-mortem evaluation and magnetic resonance imaging, lie in subcortical areas. However, recent results obtained post-mortem, particularly in severe cases, have highlighted the burden of cortex lesions such as microinfarcts and diffuse neuronal loss. The recent development of image post-processing methods allows now assessing in vivo multiple aspects of the cerebral cortex. This systematic review aimed to analyze in vivo magnetic resonance imaging studies evaluating cortex alterations at different stages of small vessel diseases. Studies assessing the relationships between small vessel disease magnetic resonance imaging markers obtained at the subcortical level and cortex estimates were reviewed both in community-dwelling elderly and in patients with symptomatic small vessel diseases. Thereafter, studies analyzing cortex estimates in small vessel disease patients compared with healthy subjects were evaluated. The results support that important cortex alterations develop along the course of small vessel diseases independently of concomitant neurodegenerative processes. Easy detection and quantification of cortex changes in small vessel diseases as well as understanding their underlying mechanisms are challenging tasks for better understanding cognitive decline in small vessel diseases.
Collapse
Affiliation(s)
- Roxane Peres
- Univ Paris Diderot, Sorbonne Paris Cité, UMR-S 1161 INSERM, Paris, France DHU NeuroVasc Sorbonne Paris Cité, Paris, France
| | - François De Guio
- Univ Paris Diderot, Sorbonne Paris Cité, UMR-S 1161 INSERM, Paris, France DHU NeuroVasc Sorbonne Paris Cité, Paris, France
| | - Hugues Chabriat
- Univ Paris Diderot, Sorbonne Paris Cité, UMR-S 1161 INSERM, Paris, France DHU NeuroVasc Sorbonne Paris Cité, Paris, France Department of Neurology, AP-HP, Lariboisière Hosp, F-75475 Paris, France
| | - Eric Jouvent
- Univ Paris Diderot, Sorbonne Paris Cité, UMR-S 1161 INSERM, Paris, France DHU NeuroVasc Sorbonne Paris Cité, Paris, France Department of Neurology, AP-HP, Lariboisière Hosp, F-75475 Paris, France
| |
Collapse
|
36
|
Cho EB, Shin HY, Park SE, Chun P, Jang HR, Yang JJ, Kim HJ, Kim YJ, Jung NY, Lee JS, Lee J, Jang YK, Jang EY, Kang M, Lee JM, Kim C, Min JH, Ryu S, Na DL, Seo SW. Albuminuria, Cerebrovascular Disease and Cortical Atrophy: among Cognitively Normal Elderly Individuals. Sci Rep 2016; 6:20692. [PMID: 26878913 PMCID: PMC4754729 DOI: 10.1038/srep20692] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 01/11/2016] [Indexed: 01/19/2023] Open
Abstract
We tested the hypothesis that decreased glomerular filtration rate and albuminuria have different roles in brain structure alterations. We enrolled 1,215 cognitively normal individuals, all of whom underwent high-resolution T1-weighted volumetric magnetic resonance imaging scans. The cerebral small vessel disease burdens were assessed with white matter hyperintensities (WMH), lacunes, and microbleeds. Subjects were considered to have an abnormally elevated urine albumin creatinine ratio if the value was ≥17 mg/g for men and ≥25 mg/g for women. Albuminuria, but not estimated glomerular filtration rate (eGFR), was associated with increased WMH burdens (p = 0.002). The data was analyzed after adjusting for age, sex, education, history of hypertension, diabetes mellitus, hyperlipidemia, ischemic heart disease, stroke, total cholesterol level, body mass index, status of smoking and alcohol drinking, and intracranial volume. Albuminuria was also associated with cortical thinning, predominantly in the frontal and occipital regions (both p < 0.01) in multiple linear regression analysis. However, eGFR was not associated with cortical thickness. Furthermore, path analysis for cortical thickness showed that albuminuria was associated with frontal thinning partially mediated by WMH burdens. The assessment of albuminuria is needed to improve our ability to identify individuals with high risk for cognitive impairments, and further institute appropriate preventive measures.
