1
|
Ip BYM, Ko H, Lam BYK, Au LWC, Lau AYL, Huang J, Kwok AJ, Leng X, Cai Y, Leung TWH, Mok VCT. Current and Future Treatments of Vascular Cognitive Impairment. Stroke 2024; 55:822-839. [PMID: 38527144 DOI: 10.1161/strokeaha.123.044174] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Affiliation(s)
- Bonaventure Yiu Ming Ip
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
- Kwok Tak Seng Centre for Stroke Research and Intervention, Hong Kong SAR, China (B.Y.M.I., X.L., T.W.H.L.)
| | - Ho Ko
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Bonnie Yin Ka Lam
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Lisa Wing Chi Au
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Alexander Yuk Lun Lau
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
| | - Junzhe Huang
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Andrew John Kwok
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Xinyi Leng
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Kwok Tak Seng Centre for Stroke Research and Intervention, Hong Kong SAR, China (B.Y.M.I., X.L., T.W.H.L.)
| | - Yuan Cai
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Thomas Wai Hong Leung
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Kwok Tak Seng Centre for Stroke Research and Intervention, Hong Kong SAR, China (B.Y.M.I., X.L., T.W.H.L.)
| | - Vincent Chung Tong Mok
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| |
Collapse
|
2
|
Goligorsky MS. Permissive role of vascular endothelium in fibrosis: focus on the kidney. Am J Physiol Cell Physiol 2024; 326:C712-C723. [PMID: 38223932 PMCID: PMC11193458 DOI: 10.1152/ajpcell.00526.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
Fibrosis, the morphologic end-result of a plethora of chronic conditions and the scorch for organ function, has been thoroughly investigated. One aspect of its development and progression, namely the permissive role of vascular endothelium, has been overshadowed by studies into (myo)fibroblasts and TGF-β; thus, it is the subject of the present review. It has been established that tensile forces of the extracellular matrix acting on cells are a prerequisite for mechanochemical coupling, leading to liberation of TGF-β and formation of myofibroblasts. Increased tensile forces are prompted by elevated vascular permeability in response to diverse stressors, resulting in the exudation of fibronectin, fibrinogen/fibrin, and other proteins, all stiffening the extracellular matrix. These processes lead to the development of endothelial cells dysfunction, endothelial-to-mesenchymal transition, premature senescence of endothelial cells, perturbation of blood flow, and gradual obliteration of microvasculature, leaving behind "string" vessels. The resulting microvascular rarefaction is not only a constant companion of fibrosis but also an adjunct mechanism of its progression. The deepening knowledge of the above chain of pathogenetic events involving endothelial cells, namely increased permeability-stiffening of the matrix-endothelial dysfunction-microvascular rarefaction-tissue fibrosis, may provide a roadmap for therapeutic interventions deemed to curtail and reverse fibrosis.
Collapse
Affiliation(s)
- Michael S Goligorsky
- Department of Medicine, New York Medical College, Touro University, Valhalla, New York, United States
- Department of Pharmacology, New York Medical College, Touro University, Valhalla, New York, United States
- Department of Physiology, New York Medical College, Touro University, Valhalla, New York, United States
| |
Collapse
|
3
|
Kreutzer AG, Parrocha CMT, Haerianardakani S, Guaglianone G, Nguyen JT, Diab MN, Yong W, Perez-Rosendahl M, Head E, Nowick JS. Antibodies Raised Against an Aβ Oligomer Mimic Recognize Pathological Features in Alzheimer's Disease and Associated Amyloid-Disease Brain Tissue. ACS CENTRAL SCIENCE 2024; 10:104-121. [PMID: 38292607 PMCID: PMC10823522 DOI: 10.1021/acscentsci.3c00592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 11/27/2023] [Accepted: 11/27/2023] [Indexed: 02/01/2024]
Abstract
Antibodies that target the β-amyloid peptide (Aβ) and its associated assemblies are important tools in Alzheimer's disease research and have emerged as promising Alzheimer's disease therapies. This paper reports the creation and characterization of a triangular Aβ trimer mimic composed of Aβ17-36 β-hairpins and the generation and study of polyclonal antibodies raised against the Aβ trimer mimic. The Aβ trimer mimic is covalently stabilized by three disulfide bonds at the corners of the triangular trimer to create a homogeneous oligomer. Structural, biophysical, and cell-based studies demonstrate that the Aβ trimer mimic shares characteristics with oligomers of full-length Aβ. X-ray crystallography elucidates the structure of the trimer and reveals that four copies of the trimer assemble to form a dodecamer. SDS-PAGE, size exclusion chromatography, and dynamic light scattering reveal that the trimer also forms higher-order assemblies in solution. Cell-based toxicity assays show that the trimer elicits LDH release, decreases ATP levels, and activates caspase-3/7 mediated apoptosis. Immunostaining studies on brain slices from people who lived with Alzheimer's disease and people who lived with Down syndrome reveal that the polyclonal antibodies raised against the Aβ trimer mimic recognize pathological features including different types of Aβ plaques and cerebral amyloid angiopathy.
Collapse
Affiliation(s)
- Adam G. Kreutzer
- Department
of Chemistry, University of California Irvine, Irvine, California 92697, United States
| | - Chelsea Marie T. Parrocha
- Department
of Pharmaceutical Sciences, University of
California Irvine, Irvine, California 92697, United States
| | - Sepehr Haerianardakani
- Department
of Chemistry, University of California Irvine, Irvine, California 92697, United States
| | - Gretchen Guaglianone
- Department
of Chemistry, University of California Irvine, Irvine, California 92697, United States
| | - Jennifer T. Nguyen
- Department
of Pharmaceutical Sciences, University of
California Irvine, Irvine, California 92697, United States
| | - Michelle N. Diab
- Department
of Chemistry, University of California Irvine, Irvine, California 92697, United States
| | - William Yong
- Department
of Pathology and Laboratory Medicine, University
of California Irvine, Irvine, California 92697, United States
| | - Mari Perez-Rosendahl
- Department
of Pathology and Laboratory Medicine, University
of California Irvine, Irvine, California 92697, United States
| | - Elizabeth Head
- Department
of Pathology and Laboratory Medicine, University
of California Irvine, Irvine, California 92697, United States
| | - James S. Nowick
- Department
of Chemistry, University of California Irvine, Irvine, California 92697, United States
- Department
of Pharmaceutical Sciences, University of
California Irvine, Irvine, California 92697, United States
| |
Collapse
|
4
|
Yuan L, Chen X, Jankovic J, Deng H. CADASIL: A NOTCH3-associated cerebral small vessel disease. J Adv Res 2024:S2090-1232(24)00001-8. [PMID: 38176524 DOI: 10.1016/j.jare.2024.01.001] [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: 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
|
5
|
Miyagi T, Ishida A, Shinzato T, Ohya Y. Arterial Stiffness Is Associated With Small Vessel Disease Irrespective of Blood Pressure in Stroke-Free Individuals. Stroke 2023; 54:2814-2821. [PMID: 37846566 DOI: 10.1161/strokeaha.123.042512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 08/03/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND Arterial stiffness and hypertension are important risk factors for cerebral small vessel disease (CSVD). Clinically, there are hypertensive patients with low pulse wave velocity (PWV) and nonhypertensive individuals with high PWV. We aimed to determine the effects of arterial stiffness on CSVD in normotensive individuals. METHODS An observational cross-sectional study was conducted in 1894 stroke-free participants who underwent brain magnetic resonance imaging and brachial-ankle pulse wave velocity (baPWV) measurements at a health checkup between 2013 and 2020. CSVD was defined as any of following: white matter hyperintensities, cerebral microbleeds, silent lacunar infarcts, and enlarged perivascular spaces. baPWV was measured using an automatic oscillometric device. Participants were divided into 4 groups according to the following cutoff points: low blood pressure (BP, <120/80 mm Hg) with low baPWV (<14.63 m/s, a cutoff value that predicted CSVD); high BP (≥120/80 mm Hg) with low baPWV; low BP with high baPWV (≥14.63 m/s); and high BP with high baPWV. RESULTS The mean age of the participants was 57±13 years (41% women). The prevalence of CSVD was 718 (38%), which was higher in the low BP with high baPWV (56%) and high BP with high baPWV (55%) groups than in the high BP with low baPWV (24%) and low BP with low baPWV (22%) groups. Compared with the low BP with low baPWV group, the low BP with high baPWV group (odds ratio, 1.63 [95% CI, 1.09-2.43]) and the high BP with high baPWV group (odds ratio, 1.86 [95% CI, 1.39-2.49]) had a significantly higher multivariable-adjusted risk for CSVD. CONCLUSIONS Individuals with a high baPWV had a higher prevalence of CSVD, independent of BP status. Higher arterial stiffness is likely to be a more important risk factor for CSVD than BP status in stroke-free individuals.