Collapse
Affiliation(s)
- Eun Bin Cho
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Neuroscience Center, Samsung Medical Center, Seoul, Korea
- Department of Neurology, Gyeongsang National University Changwon Hospital, Gyeongsang National University School of Medicine, Changwon, Korea
| | - Hee-Young Shin
- Center for Health Promotion, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sang Eon Park
- Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Korea
| | - Phillip Chun
- Department of Emergency Medicine Behavioral Emergencies Research Lab, San Diego, CA, USA
- Department of Biology, University of California San Diego, CA, USA
| | - Hye Ryoun Jang
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jin-ju Yang
- Department of Biomedical Engineering, Hanyang University, Seoul, Korea
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Yeo Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Na-Yeon Jung
- Department of Neurology, Pusan National University Hospital, Pusan National University College of Medicine and Biomedical Research Institute, Busan, Korea
| | - Jin San Lee
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Juyoun Lee
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Young Kyoung Jang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Eun Young Jang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Mira Kang
- Center for Health Promotion, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jong-Min Lee
- Department of Biomedical Engineering, Hanyang University, Seoul, Korea
| | - Changsoo Kim
- Department of Preventive Medicine and the Institute for Environmental Research, Yonsei University College of Medicine, Seoul, Korea
- Divison of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Ju-Hong Min
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Seungho Ryu
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Duk L. Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Neuroscience Center, Samsung Medical Center, Seoul, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
- Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
37
|
Hughes TM, Sink KM. Hypertension and Its Role in Cognitive Function: Current Evidence and Challenges for the Future. Am J Hypertens 2016; 29:149-57. [PMID: 26563965 DOI: 10.1093/ajh/hpv180] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/06/2015] [Indexed: 12/17/2022] Open
Abstract
This review summarizes evidence from studies of blood pressure and dementia-related biomarkers into our understanding of cognitive health and highlights the challenges facing studies, particularly randomized trials, of hypertension and cognition. Several lines of research suggest that elevated blood pressure, especially at midlife, is associated with cognitive decline and dementia and that treatment of hypertension could prevent these conditions. Further, studies of hypertension and brain structure show that blood pressure is associated with several forms of small vessel disease that can result in vascular dementia or interact with Alzheimer's pathology to lower the pathologic threshold at which Alzheimer's signs and symptoms manifest. In addition, recent studies of hypertension and Alzheimer's biomarkers show that elevated blood pressure and pulse pressure are associated with the extent of brain beta amyloid (Aβ) deposition and altered cerebral spinal fluid profiles of Aβ and tau indicative of Alzheimer's pathology. However, in spite of strong evidence of biological mechanisms, results from randomized trials of antihypertensive therapy for the prevention of cardiovascular or cerebrovascular disease that include cognitive endpoints do not strongly support the observational evidence that treatment of hypertension should be better for cognition. We propose that future clinical trials should consider including dementia biomarkers and assess genetic and cardiometabolic risk factors that have been associated with progression of the underlying disease pathology to help bridge these gaps.
Collapse
Affiliation(s)
- Timothy M Hughes
- Department of Internal Medicine, Division of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Kaycee M Sink
- Department of Internal Medicine, Division of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.
| |
Collapse
|
38
|
Hughes TM, Craft S, Lopez OL. Review of 'the potential role of arterial stiffness in the pathogenesis of Alzheimer's disease'. Neurodegener Dis Manag 2016; 5:121-35. [PMID: 25894876 DOI: 10.2217/nmt.14.53] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Arterial stiffness is emerging as an important risk marker for poor brain aging and dementia through its associations with cerebral small vessel disease, stroke, β-amyloid deposition, brain atrophy and cognitive impairment. Arterial stiffness directly relates the detrimental effects of hypertension on peripheral organs with dire consequences for the extensive microvasculature structure of the kidneys and brain. In this review, we discuss the evidence linking arterial stiffness, hypertension and brain structural abnormalities in older adults. In particular, we discuss the potential mechanisms linking arterial stiffness to brain β-amyloid deposition and dementia and potential therapeutic strategies to prevent hypertension's adverse effects on the brain.
Collapse
Affiliation(s)
- Timothy M Hughes
- Department of Internal Medicine, Division of Gerontology & Geriatric Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | | | | |
Collapse
|
39
|
Hughes TM, Craft S. The role of insulin in the vascular contributions to age-related dementia. Biochim Biophys Acta Mol Basis Dis 2015; 1862:983-91. [PMID: 26657615 DOI: 10.1016/j.bbadis.2015.11.013] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/17/2015] [Accepted: 11/29/2015] [Indexed: 01/12/2023]
Abstract
In addition to its well-known role in energy metabolism in the body, insulin is a vasoactive hormone that regulates peripheral and cerebral blood flow and neuronal function. Vascular and metabolic dysfunctions are emerging risk factors for Alzheimer's disease (AD) and age-related dementias, and recent evidence suggests that the two pathways are constitutive and interrelated. As a result, an emphasis on correcting metabolic disorders is emerging as an important strategy in the treatment and prevention of age-related cognitive impairment and AD. We review the evidence regarding the unique and interactive effects of vascular and metabolic disorders in pathological brain aging, with special consideration of the role of insulin dysregulation in promoting AD pathologic processes and vascular brain injury. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
Affiliation(s)
- Timothy M Hughes
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Wake Forest University, USA
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Wake Forest University, USA.
| |
Collapse
|
40
|
Ghosh M, Balbi M, Hellal F, Dichgans M, Lindauer U, Plesnila N. Pericytes are involved in the pathogenesis of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. Ann Neurol 2015; 78:887-900. [PMID: 26312599 DOI: 10.1002/ana.24512] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 08/24/2015] [Accepted: 08/24/2015] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), the most common inherited small-vessel disease, is associated with vascular aggregation of mutant Notch3 protein, dysfunction of cerebral vessels, and dementia. Pericytes, perivascular cells involved in microvascular function, express Notch3. Therefore, we hypothesize that these cells may play a role in the pathogenesis of CADASIL. METHODS Two-, 7-, and 12-month-old CADASIL mutant mice (TgNotch3(R169C) ) and wild-type controls were examined regarding Notch3 aggregation in pericytes, the coverage of cerebral vessels by pericytes, pericyte numbers, capillary density, blood-brain barrier (BBB) integrity, astrocytic end-feet, and the expression of astrocytic gap junction and endothelial adherens junction protein using immunostaining and Western blot analysis. In addition, we examined cerebrovascular CO2 reactivity using laser Doppler fluxmetry and in vivo microscopy. RESULTS With increasing age, mutated Notch3 aggregated around pericytes and smooth muscle cells. Notch3 aggregation caused significant reduction of pericyte number and coverage of capillaries by pericyte processes (p < 0.01). These changes were associated with detachment of astrocytic end-feet from cerebral microvessels, leakage of plasma proteins, reduction in expression of endothelial adherens junction protein, and reduced microvascular reactivity to CO2 . Smooth muscle cells were not affected by Notch3 accumulation. INTERPRETATION Our results show that pericytes are the first cells affected by Notch3 aggregation in CADASIL mice. Pericyte pathology causes opening of the BBB and microvascular dysfunction. Therefore, protecting pericytes may represent a novel therapeutic strategy for vascular dementia.