Collapse
Affiliation(s)
- Tomo Miyagi
- Department of Cardiovascular Medicine, Nephrology and Neurology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan (T.M., A.I., Y.O.)
| | - Akio Ishida
- Department of Cardiovascular Medicine, Nephrology and Neurology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan (T.M., A.I., Y.O.)
| | | | - Yusuke Ohya
- Department of Cardiovascular Medicine, Nephrology and Neurology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan (T.M., A.I., Y.O.)
| |
Collapse
|
6
|
Arthur E, Ravichandran S, Snyder PJ, Alber J, Strenger J, Bittner AK, Khankan R, Adams SL, Putnam NM, Lypka KR, Piantino JA, Sinoff S. Retinal mid-peripheral capillary free zones are enlarged in cognitively unimpaired older adults at high risk for Alzheimer's disease. Alzheimers Res Ther 2023; 15:172. [PMID: 37828548 PMCID: PMC10568786 DOI: 10.1186/s13195-023-01312-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND Compared to standard neuro-diagnostic techniques, retinal biomarkers provide a probable low-cost and non-invasive alternative for early Alzheimer's disease (AD) risk screening. We have previously quantified the periarteriole and perivenule capillary free zones (mid-peripheral CFZs) in cognitively unimpaired (CU) young and older adults as novel metrics of retinal tissue oxygenation. There is a breakdown of the inner retinal blood barrier, pericyte loss, and capillary non-perfusion or dropout in AD leading to potential enlargement of the mid-peripheral CFZs. We hypothesized the mid-peripheral CFZs will be enlarged in CU older adults at high risk for AD compared to low-risk individuals. METHODS 20 × 20° optical coherence tomography angiography images consisting of 512 b-scans, 512 A-scans per b-scan, 12-µm spacing between b-scans, and 5 frames averaged per each b-scan location of the central fovea and of paired major arterioles and venules with their surrounding capillaries inferior to the fovea of 57 eyes of 37 CU low-risk (mean age: 66 years) and 50 eyes of 38 CU high-risk older adults (mean age: 64 years; p = 0.24) were involved in this study. High-risk participants were defined as having at least one APOE e4 allele and a positive first-degree family history of AD while low-risk participants had neither of the two criteria. All participants had Montreal Cognitive Assessment scores ≥ 26. The mid-peripheral CFZs were computed in MATLAB and compared between the two groups. RESULTS The periarteriole CFZ of the high-risk group (75.8 ± 9.19 µm) was significantly larger than that of the low-risk group (71.3 ± 7.07 µm), p = 0.005, Cohen's d = 0.55. The perivenule CFZ of the high-risk group (60.4 ± 8.55 µm) was also significantly larger than that of the low-risk group (57.3 ± 6.40 µm), p = 0.034, Cohen's d = 0.42. There were no significant differences in foveal avascular zone (FAZ) size, FAZ effective diameter, and vessel density between the two groups, all p > 0.05. CONCLUSIONS Our results show larger mid-peripheral CFZs in CU older adults at high risk for AD, with the potential for the periarteriole CFZ to serve as a novel retinal vascular biomarker for early AD risk detection.
Collapse
Affiliation(s)
- Edmund Arthur
- School of Optometry, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Swetha Ravichandran
- School of Optometry, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Peter J Snyder
- Department of Neurology, Alpert Medical School of Brown University, Providence, RI, USA
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Jessica Alber
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
- Butler Hospital Memory & Aging Program, Providence, RI, USA
| | - Jennifer Strenger
- Butler Hospital Memory & Aging Program, Providence, RI, USA
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
| | - Ava K Bittner
- Department of Ophthalmology, Stein Eye Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Rima Khankan
- Southern California College of Optometry, Marshall B. Ketchum University, Fullerton, CA, USA
| | | | - Nicole M Putnam
- State University of New York College of Optometry, New York, NY, USA
| | - Karin R Lypka
- Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA
| | - Juan A Piantino
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | | |
Collapse
|
7
|
Liu Y, Yan Z, Ren Y, Wang W, Ke Y, Wang Y, Qi R. Electroacupuncture inhibits hippocampal neuronal apoptosis and improves cognitive dysfunction in mice with vascular dementia via the JNK signaling pathway. Acupunct Med 2023; 41:284-296. [PMID: 36482691 DOI: 10.1177/09645284221136878] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
BACKGROUND Electroacupuncture (EA) has been shown to reduce cognitive impairment in vascular dementia (VaD) patients. However, the mechanism of action remains unknown. OBJECTIVE The c-Jun N-terminal kinase (JNK) signaling pathway plays an important role in apoptosis. Herein, we focused on whether EA can inhibit apoptosis and alleviate cognitive impairment by regulating the JNK signaling pathway using a mouse model of VaD induced by modified bilateral common carotid artery occlusion (BCCAo). METHODS In experiment I, 60 mice were randomly divided into a Sham group, BCCAo group, BCCAo + EA group, BCCAo + Sham-EA group, BCCAo + SP group (receiving the selective JNK inhibitor SP600125) and BCCAo + SP + EA group. Morris water maze tests, TdT-mediated dUTP-biotin nick end labeling (TUNEL) staining and flow cytometry were used to evaluate the effect of the EA intervention on VaD. In experiment II, 30 mice were randomly divided into a Sham group, BCCAo group, BCCAo + EA group, BCCAo + SP group and BCCAo + SP + EA group. Western blotting and real-time reverse transcription polymerase chain reaction were used to detect protein and mRNA expression of key factors in the JNK signaling pathway in the hippocampus. RESULTS EA, SP600125 and EA + SP600125 significantly inhibited hippocampal apoptosis and improved cognitive impairment in VaD model mice. There were no significant differences between the BCCAo group and the BCCAo + Sham-EA group. EA, EA + SP600125 and SP600125 inhibited the phosphorylation of JNK and caspase-3. EA and EA + SP600125 promoted protein and mRNA expression of B-cell lymphoma 2 (Bcl-2) in the hippocampus of VaD mice and inhibited protein and mRNA expression of activator protein (AP)-1, p53 and Bax. CONCLUSION EA can reverse cognitive deficits and inhibit hippocampal neuronal apoptosis in VaD model mice, at least partially through inhibition of the JNK signaling pathway and regulation of apoptosis signals.
Collapse
Affiliation(s)
- Yaru Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Zhenyang Yan
- Weifang Traditional Chinese Hospital, Weifang, China
| | - Yafei Ren
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Woyu Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yinze Ke
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yifan Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Rongming Qi
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
8
|
Xia H, Jin B, Su C, Zhao K, Ma R. Early diagnostic value of intima-media thickness and D-dimer levels for vascular dementia. Medicine (Baltimore) 2023; 102:e34149. [PMID: 37352042 PMCID: PMC10289637 DOI: 10.1097/md.0000000000034149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/07/2023] [Accepted: 06/08/2023] [Indexed: 06/25/2023] Open
Abstract
To evaluate the diagnostic value of combination of D-dimer and Doppler Intima-Media Thickness (IMT) based on clinical data for vascular dementia (VaD). In this study, 100 cases of initial VaD were diagnosed by Zhenjiang Fourth People Hospital as VaD group, and 100 healthy were engaged as control group, medical history and test results were collected for evaluation. IMT and D-dimer were highly correlated with the onset of VaD. The sensitivity of combination diagnosis for early VaD: 90.2%, the specificity: 87.9%, the area under the curve (AUC) is 0.872, which were superior to IMT or D-dimer alone. D-dimer and IMT are significantly increased in the initial onset of cerebrovascular diseases, and combined detection of them is conducive to early diagnosis and evaluation of the disease. Given the limitations of D-dimer and IMT, combined detection is more conducive to early diagnosis and prognosis, and can be used as screening and routine examination items, even evaluate the severity of cognitive decline.