Collapse
Affiliation(s)
- Mitrajit Ghosh
- Institute for Stroke and Dementia Research, University of Munich Medical Center, Ludwig Maximilian University, Munich.,Experimental Neurosurgery, Department of Neurosurgery, Rechts der Isar Hospital, Technical University Munich, Munich.,Munich Cluster for Systems Neurology (SyNergy), Munich
| | - Matilde Balbi
- Institute for Stroke and Dementia Research, University of Munich Medical Center, Ludwig Maximilian University, Munich.,Graduate School of Systemic Neurosciences, Ludwig Maximilian University, Munich
| | - Farida Hellal
- Institute for Stroke and Dementia Research, University of Munich Medical Center, Ludwig Maximilian University, Munich.,Munich Cluster for Systems Neurology (SyNergy), Munich
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University of Munich Medical Center, Ludwig Maximilian University, Munich.,Munich Cluster for Systems Neurology (SyNergy), Munich
| | - Ute Lindauer
- Experimental Neurosurgery, Department of Neurosurgery, Rechts der Isar Hospital, Technical University Munich, Munich.,Munich Cluster for Systems Neurology (SyNergy), Munich.,Translational Neurosurgery and Neurobiology, Department of Neurosurgery, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research, University of Munich Medical Center, Ludwig Maximilian University, Munich.,Munich Cluster for Systems Neurology (SyNergy), Munich
| |
Collapse
|
41
|
Flanagan M, Larson EB, Latimer CS, Cholerton B, Crane PK, Montine KS, White LR, Keene CD, Montine TJ. Clinical-pathologic correlations in vascular cognitive impairment and dementia. Biochim Biophys Acta Mol Basis Dis 2015; 1862:945-51. [PMID: 26319420 DOI: 10.1016/j.bbadis.2015.08.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 08/22/2015] [Accepted: 08/24/2015] [Indexed: 11/30/2022]
Abstract
The most common causes of cognitive impairment and dementia are Alzheimer's disease (AD) and vascular brain injury (VBI), either independently, in combination, or in conjunction with other neurodegenerative disorders. The contribution of VBI to cognitive impairment and dementia, particularly in the context of AD pathology, has been examined extensively yet remains difficult to characterize due to conflicting results. Describing the relative contribution and mechanisms of VBI in dementia is important because of the profound impact of dementia on individuals, caregivers, families, and society, particularly the stability of health care systems with the rapidly increasing age of our population. Here we discuss relationships between pathologic processes of VBI and clinical expression of dementia, specific subtypes of VBI including microvascular brain injury, and what is currently known regarding contributions of VBI to the development and pathogenesis of the dementia syndrome. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
Affiliation(s)
- Margaret Flanagan
- Department of Pathology, University of Washington, Seattle, WA, United States
| | - Eric B Larson
- Group Health Research Institute, Seattle, WA, United States
| | - Caitlin S Latimer
- Department of Pathology, University of Washington, Seattle, WA, United States
| | - Brenna Cholerton
- Department of Psychiatry & Behavioral Sciences, University of Washington, Seattle, WA, United States
| | - Paul K Crane
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Kathleen S Montine
- Department of Pathology, University of Washington, Seattle, WA, United States
| | - Lon R White
- Pacific Health Research and Education Institute, Honolulu, HI, United States; Department of Geriatric Medicine, University of Hawaii John A. Burns School of Medicine, Honolulu, HI, United States
| | - C Dirk Keene
- Department of Pathology, University of Washington, Seattle, WA, United States
| | - Thomas J Montine
- Department of Pathology, University of Washington, Seattle, WA, United States.
| |
Collapse
|
42
|
Weller RO, Hawkes CA, Kalaria RN, Werring DJ, Carare RO. White matter changes in dementia: role of impaired drainage of interstitial fluid. Brain Pathol 2015; 25:63-78. [PMID: 25521178 DOI: 10.1111/bpa.12218] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 10/08/2014] [Indexed: 12/22/2022] Open
Abstract
White matter abnormalities on magnetic resonance imaging (MRI) are associated with dementia and include white matter hyperintensities (WMH; also termed leukoaraiosis) and visible perivascular spaces (PVS). We review the potential role of impaired drainage of interstitial fluid in the pathogenesis of WMH and PVS. Whereas the volume of extracellular space in the grey matter is tightly controlled, fluid accumulates and expands the extracellular spaces of the white matter in acute hydrocephalus, vasogenic edema and WMH. Although there are no conventional lymphatic vessels in the brain, there is very effective lymphatic drainage for fluid and solutes along restricted pathways in the basement membranes of cerebral capillaries and arteries in young individuals. Lymphatic drainage of the brain is impaired with age and in association with apolipoprotein E ε4, risk factors for Alzheimer's disease and cerebral amyloid angiopathy (CAA). Deposition of proteins in the lymphatic drainage pathways in the walls of cerebral arteries with age is recognized as protein elimination failure angiopathy (PEFA), as in CAA and cerebral autosomal dominant arteriopathy and leukoencephalopathy (CADASIL). Facilitating perivascular lymphatic drainage from the aging brain may play a significant role in the prevention of CAA, WMH and Alzheimer's disease and may enhance the efficacy of immunotherapy for Alzheimer's disease.