Collapse
Affiliation(s)
- Haiping Xia
- Zhenjiang 4th People’s Hospital Affiliated to Jiangsu University, Jiangsu, China
| | - Biao Jin
- Zhenjiang 4th People’s Hospital Affiliated to Jiangsu University, Jiangsu, China
| | - Cen Su
- Zhenjiang 4th People’s Hospital Affiliated to Jiangsu University, Jiangsu, China
| | - Kangren Zhao
- Zhenjiang 4th People’s Hospital Affiliated to Jiangsu University, Jiangsu, China
| | - Rui Ma
- Department of Anatomy, Medical School, Jiangsu University, Jiangsu, China
| |
Collapse
|
9
|
Pansieri J, Hadley G, Lockhart A, Pisa M, DeLuca GC. Regional contribution of vascular dysfunction in white matter dementia: clinical and neuropathological insights. Front Neurol 2023; 14:1199491. [PMID: 37396778 PMCID: PMC10313211 DOI: 10.3389/fneur.2023.1199491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/25/2023] [Indexed: 07/04/2023] Open
Abstract
The maintenance of adequate blood supply and vascular integrity is fundamental to ensure cerebral function. A wide range of studies report vascular dysfunction in white matter dementias, a group of cerebral disorders characterized by substantial white matter damage in the brain leading to cognitive impairment. Despite recent advances in imaging, the contribution of vascular-specific regional alterations in white matter dementia has been not extensively reviewed. First, we present an overview of the main components of the vascular system involved in the maintenance of brain function, modulation of cerebral blood flow and integrity of the blood-brain barrier in the healthy brain and during aging. Second, we review the regional contribution of cerebral blood flow and blood-brain barrier disturbances in the pathogenesis of three distinct conditions: the archetypal white matter predominant neurocognitive dementia that is vascular dementia, a neuroinflammatory predominant disease (multiple sclerosis) and a neurodegenerative predominant disease (Alzheimer's). Finally, we then examine the shared landscape of vascular dysfunction in white matter dementia. By emphasizing the involvement of vascular dysfunction in the white matter, we put forward a hypothetical map of vascular dysfunction during disease-specific progression to guide future research aimed to improve diagnostics and facilitate the development of tailored therapies.
Collapse
|
10
|
Magaki SD, Vinters HV. Evaluating when (and how) hypertension may be 'good for your brain'. Brain Commun 2023; 5:fcad127. [PMID: 37113316 PMCID: PMC10128875 DOI: 10.1093/braincomms/fcad127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 03/16/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
This scientific commentary refers to 'Elevated late-life blood pressure may maintain brain oxygenation and slow amyloid-β accumulation, at the expense of cerebral vascular damage', by Tayler et al. (https://doi.org/10.1093/braincomms/fcad112).
Collapse
Affiliation(s)
- Shino D Magaki
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Harry V Vinters
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, CA 90095, USA
- Department of Neurology, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, CA 90095, USA
- Brain Research Institute, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
11
|
Magaki S, Chen Z, Severance A, Williams CK, Diaz R, Fang C, Khanlou N, Yong WH, Paganini-Hill A, Kalaria RN, Vinters HV, Fisher M. Neuropathology of microbleeds in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). J Neuropathol Exp Neurol 2023; 82:333-344. [PMID: 36715085 PMCID: PMC10025882 DOI: 10.1093/jnen/nlad004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Cerebral microbleeds (CMBs) detected on magnetic resonance imaging are common in patients with cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). The neuropathologic correlates of CMBs are unclear. In this study, we characterized findings relevant to CMBs in autopsy brain tissue of 8 patients with genetically confirmed CADASIL and 10 controls within the age range of the CADASIL patients by assessing the distribution and extent of hemosiderin/iron deposits including perivascular hemosiderin leakage (PVH), capillary hemosiderin deposits, and parenchymal iron deposits (PID) in the frontal cortex and white matter, basal ganglia and cerebellum. We also characterized infarcts, vessel wall thickening, and severity of vascular smooth muscle cell degeneration. CADASIL subjects had a significant increase in hemosiderin/iron deposits compared with controls. This increase was principally seen with PID. Hemosiderin/iron deposits were seen in the majority of CADASIL subjects in all brain areas. PVH was most pronounced in the frontal white matter and basal ganglia around small to medium sized arterioles, with no predilection for the vicinity of vessels with severe vascular changes or infarcts. CADASIL subjects have increased brain hemosiderin/iron deposits but these do not occur in a periarteriolar distribution. Pathogenesis of these lesions remains uncertain.
Collapse
Affiliation(s)
- Shino Magaki
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, California, USA
| | - Zesheng Chen
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, California, USA
| | - Alyscia Severance
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, California, USA
| | - Christopher K Williams
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, California, USA
| | - Ramiro Diaz
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, California, USA
| | - Chuo Fang
- Department of Neurology, University of California-Irvine School of Medicine, Irvine, California, USA
| | - Negar Khanlou
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, California, USA
| | - William H Yong
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, California, USA
| | - Annlia Paganini-Hill
- Department of Neurology, University of California-Irvine School of Medicine, Irvine, California, USA
| | - Rajesh N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - Harry V Vinters
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, California, USA
- Department of Neurology, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, California, USA
- Brain Research Institute, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, California, USA
| | - Mark Fisher
- Department of Neurology, University of California-Irvine School of Medicine, Irvine, California, USA
- Department of Pathology and Laboratory Medicine, University of California-Irvine School of Medicine, Irvine, California, USA
| |
Collapse
|
12
|
Fang C, Magaki SD, Kim RC, Kalaria RN, Vinters HV, Fisher M. Arteriolar neuropathology in cerebral microvascular disease. Neuropathol Appl Neurobiol 2023; 49:e12875. [PMID: 36564356 DOI: 10.1111/nan.12875] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/14/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022]
Abstract
Cerebral microvascular disease (MVD) is an important cause of vascular cognitive impairment. MVD is heterogeneous in aetiology, ranging from universal ageing to the sporadic (hypertension, sporadic cerebral amyloid angiopathy [CAA] and chronic kidney disease) and the genetic (e.g., familial CAA, cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy [CADASIL] and cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy [CARASIL]). The brain parenchymal consequences of MVD predominantly consist of lacunar infarcts (lacunes), microinfarcts, white matter disease of ageing and microhaemorrhages. MVD is characterised by substantial arteriolar neuropathology involving ubiquitous vascular smooth muscle cell (SMC) abnormalities. Cerebral MVD is characterised by a wide variety of arteriolar injuries but only a limited number of parenchymal manifestations. We reason that the cerebral arteriole plays a dominant role in the pathogenesis of each type of MVD. Perturbations in signalling and function (i.e., changes in proliferation, apoptosis, phenotypic switch and migration of SMC) are prominent in the pathogenesis of cerebral MVD, making 'cerebral angiomyopathy' an appropriate term to describe the spectrum of pathologic abnormalities. The evidence suggests that the cerebral arteriole acts as both source and mediator of parenchymal injury in MVD.
Collapse
Affiliation(s)
- Chuo Fang
- Department of Neurology, University of California, Irvine Medical Center, 101 The City Drive South Shanbrom Hall (Building 55), Room 121, Orange, 92868, California, USA
| | - Shino D Magaki
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Ronald C Kim
- Department of Pathology & Laboratory Medicine, University of California, Irvine, Orange, California, USA
| | - Raj N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Harry V Vinters
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA.,Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Mark Fisher
- Department of Neurology, University of California, Irvine Medical Center, 101 The City Drive South Shanbrom Hall (Building 55), Room 121, Orange, 92868, California, USA.,Department of Pathology & Laboratory Medicine, University of California, Irvine, Orange, California, USA
| |
Collapse
|
13
|
de Liyis BG, Sutedja JC, Kesuma PMI, Liyis S, Widyadharma IPE. A review of literature on Compound 21-loaded gelatin nanoparticle: a promising nose-to-brain therapy for multi-infarct dementia. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2023. [DOI: 10.1186/s41983-023-00621-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
AbstractMulti-infarct dementia (MID) is described as a chronic progressive decline in cortical cognitive function due to the occurrence of multiple infarcts in the cerebral vascularization throughout the gray and white matter. Current therapies of MID mostly focus only on slowing down MID progression and symptomatic medications. A novel therapy which is able to provide both preventive and curative properties for MID is of high interest. The purpose of this review is to identify the potential of Compound 21 (C21) gelatin nanoparticle through the nose-to-brain route as therapy for MID. C21, an angiotensin II type 2 receptor (AT2R) agonist, has shown to reduce the size of cerebral infarct in rodent models, resulting in the preservation and improvement of overall cognitive function and prevention of secondary neurodegenerative effects. It is also shown that C21 decreases neuronal apoptosis, improves damaged axons, and encourage synapse development. The challenge remains in preventing systemic AT2R activation and increasing its low oral bioavailability which can be overcome through nose-to-brain administration of C21. Nose-to-brain drug delivery of C21 significantly increases drug efficiency and limits C21 exposure in order to specifically target the multiple infarcts located in the cerebral cortex. Adhering C21 onto gelatin nanoparticles may enable longer contact time with the olfactory and the trigeminal nerve endings, increasing the potency of C21. In summary, treatment of C21 gelatin nanoparticle through nose-to-brain delivery shows high potential as therapy for vascular dementia. However, clinical trials must be further studied in order to test the safety and efficacy of C21.