Collapse
Affiliation(s)
- Roy O Weller
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | | | | | | |
Collapse
|
43
|
Liu XY, Gonzalez-Toledo ME, Fagan A, Duan WM, Liu Y, Zhang S, Li B, Piao CS, Nelson L, Zhao LR. Stem cell factor and granulocyte colony-stimulating factor exhibit therapeutic effects in a mouse model of CADASIL. Neurobiol Dis 2015; 73:189-203. [DOI: 10.1016/j.nbd.2014.09.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 08/27/2014] [Accepted: 09/12/2014] [Indexed: 11/28/2022] Open
|
44
|
Alten F, Motte J, Ewering C, Osada N, Clemens CR, Kadas EM, Eter N, Paul F, Marziniak M. Multimodal retinal vessel analysis in CADASIL patients. PLoS One 2014; 9:e112311. [PMID: 25372785 PMCID: PMC4221286 DOI: 10.1371/journal.pone.0112311] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Accepted: 10/06/2014] [Indexed: 11/18/2022] Open
Abstract
PURPOSE To further elucidate retinal findings and retinal vessel changes in Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) patients by means of high resolution retinal imaging. METHODS 28 eyes of fourteen CADASIL patients and an equal number of control subjects underwent confocal scanning laser ophthalmoscopy (cSLO), spectral-domain optical coherence tomography (SD-OCT), retinal nerve fibre layer (RNFL) measurements, fluorescein and indocyanine angiography. Three vessel measurement techniques were applied: RNFL thickness, a semiautomatic software tool based on cSLO images and manual vessel outlining based on SD-OCT. RESULTS Mean age of patients was 56.2 ± 11.6 years. Arteriovenous nicking was present in 22 (78.6%) eyes and venous dilation in 24 (85.7%) eyes. Retinal volume and choroidal volume were 8.77 ± 0.46 mm(3) and 8.83 ± 2.24 mm(3). RNFL measurements showed a global increase of 105.2 µm ( CONTROL GROUP 98.4 µm; p = 0.015). Based on semi-automatic cSLO measurements, maximum diameters of arteries and veins were 102.5 µm (106.0 µm; p = 0.21) and 128.6 µm (124.4 µm; p = 0.27) respectively. Manual SD-OCT measurements revealed significantly increased mean arterial 138.7 µm (125.4 µm; p<0.001) and venous 160.0 µm (146.9; p = 0.003) outer diameters as well as mean arterial 27.4 µm (19.2 µm; p<0.001) and venous 18.3 µm (15.7 µm; p<0.001) wall thicknesses in CADASIL patients. CONCLUSIONS The findings reflect current knowledge on pathophysiologic changes in vessel morphology in CADASIL patients. SD-OCT may serve as a complementary tool to diagnose and follow-up patients suffering from cerebral small-vessel diseases.