Collapse
|
14
|
Gao L, Liu F, Liu R. The Mechanism of Aerobic Exercise Regulating the PI3K/Akt-mTOR Signaling Pathway Intervenes in Hippocampal Neuronal Apoptosis in Vascular Dementia Rats. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:ijerph20031893. [PMID: 36767264 PMCID: PMC9914742 DOI: 10.3390/ijerph20031893] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 06/14/2023]
Abstract
BACKGROUND The purpose of this paper is to explore the mechanism of aerobic exercise regulating autophagy through the PI3K/Akt-mTOR signaling pathway and its participation in apoptosis, to protect the hippocampal nerves from damage in vascular dementia rats. METHODS Thirty-six healthy male SD rats were randomly divided into a sham group, a model group, and a model exercise group. A neurobehavioral assessment was used to determine the memory and exploration abilities of the rats. A TUNEL assay was used to detect hippocampal neuron apoptosis. Immunohistochemical and Western blot analyses were used to analyze LC3Ⅱ and the beclin-1 protein. An RT-PCR detected the differential expression of mRNA. RESULTS The results of the neurobehavioral tests showed that the platform latency time of the rats with vascular dementia was prolonged. Aerobic exercise significantly shortens the swimming time of rats in platform latency. The TUNEL results showed that the TUNEL-positive cells of the hippocampal neurons in the model group increased; the expression of pro-apoptotic genes caspase-3 and Bax mRNA was up-regulated, and the expression of Bcl-2 mRNA was down-regulated. Aerobic exercise reduced hippocampal neuronal apoptosis, up-regulated Bcl-2 mRNA, and down-regulated caspase-3 and Bax mRNA. The LC3Ⅱ and Beclin-1 proteins, detected by immunohistochemistry and a Western blot analysis, showed that the protein expression in the hippocampi of rats with vascular dementia increased. Aerobic exercise reduced LC3Ⅱ and Beclin-1 protein expression. The results of the RT-PCR showed similar changes. CONCLUSIONS Aerobic exercise could improve the learning and memory abilities of vascular dementia rats, moderately regulate the process of autophagy, reduce the TUNEL-positive cells of hippocampal neurons, repair damaged hippocampal neurons by regulating the autophagy signaling pathway PI3K/Akt-mTOR, and improve hippocampal function.
Collapse
Affiliation(s)
- Lei Gao
- Department of Physical Education, Yuzhang Normal University, Nanchang 330103, China
| | - Fushun Liu
- Police Sports Department, Zhejiang Police College, Hangzhou 310053, China
| | - Ruilian Liu
- College of Physical Education, Yichun University, Yichun 336000, China
| |
Collapse
|
15
|
Dover M, Moseley T, Biskaduros A, Paulchakrabarti M, Hwang SH, Hammock B, Choudhury B, Kaczor-Urbanowicz KE, Urbanowicz A, Morselli M, Dang J, Pellegrini M, Paul K, Bentolila LA, Fiala M. Polyunsaturated Fatty Acids Mend Macrophage Transcriptome, Glycome, and Phenotype in the Patients with Neurodegenerative Diseases, Including Alzheimer's Disease. J Alzheimers Dis 2023; 91:245-261. [PMID: 36373322 PMCID: PMC9881025 DOI: 10.3233/jad-220764] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Macrophages of healthy subjects have a pro-resolution phenotype, upload amyloid-β (Aβ) into endosomes, and degrade Aβ, whereas macrophages of patients with Alzheimer's disease (AD) generally have a pro-inflammatory phenotype and lack energy for brain clearance of Aβ. OBJECTIVE To clarify the pathogenesis of sporadic AD and therapeutic effects of polyunsaturated fatty acids (PUFA) with vitamins B and D and antioxidants on monocyte/macrophage (MM) migration in the AD brain, MM transcripts in energy and Aβ degradation, MM glycome, and macrophage clearance of Aβ. METHODS We followed for 31.3 months (mean) ten PUFA-supplemented neurodegenerative patients: 3 with subjective cognitive impairment (SCI), 2 with mild cognitive impairment (MCI), 3 MCI/vascular cognitive impairment, 2 with dementia with Lewy bodies, and 7 non-supplemented caregivers. We examined: monocyte migration in the brain and a blood-brain barrier model by immunochemistry and electron microscopy; macrophage transcriptome by RNAseq; macrophage glycome by N-glycan profiling and LTQ-Orbitrap mass spectrometry; and macrophage phenotype and phagocytosis by immunofluorescence. RESULTS MM invade Aβ plaques, upload but do not degrade Aβ, and release Aβ into vessels, which develop cerebrovascular amyloid angiopathy (CAA); PUFA upregulate energy and Aβ degradation enzyme transcripts in macrophages; PUFA enhance sialylated N-glycans in macrophages; PUFA reduce oxidative stress and increase pro-resolution MM phenotype, mitochondrial membrane potential, and Aβ phagocytosis (p < 0.001). CONCLUSION Macrophages of SCI, MCI, and AD patients have interrelated defects in the transcriptome, glycome, Aβ phagocytosis, and Aβ degradation. PUFA mend macrophage transcriptome, enrich glycome, enhance Aβ clearance, and benefit the cognition of early-stage AD patients.
Collapse
Affiliation(s)
- Mary Dover
- Department of Molecular, Cell and Developmental Biology, UCLA School of Life Sciences, Los Angeles, CA, USA
- Department of Integrated Biology and Physiology, UCLA School of Life Sciences, Los Angeles, CA, USA
| | - Taylor Moseley
- Department of Molecular, Cell and Developmental Biology, UCLA School of Life Sciences, Los Angeles, CA, USA
| | - Adrienne Biskaduros
- Department of Molecular, Cell and Developmental Biology, UCLA School of Life Sciences, Los Angeles, CA, USA
| | | | - Sung Hee Hwang
- Department of Entomology and Nematology, and UCDavis Comprehensive Cancer Center, University of California – Davis, Davis, CA, USA
| | - Bruce Hammock
- Department of Entomology and Nematology, and UCDavis Comprehensive Cancer Center, University of California – Davis, Davis, CA, USA
| | - Biswa Choudhury
- GlycoAnalytics Core, University of California SanDiego Health Sciences, La Jolla, CA, USA
| | | | - Andrzej Urbanowicz
- Institute of Control and Computation Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Marco Morselli
- Department of Molecular, Cell and Developmental Biology, UCLA School of Life Sciences, Los Angeles, CA, USA
| | - Johnny Dang
- Department of Molecular, Cell and Developmental Biology, UCLA School of Life Sciences, Los Angeles, CA, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, UCLA School of Life Sciences, Los Angeles, CA, USA
| | - Ketema Paul
- Department of Integrated Biology and Physiology, UCLA School of Life Sciences, Los Angeles, CA, USA
| | - Laurent A. Bentolila
- Advanced Light Microscopy and Spectroscopy Laboratory, California Nano Systems Institute, UCLA, Los Angeles, CA, USA
| | - Milan Fiala
- Department of Molecular, Cell and Developmental Biology, UCLA School of Life Sciences, Los Angeles, CA, USA
- Department of Integrated Biology and Physiology, UCLA School of Life Sciences, Los Angeles, CA, USA
| |
Collapse
|
16
|
Zhao J, Li Q, Meng L, Wang F, Li Q, Yang F, Wang M, Yu M, Zhang J, Li S, Ji S. Relationship between MMP-9 serum levels and tHcy levels and total imaging load and cognitive dysfunction. J Stroke Cerebrovasc Dis 2022; 31:106759. [DOI: 10.1016/j.jstrokecerebrovasdis.2022.106759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 08/28/2022] [Accepted: 09/04/2022] [Indexed: 11/05/2022] Open
|
17
|
Brier MR, Blazey T, Raichle ME, Morris JC, Benzinger TLS, Vlassenko AG, Snyder AZ, Goyal MS. Increased white matter glycolysis in humans with cerebral small vessel disease. NATURE AGING 2022; 2:991-999. [PMID: 37118084 PMCID: PMC10155263 DOI: 10.1038/s43587-022-00303-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 10/03/2022] [Indexed: 04/30/2023]
Abstract
White matter lesions in cerebral small vessel disease are related to ischemic injury and increase the risk of stroke and cognitive decline. Pathological changes due to cerebral small vessel disease are increasingly recognized outside of discrete lesions, but the metabolic alterations in nonlesional tissue has not been described. Aerobic glycolysis is critical to white matter myelin homeostasis and repair. In this study, we examined cerebral metabolism of glucose and oxygen as well as blood flow in individuals with and without cerebral small vessel disease using multitracer positron emission tomography. We show that glycolysis is relatively elevated in nonlesional white matter in individuals with small vessel disease relative to healthy, age-matched controls. On the other hand, in young healthy individuals, glycolysis is relatively low in areas of white matter susceptible to lesion formation. These results suggest that increased white matter glycolysis is a marker of pathology associated with small vessel disease.