Collapse
Affiliation(s)
- Florian Alten
- Department of Ophthalmology, University of Muenster Medical Center, Muenster, Germany
- * E-mail:
| | - Jeremias Motte
- Department of Neurology, University of Muenster Medical Center, Muenster, Germany
| | - Carina Ewering
- Department of Neurology, University of Muenster Medical Center, Muenster, Germany
| | - Nani Osada
- Department of Medical Informatics and Biomathematics, University of Muenster, Muenster, Germany
| | - Christoph R. Clemens
- Department of Ophthalmology, University of Muenster Medical Center, Muenster, Germany
| | - Ella M. Kadas
- Department of Neurology, Charite University Medicine Berlin, Berlin, German
| | - Nicole Eter
- Department of Ophthalmology, University of Muenster Medical Center, Muenster, Germany
| | - Friedemann Paul
- Department of Neurology, Charite University Medicine Berlin, Berlin, German
- NeuroCure Clinical Research Center, Berlin, Germany
| | - Martin Marziniak
- Department of Neurology, University of Muenster Medical Center, Muenster, Germany
| |
Collapse
|
45
|
Craggs LJ, Yamamoto Y, Deramecourt V, Kalaria RN. Microvascular pathology and morphometrics of sporadic and hereditary small vessel diseases of the brain. Brain Pathol 2014; 24:495-509. [PMID: 25323665 PMCID: PMC4228759 DOI: 10.1111/bpa.12177] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 06/27/2014] [Indexed: 12/12/2022] Open
Abstract
Small vessel diseases (SVDs) of the brain are likely to become increasingly common in tandem with the rise in the aging population. In recent years, neuroimaging and pathological studies have informed on the pathogenesis of sporadic SVD and several single gene (monogenic) disorders predisposing to subcortical strokes and diffuse white matter disease. However, one of the limitations toward studying SVD lies in the lack of consistent assessment criteria and lesion burden for both clinical and pathological measures. Arteriolosclerosis and diffuse white matter changes are the hallmark features of both sporadic and hereditary SVDs. The pathogenesis of the arteriopathy is the key to understanding the differential progression of disease in various SVDs. Remarkably, quantification of microvascular abnormalities in sporadic and hereditary SVDs has shown that qualitatively the processes involved in arteriolar degeneration are largely similar in sporadic SVD compared with hereditary disorders such as cerebral autosomal arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). Important significant regional differences in lesion location within the brain may enable one to distinguish SVDs, where frontal lobe involvement appears consistently with almost every SVD, but others bear specific pathologies in other lobes, such as the temporal pole in CADASIL and the pons in pontine autosomal dominant microangiopathy and leukoencephalopathy or PADMAL. Additionally, degenerative changes in the vascular smooth muscle cells, the cerebral endothelium and the basal lamina are often rapid and more aggressive in genetic disorders. Further quantification of other microvascular elements and even neuronal cells is needed to fully characterize SVD pathogenesis and to differentiate the usefulness of vascular interventions and treatments on the resulting pathology.
Collapse
Affiliation(s)
| | - Yumi Yamamoto
- Department of Regenerative Medicine and Tissue EngineeringNational Cerebral and Cardiovascular CenterNational Cerebral and Cardiovascular Center Research InstituteOsakaJapan
| | | | - Raj N. Kalaria
- Institute for Ageing and HealthNewcastle UniversityNewcastle upon TyneUK
| |
Collapse
|
46
|
De Guio F, Reyes S, Vignaud A, Duering M, Ropele S, Duchesnay E, Chabriat H, Jouvent E. In vivo high-resolution 7 Tesla MRI shows early and diffuse cortical alterations in CADASIL. PLoS One 2014; 9:e106311. [PMID: 25165824 PMCID: PMC4148432 DOI: 10.1371/journal.pone.0106311] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 07/24/2014] [Indexed: 11/19/2022] Open
Abstract
Background and Purpose Recent data suggest that early symptoms may be related to cortex alterations in CADASIL (Cerebral Autosomal-Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy), a monogenic model of cerebral small vessel disease (SVD). The aim of this study was to investigate cortical alterations using both high-resolution T2* acquisitions obtained with 7 Tesla MRI and structural T1 images with 3 Tesla MRI in CADASIL patients with no or only mild symptomatology (modified Rankin’s scale ≤1 and Mini Mental State Examination (MMSE) ≥24). Methods Complete reconstructions of the cortex using 7 Tesla T2* acquisitions with 0.7 mm isotropic resolution were obtained in 11 patients (52.1±13.2 years, 36% male) and 24 controls (54.8±11.0 years, 42% male). Seven Tesla T2* within the cortex and cortical thickness and morphology obtained from 3 Tesla images were compared between CADASIL and control subjects using general linear models. Results MMSE, brain volume, cortical thickness and global sulcal morphology did not differ between groups. By contrast, T2* measured by 7 Tesla MRI was significantly increased in frontal, parietal, occipital and cingulate cortices in patients after correction for multiple testing. These changes were not related to white matter lesions, lacunes or microhemorrhages in patients having no brain atrophy compared to controls. Conclusions Seven Tesla MRI, by contrast to state of the art post-processing of 3 Tesla acquisitions, shows diffuse T2* alterations within the cortical mantle in CADASIL whose origin remains to be determined.