Collapse
Affiliation(s)
- Matthew R Brier
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Tyler Blazey
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Marcus E Raichle
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrei G Vlassenko
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Abraham Z Snyder
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Manu S Goyal
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
18
|
Nägga K, Bränsvik V, Stomrud E, Melander O, Nilsson PM, Gustavsson AM, Hansson O. Prevalence and Ascertainment of Dementia Cases in the Malmö Diet and Cancer Study. J Alzheimers Dis Rep 2022; 6:529-538. [PMID: 36275419 PMCID: PMC9535608 DOI: 10.3233/adr-220003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 08/14/2022] [Indexed: 11/15/2022] Open
Abstract
Background: Register diagnoses, both hospital-based and from open clinic care, are often used in research studies in Sweden. The validity of such diagnoses has been debated and a validation assessment can improve the diagnostic accuracy for use in research studies. Objective: The aim of this study was to investigate the quality of register-derived dementia diagnoses in the Malmö Diet and Cancer population study (MDCS) and to validate these diagnoses using systematic criteria. Methods: MDCS is a population-based prospective study comprising 30,446 participants. Register diagnoses of dementia for the MDCS population were derived from the Swedish National Patient Register (NPR) and validated through re-evaluation of available medical records by physicians. Results: In the MDCS cohort, 2,206 participants were diagnosed with dementia according to the NPR during a mean follow-up of 18.1 years. The general dementia diagnosis was valid in 96% of the cases, but 40% of the specific dementia diagnoses were changed during the process of reevaluation. The diagnostic validity varied between 25.2% and 82.9% for the different diagnoses. The results from the validity assessment per diagnostic category revealed that the validity of the NPR diagnoses was higher for the more specific diagnoses and lower for unspecified dementia. The major diagnostic shift during the re-evaluation was from unspecified dementia to more specific diagnoses. Conclusion: Validation of dementia diagnoses using medical records results in more precise diagnoses. Dementia diagnoses derived from registers should be validated in order to study associations between influential factors and different dementia diagnoses.
Collapse
Affiliation(s)
- Katarina Nägga
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
- Department of Acute Internal Medicine and Geriatrics, Linköping University, Linköping, Sweden
| | - Vanja Bränsvik
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences Malmö, Lund University, Sweden
| | | | - Anna-Märta Gustavsson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
19
|
Liu X, Sun P, Yang J, Fan Y. Biomarkers involved in the pathogenesis of cerebral small-vessel disease. Front Neurol 2022; 13:969185. [PMID: 36119691 PMCID: PMC9475115 DOI: 10.3389/fneur.2022.969185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Cerebral small-vessel disease (CSVD) has been found to have a strong association with vascular cognitive impairment (VCI) and functional loss in elderly patients. At present, the diagnosis of CSVD mainly relies on brain neuroimaging markers, but they cannot fully reflect the overall picture of the disease. Currently, some biomarkers were found to be related to CSVD, but the underlying mechanisms remain unclear. We aimed to systematically review and summarize studies on the progress of biomarkers related to the pathogenesis of CSVD, which is mainly the relationship between these indicators and neuroimaging markers of CSVD. Concerning the pathophysiological mechanism of CSVD, the biomarkers of CSVD have been described as several categories related to sporadic and genetic factors. Monitoring of biomarkers might contribute to the early diagnosis and progression prediction of CSVD, thus providing ideas for better diagnosis and treatment of CSVD.
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW This article gives a broad overview of vascular cognitive impairment and dementia, including epidemiology, pathophysiology, clinical approach, and management. Emphasis is placed on understanding the common underlying types of cerebrovascular disease (including atherosclerosis, arteriolosclerosis, and cerebral amyloid angiopathy) and awareness of rare inherited cerebrovascular disorders. RECENT FINDINGS The pathophysiology of vascular cognitive impairment and dementia is heterogeneous, and the most recent diagnostic criteria for vascular cognitive impairment and dementia break down the diagnosis of major vascular dementia into four phenotypic categories, including subcortical ischemic vascular dementia, poststroke dementia, multi-infarct dementia, and mixed dementia. Control of cardiovascular risk factors, including management of midlife blood pressure, cholesterol, and blood sugars, remains the mainstay of prevention for vascular cognitive impairment and dementia. Cerebral amyloid angiopathy requires special consideration when it comes to risk factor management given the increased risk of spontaneous intracerebral hemorrhage. Recent trials suggest some improvement in global cognitive function in patients with vascular cognitive impairment and dementia with targeted cognitive rehabilitation. SUMMARY Thorough clinical evaluation and neuroimaging form the basis for diagnosis. As vascular cognitive impairment and dementia is the leading nondegenerative cause of dementia, identifying risk factors and optimizing their management is paramount. Once vascular brain injury has occurred, symptomatic management should be offered and secondary prevention pursued.
Collapse
|
21
|
Cerebral small vessel disease alters neurovascular unit regulation of microcirculation integrity involved in vascular cognitive impairment. Neurobiol Dis 2022; 170:105750. [DOI: 10.1016/j.nbd.2022.105750] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/09/2022] [Accepted: 05/08/2022] [Indexed: 12/25/2022] Open
|
22
|
Nguyen ML, Huie EZ, Whitmer RA, George KM, Dugger BN. Neuropathology Studies of Dementia in US Persons other than Non-Hispanic Whites. FREE NEUROPATHOLOGY 2022; 3. [PMID: 35425946 PMCID: PMC9007571 DOI: 10.17879/freeneuropathology-2022-3795] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) and vascular dementia are two of the most prevalent dementias that afflict the aging population in the United States (US). Studies have made great strides in understanding the neuropathology of these diseases; however, many studies are conducted in the context of non-Hispanic whites (NHWs), and few include the rapidly growing underrepresented populations that reside in the US. We sought to characterize current knowledge of the neuropathologic landscape of AD and vascular dementia of the largest growing US minority groups, namely Latinos/Hispanics, Black Americans, and Asian Americans, compared with NHWs being the majority group. It is vital to note these historic categories are social constructs and cultural and social associations may underlie differences. We conducted a literature search utilizing specific criteria to yield neuropathology papers that addressed the demographics and neuropathologies of relevance, then collated the findings into this review. We reveal that while there has been much progress in neuropathological research involving Latinos/Hispanics and Black Americans in the past decade, no cohesive conclusions could be extrapolated from the existing data due to the dearth of minority participants and even smaller amount of information related to the heterogeneity within each minority group, especially Latinos/Hispanics. Furthermore, we reveal an even greater scarcity in neuropathological studies involving Asian Americans, also a very heterogeneous group. We hope the presented findings will illuminate the paucity of minority representation in not just neuropathological research but the field of clinical research overall and serve to inspire clinicians and researchers to help reduce the health disparities underrepresented groups in the US face.
Collapse
Affiliation(s)
- My-le Nguyen
- Department of Pathology and Laboratory Medicine, University of California, Davis
| | - Emily Z Huie
- Department of Pathology and Laboratory Medicine, University of California, Davis
| | - Rachel A Whitmer
- Department of Public Health Sciences, University of California, Davis
| | - Kristen M George
- Department of Public Health Sciences, University of California, Davis
| | - Brittany N Dugger
- Department of Pathology and Laboratory Medicine, University of California, Davis
| |
Collapse
|
23
|
Lima AA, Mridha MF, Das SC, Kabir MM, Islam MR, Watanobe Y. A Comprehensive Survey on the Detection, Classification, and Challenges of Neurological Disorders. BIOLOGY 2022; 11:469. [PMID: 35336842 PMCID: PMC8945195 DOI: 10.3390/biology11030469] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/12/2022] [Accepted: 03/14/2022] [Indexed: 01/19/2023]
Abstract
Neurological disorders (NDs) are becoming more common, posing a concern to pregnant women, parents, healthy infants, and children. Neurological disorders arise in a wide variety of forms, each with its own set of origins, complications, and results. In recent years, the intricacy of brain functionalities has received a better understanding due to neuroimaging modalities, such as magnetic resonance imaging (MRI), magnetoencephalography (MEG), and positron emission tomography (PET), etc. With high-performance computational tools and various machine learning (ML) and deep learning (DL) methods, these modalities have discovered exciting possibilities for identifying and diagnosing neurological disorders. This study follows a computer-aided diagnosis methodology, leading to an overview of pre-processing and feature extraction techniques. The performance of existing ML and DL approaches for detecting NDs is critically reviewed and compared in this article. A comprehensive portion of this study also shows various modalities and disease-specified datasets that detect and records images, signals, and speeches, etc. Limited related works are also summarized on NDs, as this domain has significantly fewer works focused on disease and detection criteria. Some of the standard evaluation metrics are also presented in this study for better result analysis and comparison. This research has also been outlined in a consistent workflow. At the conclusion, a mandatory discussion section has been included to elaborate on open research challenges and directions for future work in this emerging field.
Collapse
Affiliation(s)
- Aklima Akter Lima
- Department of Computer Science and Engineering, Bangladesh University of Business and Technology, Dhaka 1216, Bangladesh; (A.A.L.); (M.F.M.); (S.C.D.); (M.M.K.)
| | - M. Firoz Mridha
- Department of Computer Science and Engineering, Bangladesh University of Business and Technology, Dhaka 1216, Bangladesh; (A.A.L.); (M.F.M.); (S.C.D.); (M.M.K.)
| | - Sujoy Chandra Das
- Department of Computer Science and Engineering, Bangladesh University of Business and Technology, Dhaka 1216, Bangladesh; (A.A.L.); (M.F.M.); (S.C.D.); (M.M.K.)
| | - Muhammad Mohsin Kabir
- Department of Computer Science and Engineering, Bangladesh University of Business and Technology, Dhaka 1216, Bangladesh; (A.A.L.); (M.F.M.); (S.C.D.); (M.M.K.)
| | - Md. Rashedul Islam
- Department of Computer Science and Engineering, University of Asia Pacific, Dhaka 1216, Bangladesh
| | - Yutaka Watanobe
- Department of Computer Science and Engineering, University of Aizu, Aizu-Wakamatsu 965-8580, Japan;
| |
Collapse
|
24
|
Wardlaw JM, Benveniste H, Williams A. Cerebral Vascular Dysfunctions Detected in Human Small Vessel Disease and Implications for Preclinical Studies. Annu Rev Physiol 2022; 84:409-434. [PMID: 34699267 DOI: 10.1146/annurev-physiol-060821-014521] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cerebral small vessel disease (SVD) is highly prevalent and a common cause of ischemic and hemorrhagic stroke and dementia, yet the pathophysiology is poorly understood. Its clinical expression is highly varied, and prognostic implications are frequently overlooked in clinics; thus, treatment is currently confined to vascular risk factor management. Traditionally, SVD is considered the small vessel equivalent of large artery stroke (occlusion, rupture), but data emerging from human neuroimaging and genetic studies refute this, instead showing microvessel endothelial dysfunction impacting on cell-cell interactions and leading to brain damage. These dysfunctions reflect defects that appear to be inherited and secondary to environmental exposures, including vascular risk factors. Interrogation in preclinical models shows consistent and converging molecular and cellular interactions across the endothelial-glial-neural unit that increasingly explain the human macroscopic observations and identify common patterns of pathology despite different triggers. Importantly, these insights may offer new targets for therapeutic intervention focused on restoring endothelial-glial physiology.
Collapse
Affiliation(s)
- Joanna M Wardlaw
- Division of Neuroimaging Sciences, Centre for Clinical Brain Sciences; UK Dementia Research Institute; and Edinburgh Imaging, University of Edinburgh, Edinburgh, United Kingdom;
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Anna Williams
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
25
|
Yu Y, Xia T, Tan Z, Xia H, He S, Sun H, Wang X, Song H, Chen W. A2DS2 Score Combined With Clinical and Neuroimaging Factors Better Predicts Stroke-Associated Pneumonia in Hyperacute Cerebral Infarction. Front Neurol 2022; 13:800614. [PMID: 35185764 PMCID: PMC8855060 DOI: 10.3389/fneur.2022.800614] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/04/2022] [Indexed: 12/01/2022] Open
Abstract
Objective To investigate the predictors of stroke-associated pneumonia (SAP) and poor functional outcome in patients with hyperacute cerebral infarction (HCI) by combining clinical factors, laboratory tests and neuroimaging features. Methods We included 205 patients with HCI from November 2018 to December 2019. The diagnostic criterion for SAP was occurrence within 7 days of the onset of stroke. Poor outcome was defined as a functional outcome based on a 3-months MRS score >3. The relationship of demographic, laboratory and neuroimaging variables with SAP and poor outcome was investigated using univariate and multivariate analyses. Results Fifty seven (27.8%) patients were diagnosed with SAP and 40 (19.5%) developed poor outcomes. A2DS2 score (OR = 1.284; 95% CI: 1.048–1.574; P = 0.016), previous stroke (OR = 2.630; 95% CI: 1.122–6.163; P = 0.026), consciousness (OR = 2.945; 95% CI: 1.514–5.729; P < 0.001), brain atrophy (OR = 1.427; 95% CI: 1.040–1.959; P = 0.028), and core infarct volume (OR = 1.715; 95% CI: 1.163–2.528; P = 0.006) were independently associated with the occurrence of SAP. Therefore, we combined these variables into a new SAP prediction model with the C-statistic of 0.84 (95% CI: 0.78–0.90). Fasting plasma glucose (OR = 1.404; 95% CI: 1.202–1.640; P < 0.001), NIHSS score (OR = 1.088; 95% CI: 1.010–1.172; P = 0.026), previous stroke (OR = 4.333; 95% CI: 1.645–11.418; P = 0.003), SAP (OR = 3.420; 95% CI: 1.332–8.787; P = 0.011), basal ganglia-dilated perivascular spaces (BG-dPVS) (OR = 2.124; 95% CI: 1.313–3.436; P = 0.002), and core infarct volume (OR = 1.680; 95% CI: 1.166–2.420; P = 0.005) were independently associated with poor outcome. The C-statistic of the outcome model was 0.87 (95% CI: 0.81–0.94). Furthermore, the SAP model significantly improved discrimination and net benefit more than the A2DS2 scale, with a C-statistic of 0.76 (95% CI: 0.69–0.83). Conclusions After the addition of neuroimaging features, the models exhibit good differentiation and calibration for the prediction of the occurrence of SAP and the development of poor outcomes in HCI patients. The SAP model could better predict the SAP, representing a helpful and valid tool to obtain a net benefit compared with the A2DS2 scale.
Collapse
Affiliation(s)
- Yaoyao Yu
- Radiology Imaging Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tianyi Xia
- Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Zhouli Tan
- Radiology Imaging Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huwei Xia
- Radiology Imaging Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shenping He
- Radiology Imaging Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Han Sun
- Radiology Imaging Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xifan Wang
- Radiology Imaging Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haolan Song
- Radiology Imaging Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weijian Chen
- Radiology Imaging Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Weijian Chen
| |
Collapse
|
26
|
Zellner A, Müller SA, Lindner B, Beaufort N, Rozemuller AJM, Arzberger T, Gassen NC, Lichtenthaler SF, Kuster B, Haffner C, Dichgans M. Proteomic profiling in cerebral amyloid angiopathy reveals an overlap with CADASIL highlighting accumulation of HTRA1 and its substrates. Acta Neuropathol Commun 2022; 10:6. [PMID: 35074002 PMCID: PMC8785498 DOI: 10.1186/s40478-021-01303-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) is an age-related condition and a major cause of intracerebral hemorrhage and cognitive decline that shows close links with Alzheimer's disease (AD). CAA is characterized by the aggregation of amyloid-β (Aβ) peptides and formation of Aβ deposits in the brain vasculature resulting in a disruption of the angioarchitecture. Capillaries are a critical site of Aβ pathology in CAA type 1 and become dysfunctional during disease progression. Here, applying an advanced protocol for the isolation of parenchymal microvessels from post-mortem brain tissue combined with liquid chromatography tandem mass spectrometry (LC-MS/MS), we determined the proteomes of CAA type 1 cases (n = 12) including a patient with hereditary cerebral hemorrhage with amyloidosis-Dutch type (HCHWA-D), and of AD cases without microvascular amyloid pathology (n = 13) in comparison to neurologically healthy controls (n = 12). ELISA measurements revealed microvascular Aβ1-40 levels to be exclusively enriched in CAA samples (mean: > 3000-fold compared to controls). The proteomic profile of CAA type 1 was characterized by massive enrichment of multiple predominantly secreted proteins and showed significant overlap with the recently reported brain microvascular proteome of patients with cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), a hereditary cerebral small vessel disease (SVD) characterized by the aggregation of the Notch3 extracellular domain. We found this overlap to be largely attributable to the accumulation of high-temperature requirement protein A1 (HTRA1), a serine protease with an established role in the brain vasculature, and several of its substrates. Notably, this signature was not present in AD cases. We further show that HTRA1 co-localizes with Aβ deposits in brain capillaries from CAA type 1 patients indicating a pathologic recruitment process. Together, these findings suggest a central role of HTRA1-dependent protein homeostasis in the CAA microvasculature and a molecular connection between multiple types of brain microvascular disease.
Collapse
Affiliation(s)
- Andreas Zellner
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Barbara Lindner
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Nathalie Beaufort
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Annemieke J M Rozemuller
- Department of Pathology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Thomas Arzberger
- Department of Psychiatry and Psychotherapy, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Nils C Gassen
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
| | - Christof Haffner
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany.
- Department of Psychiatry and Psychotherapy, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany.