Collapse
Affiliation(s)
- François De Guio
- Univ Paris Diderot, Sorbonne Paris Cité, UMR-S 1161 INSERM, Paris, France
- DHU NeuroVasc Sorbonne Paris Cité, Paris, France
| | - Sonia Reyes
- AP-HP, Lariboisière Hosp, Department of Neurology, Paris, France
| | | | - Marco Duering
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany
| | - Stefan Ropele
- Department of Neurology, Medical University of Graz, Graz, Austria
| | | | - Hugues Chabriat
- Univ Paris Diderot, Sorbonne Paris Cité, UMR-S 1161 INSERM, Paris, France
- DHU NeuroVasc Sorbonne Paris Cité, Paris, France
- AP-HP, Lariboisière Hosp, Department of Neurology, Paris, France
| | - Eric Jouvent
- Univ Paris Diderot, Sorbonne Paris Cité, UMR-S 1161 INSERM, Paris, France
- DHU NeuroVasc Sorbonne Paris Cité, Paris, France
- AP-HP, Lariboisière Hosp, Department of Neurology, Paris, France
- * E-mail:
| |
Collapse
|
47
|
Cognat E, Cleophax S, Domenga-Denier V, Joutel A. Early white matter changes in CADASIL: evidence of segmental intramyelinic oedema in a pre-clinical mouse model. Acta Neuropathol Commun 2014; 2:49. [PMID: 24886907 PMCID: PMC4035092 DOI: 10.1186/2051-5960-2-49] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 04/22/2014] [Indexed: 01/28/2023] Open
Abstract
Introduction Small vessel disease (SVD) of the brain is a leading cause of age- and hypertension-related cognitive decline and disability. Cerebral white matter changes are a consistent manifestation of SVD on neuroimaging, progressing silently for many years before becoming clinically evident. The pathogenesis of these changes remains poorly understood, despite their importance. In particular, their pathological correlate at early stages remains largely undefined. Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL), caused by dominant mutations of the NOTCH3 receptor, is regarded as a paradigm for the most common form of sporadic SVD. In this study, we used immunohistochemistry, confocal microscopy and electron microscopy, together with qualitative and quantitative analyses to assess oligodendroglial, axon and myelin damage in TgPAC-Notch3R169C mice, a model of preclinical CADASIL. Results The principal cerebral white matter changes in TgPAC-Notch3R169C mice are microvacuoles (≤1 μm diameter) in the myelin sheaths associated with focal myelin degradation and occurring in the absence of oligodendrocyte loss. Half the damaged myelin sheaths still contain an apparently intact axon. Clearance of myelin debris appears inefficient, as demonstrated by the significant but mild microglial reaction, with occasional myelin debris either contacted or internalized by microglial cells. Conclusion Our findings suggest that segmental intramyelinic oedema is an early, conspicuous white matter change in CADASIL. Brain white matter intramyelinic oedema is consistently found in patients and mouse models with compromised ion and water homeostasis. These data provide a starting point for novel mechanistic studies to investigate the pathogenesis of SVD-related white matter changes. Electronic supplementary material The online version of this article (doi:10.1186/2051-5960-2-49) contains supplementary material, which is available to authorized users.
Collapse
|
48
|
Fakhran S, Yaeger K, Alhilali L. Symptomatic White Matter Changes in Mild Traumatic Brain Injury Resemble Pathologic Features of Early Alzheimer Dementia. Radiology 2013; 269:249-57. [DOI: 10.1148/radiol.13122343] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
49
|
Carare RO, Hawkes CA, Jeffrey M, Kalaria RN, Weller RO. Review: Cerebral amyloid angiopathy, prion angiopathy, CADASIL and the spectrum of protein elimination failure angiopathies (PEFA) in neurodegenerative disease with a focus on therapy. Neuropathol Appl Neurobiol 2013; 39:593-611. [DOI: 10.1111/nan.12042] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 03/07/2013] [Indexed: 01/03/2023]
Affiliation(s)
- R. O. Carare
- Clinical Neurosciences and Experimental Sciences; Faculty of Medicine; University of Southampton; Southampton; UK
| | - C. A. Hawkes
- Clinical Neurosciences and Experimental Sciences; Faculty of Medicine; University of Southampton; Southampton; UK
| | - M. Jeffrey
- Animal Health and Veterinary Laboratories Agency (AHVLA); Bush Loan Penicuik; Edinburgh; UK
| | - R. N. Kalaria
- Centre for Brain Ageing & Vitality; Institute for Ageing and Health; Newcastle University; Newcastle Upon Tyne; UK
| | - R. O. Weller
- Clinical Neurosciences and Experimental Sciences; Faculty of Medicine; University of Southampton; Southampton; UK
| |
Collapse
|
50
|
Zaucker A, Mercurio S, Sternheim N, Talbot WS, Marlow FL. notch3 is essential for oligodendrocyte development and vascular integrity in zebrafish. Dis Model Mech 2013; 6:1246-59. [PMID: 23720232 PMCID: PMC3759344 DOI: 10.1242/dmm.012005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Accepted: 05/22/2013] [Indexed: 01/08/2023] Open
Abstract
Mutations in the human NOTCH3 gene cause CADASIL syndrome (cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy). CADASIL is an inherited small vessel disease characterized by diverse clinical manifestations including vasculopathy, neurodegeneration and dementia. Here we report two mutations in the zebrafish notch3 gene, one identified in a previous screen for mutations with reduced expression of myelin basic protein (mbp) and another caused by a retroviral insertion. Reduced mbp expression in notch3 mutant embryos is associated with fewer oligodendrocyte precursor cells (OPCs). Despite an early neurogenic phenotype, mbp expression recovered at later developmental stages and some notch3 homozygous mutants survived to adulthood. These mutants, as well as adult zebrafish carrying both mutant alleles together, displayed a striking stress-associated accumulation of blood in the head and fins. Histological analysis of mutant vessels revealed vasculopathy, including: an enlargement (dilation) of vessels in the telencephalon and fin, disorganization of the normal stereotyped arrangement of vessels in the fin, and an apparent loss of arterial morphological structure. Expression of hey1, a well-known transcriptional target of Notch signaling, was greatly reduced in notch3 mutant fins, suggesting that Notch3 acts via a canonical Notch signaling pathway to promote normal vessel structure. Ultrastructural analysis confirmed the presence of dilated vessels in notch3 mutant fins and revealed that the vessel walls of presumed arteries showed signs of deterioration. Gaps in the arterial wall and the presence of blood cells outside of vessels in mutants indicated that compromised vessel structure led to hemorrhage. In notch3 heterozygotes, we found elevated expression of both notch3 itself and target genes, indicating that specific alterations in gene expression due to partial loss of Notch3 function might contribute to the abnormalities observed in heterozygous larvae and adults. Our analysis of zebrafish notch3 mutants indicates that Notch3 regulates OPC development and mbp gene expression in larvae, and maintains vascular integrity in adults.
Collapse
Affiliation(s)
- Andreas Zaucker
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Sara Mercurio
- Department of Developmental Biology, Stanford University School of Medicine, Beckman Center B300, 279 Campus Drive, Stanford, CA 94305, USA
| | - Nitzan Sternheim
- Department of Developmental Biology, Stanford University School of Medicine, Beckman Center B300, 279 Campus Drive, Stanford, CA 94305, USA
| | - William S. Talbot
- Department of Developmental Biology, Stanford University School of Medicine, Beckman Center B300, 279 Campus Drive, Stanford, CA 94305, USA
| | - Florence L. Marlow
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| |
Collapse
|