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
27
|
Shakir MN, Dugger BN. Advances in Deep Neuropathological Phenotyping of Alzheimer Disease: Past, Present, and Future. J Neuropathol Exp Neurol 2022; 81:2-15. [PMID: 34981115 PMCID: PMC8825756 DOI: 10.1093/jnen/nlab122] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Alzheimer disease (AD) is a neurodegenerative disorder characterized pathologically by the presence of neurofibrillary tangles and amyloid beta (Aβ) plaques in the brain. The disease was first described in 1906 by Alois Alzheimer, and since then, there have been many advancements in technologies that have aided in unlocking the secrets of this devastating disease. Such advancements include improving microscopy and staining techniques, refining diagnostic criteria for the disease, and increased appreciation for disease heterogeneity both in neuroanatomic location of abnormalities as well as overlap with other brain diseases; for example, Lewy body disease and vascular dementia. Despite numerous advancements, there is still much to achieve as there is not a cure for AD and postmortem histological analyses is still the gold standard for appreciating AD neuropathologic changes. Recent technological advances such as in-vivo biomarkers and machine learning algorithms permit great strides in disease understanding, and pave the way for potential new therapies and precision medicine approaches. Here, we review the history of human AD neuropathology research to include the notable advancements in understanding common co-pathologies in the setting of AD, and microscopy and staining methods. We also discuss future approaches with a specific focus on deep phenotyping using machine learning.
Collapse
Affiliation(s)
- Mustafa N Shakir
- From the Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, California, USA (MNS, BND)
| | - Brittany N Dugger
- From the Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, California, USA (MNS, BND)
| |
Collapse
|
28
|
Canavero I, Rifino N, Montano V, Pantoni L, Gatti L, Pollaci G, Potenza A, Carrozzini T, Finsterer J, Bersano A. Cognitive aspects of MELAS and CARASAL. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2022; 3:100139. [PMID: 36324419 PMCID: PMC9616374 DOI: 10.1016/j.cccb.2022.100139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 11/18/2022]
Abstract
MELAS and CARASAL have been associated with clinical evidence of cognitive impairment and should be considered as possible causes of early onset Vascular Dementia (VaD), particularly in patients with a familial history of dementia or cerebrovascular disease. Cognitive deterioration in MELAS involves executive function, attention, language, memory, visuospatial, and motor functioning and may correlate with the total Stroke-like episodes (SLEs) lesion load. CARASIL is characterized by late and slow cognition disorders, involving episodic memory, executive functions and facial recognition.
Monogenic diseases, although rare, should be always considered in the diagnostic work up of vascular dementia (VaD), particularly in patients with early onset and a familial history of dementia or cerebrovascular disease. They include, other than CADASIL, Fabry disease, Col4A1-A2 related disorders, which are well recognized causes of VaD, other heritable diseases such as mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) and cathepsin-A related arteriopathy strokes and leukoencephalopathy (CARASAL). MELAS, caused by mtDNA (80% of adult cases m.3243A>G mutations) and more rarely POLG1 mutations, has minimum prevalence of 3.5/100,000. CARASAL, which is caused by mutations in the CTSA gene, has been described in about 19 patients so far. In both these two disorders cognitive features have not been fully explored and are described only in case series or families. This review paper is aimed at providing an update on the clinical manifestations, with particular focus on cognitive aspects, but also neuroradiological and genetic features of these less frequent monogenic diseases associated with VaD.
Collapse
Affiliation(s)
- I Canavero
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - N Rifino
- University of Milano-Bicocca, Milan, Italy
| | - V Montano
- Department of Clinical and Experimental Medicine, Neurological Institute, University of Pisa, Italy
| | - L Pantoni
- Luigi Sacco Department of Biomedical and Clinical Sciences, Stroke and Dementia Lab, University of Milan, Italy
| | - L Gatti
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - G Pollaci
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - A Potenza
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - T Carrozzini
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - J Finsterer
- Krankenanstalt Rudolfstiftung, Messerli Institute, Vienna, Austria
| | - A Bersano
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- Corresponding author.
| |
Collapse
|
29
|
Mossanen Parsi M, Duval C, Ariëns RAS. Vascular Dementia and Crosstalk Between the Complement and Coagulation Systems. Front Cardiovasc Med 2021; 8:803169. [PMID: 35004913 PMCID: PMC8733168 DOI: 10.3389/fcvm.2021.803169] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 11/29/2021] [Indexed: 01/12/2023] Open
Abstract
Vascular Dementia (VaD) is a neurocognitive disorder caused by reduced blood flow to the brain tissue, resulting in infarction, and is the second most common type of dementia. The complement and coagulation systems are evolutionary host defence mechanisms activated by acute tissue injury to induce inflammation, clot formation and lysis; recent studies have revealed that these systems are closely interlinked. Overactivation of these systems has been recognised to play a key role in the pathogenesis of neurological disorders such as Alzheimer's disease and multiple sclerosis, however their role in VaD has not yet been extensively reviewed. This review aims to bridge the gap in knowledge by collating current understanding of VaD to enable identification of complement and coagulation components involved in the pathogenesis of this disorder that may have their effects amplified or supressed by crosstalk. Exploration of these mechanisms may unveil novel therapeutic targets or biomarkers that would improve current treatment strategies for VaD.
Collapse
Affiliation(s)
| | | | - Robert A. S. Ariëns
- Discovery and Translational Science Department, School of Medicine, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
30
|
Fan Y, Xu Y, Shen M, Guo H, Zhang Z. Total Cerebral Small Vessel Disease Burden on MRI Correlates With Cognitive Impairment in Outpatients With Amnestic Disorders. Front Neurol 2021; 12:747115. [PMID: 34925212 PMCID: PMC8675386 DOI: 10.3389/fneur.2021.747115] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/26/2021] [Indexed: 01/04/2023] Open
Abstract
Objectives: The main markers of cerebral small vessel disease (cSVD) on MRI may be entered into a scoring system, with the total score representing the overall burden of cSVD. An association between total cSVD score and cognitive dysfunction has been reported in several cohorts. The present study aimed to investigate this association in outpatients with amnestic disorders. Materials and Methods: Outpatients with amnestic complaints in a memory clinic (n = 289) were recruited retrospectively. All the patients had undergone clinical and cognitive evaluation at first presentation. Cognitive function was assessed by Montreal Cognitive Assessment (MoCA) scale. The total cSVD score was based on the following markers on MRI: lacune; white matter hyperintensities, microbleed, and enlarged perivascular spaces. The association between total cSVD score and MoCA score was tested via Spearman's analysis and a linear regression model. Results: Among the 289 patients, rates for 0–4 cSVD markers respectively ranged from 30.4 to 2.8%. A multiple linear regression model revealed an inverse correlation between the total cSVD score and MoCA score. The association remained significant after adjusting for gender, age, education, levels of medial temporal lobe atrophy, and classical vascular risk factors [β = −0.729, 95% CI (−1.244, −0.213); P = 0.006]. When individual markers were individually analyzed after adjusting for the same factors, only microbleed associated with MoCA score [β = −3.007, 95% CI (−4.533, −1.480), P < 0.001]. Conclusions: A significant association was demonstrated between total cSVD score and cognitive performance in the outpatients with amnestic disorders.
Collapse
Affiliation(s)
- Yangyi Fan
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Yicheng Xu
- Department of Neurology, Aerospace Center Hospital, Beijing, China
| | - Ming Shen
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Huailian Guo
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Zhaoxu Zhang
- Department of Neurology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
31
|
Neuroprotective Potential of Carnosine in Cerebrovascular Diseases. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10342-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
32
|
Yang K, Zeng L, Ge A, Cao C, Zhang H, Bao T, Yi Y, Ge J. Systems Biology and Chemoinformatics-Based Strategies to Explore the Biological Mechanism of Fugui Wenyang Decoction in Treating Vascular Dementia Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6693955. [PMID: 34659639 PMCID: PMC8517630 DOI: 10.1155/2021/6693955] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 08/24/2021] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To explore the biological mechanism of Fugui Wenyang Decoction (FGWYD) in treating vascular dementia (VD) rats based on systems pharmacology, proteomics, and a multidirectional pharmacology integration strategy. METHODS Chemoinformatics was utilized to construct and analyze the FGWYD-VD protein-protein interaction (PPI) network. Then, the total protein in the brain tissue of the infarcted side of the rat was extracted for protein identification, pattern identification, and protein quantitative analysis. The differentially expressed proteins are analyzed by bioinformatics. Finally, the important proteins in the oxidative stress-related biological process proteins and indicators were detected through experimental pharmacology to verify the findings of systems biology and chemoinformatics. RESULTS There were a total of 73 FGWYD components with 245 FGWYD and 145 VD genes. The results of GO enrichment analysis and pathway enrichment analysis showed that MBHD may regulate the inflammation module, oxidative stress, the synaptic plasticity regulation module, and the neuronal apoptosis section module. Compared with the sham operation group, there were 23 upregulated proteins and 17 downregulated proteins in the model group (P < 0.05). Compared with the model group, there were 16 upregulated proteins and 10 downregulated proteins in the FGWYD group (P < 0.05). Bioinformatics analysis shows that those proteins were closely related to processes such as inflammation, oxidative stress, neuronal apoptosis, neuronal growth and differentiation, signaling pathways, and transcriptional regulation. Multidirectional pharmacology further verified the neuroprotective mechanism of the Nrf2/HO-1 pathway in FGWYD treatment of VD. CONCLUSION The mechanism of FGWYD in the treatment of VD may be related to inflammation, oxidative stress, angiogenesis, and neuronal apoptosis.
Collapse
Affiliation(s)
- Kailin Yang
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Liuting Zeng
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Anqi Ge
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Chuandong Cao
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Haiyan Zhang
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Tingting Bao
- Beijing University of Chinese Medicine, Beijing, China
| | - Yaqiao Yi
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Jinwen Ge
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| |
Collapse
|
33
|
Lee R, Choi H, Park KY, Kim JM, Seok JW. Prediction of post-stroke cognitive impairment using brain FDG PET: deep learning-based approach. Eur J Nucl Med Mol Imaging 2021; 49:1254-1262. [PMID: 34599654 DOI: 10.1007/s00259-021-05556-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/04/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE Post-stroke cognitive impairment can affect up to one third of stroke survivors. Since cognitive function greatly contributes to patients' quality of life, an objective quantitative biomarker for early prediction of dementia after stroke is required. We developed a deep-learning (DL)-based signature using positron emission tomography (PET) to objectively evaluate cognitive decline in patients with stroke. METHODS We built a DL model that differentiated Alzheimer's disease (AD) from normal controls (NC) using brain fluorodeoxyglucose (FDG) PET from the Alzheimer's Disease Neuroimaging Initiative database. The model was directly transferred to a prospectively enrolled cohort of patients with stroke to differentiate patients with dementia from those without dementia. The accuracy of the model was evaluated by the area under the curve values of receiver operating characteristic curves (AUC-ROC). We visualized the distribution of DL-based features and brain regions that the model weighted for classification. Correlations between cognitive signature from the DL model and clinical variables were evaluated, and survival analysis for post-stroke dementia was performed in patients with stroke. RESULTS The classification of AD vs. NC subjects was performed with AUC-ROC of 0.94 (95% confidence interval [CI], 0.89-0.98). The transferred model discriminated stroke patients with dementia (AUC-ROC = 0.75). The score of cognitive decline signature using FDG PET was positively correlated with age, neutrophil-lymphocyte ratio and platelet-lymphocyte ratio and negatively correlated with body mass index in patients with stroke. We found that the cognitive decline score was an independent risk factor for dementia following stroke (hazard ratio, 10.90; 95% CI, 3.59-33.09; P < 0.0001) after adjustment for other key variables. CONCLUSION The DL-based cognitive signature using FDG PET was successfully transferred to an independent stroke cohort. It is suggested that DL-based cognitive evaluation using FDG PET could be utilized as an objective biomarker for cognitive dysfunction in patients with cerebrovascular diseases.
Collapse
Affiliation(s)
- Reeree Lee
- Department of Nuclear Medicine, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 224-1, Heukseok-dong, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Hongyoon Choi
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kwang-Yeol Park
- Department of Neurology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 224-1, Heukseok-dong, Dongjak-gu, Seoul, 06974, Republic of Korea.
| | - Jeong-Min Kim
- Department of Neurology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ju Won Seok
- Department of Nuclear Medicine, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 224-1, Heukseok-dong, Dongjak-gu, Seoul, 06974, Republic of Korea.
| |
Collapse
|
34
|
Bir SC, Khan MW, Javalkar V, Toledo EG, Kelley RE. Emerging Concepts in Vascular Dementia: A Review. J Stroke Cerebrovasc Dis 2021; 30:105864. [PMID: 34062312 DOI: 10.1016/j.jstrokecerebrovasdis.2021.105864] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/29/2021] [Accepted: 04/28/2021] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVE Vascular dementia (VaD) is the second most common cause of dementia and a major health concern worldwide. A comprehensive review on VaD is warranted for better understanding and guidance for the practitioner. We provide an updated overview of the epidemiology, pathophysiological mechanisms, neuroimaging patterns as well as current diagnostic and therapeutic approaches. MATERIALS AND METHODS A narrative review of current literature in VaD was performed based on publications from the database of PubMed, Scopus and Google Scholar up to January, 2021. RESULTS VaD can be the result of ischemic or hemorrhagic tissue injury in a particular region of the brain which translates into clinically significant cognitive impairment. For example, a cerebral infarct in the speech area of the dominant hemisphere would translate into clinically significant impairment as would involvement of projection pathways such as the arcuate fasciculus. Specific involvement of the angular gyrus of the dominant hemisphere, with resultant Gerstman's syndrome, could have a pronounced effect on functional ability despite being termed a "minor stroke". Small vessel cerebrovascular disease can have a cumulate effect on cognitive function over time. It is unfortunately well recognized that "good" functional recovery in acute ischemic or haemorrhagic stroke, including subarachnoid haemorrhage, does not necessarily translate into good cognitive recovery. The victim may often be left unable to have gainful employment, drive a car safely or handle their affairs independently. CONCLUSIONS This review should serve as a compendium of updated information on VaD and provide guidance in terms of newer diagnostic and potential therapeutic approaches.
Collapse
Affiliation(s)
- Shyamal C Bir
- Department of Neurology Ocshner/LSU Health Sciences Center-Sheveport, Shreveport, LA, USA
| | - Muhammad W Khan
- Department of Neurology Ocshner/LSU Health Sciences Center-Sheveport, Shreveport, LA, USA
| | - Vijayakumar Javalkar
- Department of Neurology Ocshner/LSU Health Sciences Center-Sheveport, Shreveport, LA, USA
| | | | - Roger E Kelley
- Department of Neurology Ocshner/LSU Health Sciences Center-Sheveport, Shreveport, LA, USA.
| |
Collapse
|
35
|
Tabeeva GR. [Headache and cerebrovascular diseases]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:114-121. [PMID: 33728860 DOI: 10.17116/jnevro2021121021114] [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: 11/17/2022]
Abstract
Headache is a common symptom of acute and chronic cerebrovascular diseases. Headache can be symptomatic in patients with various forms of vascular pathology of the brain but primary headaches are much more common. Secondary headaches in acute cerebrovascular accidents may be the first symptom, and in some cases, a risk factor or complication of stroke. In chronic cerebrovascular diseases, headache may be the predominant symptom in the early stages and resolve in the later stages of the disease. At the same time, the severity, nature and course of headache cannot be considered as reliable signs of cerebrovascular disease. Meanwhile, the verification of the headache form is important from the point of view of determining the priorities of diagnosis and therapy.
Collapse
Affiliation(s)
- G R Tabeeva
- Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
36
|
Al-Baradie RS, Pu S, Liu D, Zeinolabediny Y, Ferris G, Sanfeli C, Corpas R, Garcia-Lara E, Alsagaby SA, Alshehri BM, Abdel-Hadi AM, Ahmad F, Moatari P, Heidari N, Slevin M. Monomeric C-Reactive Protein Localized in the Cerebral Tissue of Damaged Vascular Brain Regions Is Associated With Neuro-Inflammation and Neurodegeneration-An Immunohistochemical Study. Front Immunol 2021; 12:644213. [PMID: 33796111 PMCID: PMC8007856 DOI: 10.3389/fimmu.2021.644213] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 02/22/2021] [Indexed: 11/13/2022] Open
Abstract
Monomeric C-reactive protein (mCRP) is now accepted as having a key role in modulating inflammation and in particular, has been strongly associated with atherosclerotic arterial plaque progression and instability and neuroinflammation after stroke where a build-up of the mCRP protein within the brain parenchyma appears to be connected to vascular damage, neurodegenerative pathophysiology and possibly Alzheimer's Disease (AD) and dementia. Here, using immunohistochemical analysis, we wanted to confirm mCRP localization and overall distribution within a cohort of AD patients showing evidence of previous infarction and then focus on its co-localization with inflammatory active regions in order to provide further evidence of its functional and direct impact. We showed that mCRP was particularly seen in large amounts within brain vessels of all sizes and that the immediate micro-environment surrounding these had become laden with mCRP positive cells and extra cellular matrix. This suggested possible leakage and transport into the local tissue. The mCRP-positive regions were almost always associated with neurodegenerative, damaged tissue as hallmarked by co-positivity with pTau and β-amyloid staining. Where this occurred, cells with the morphology of neurons, macrophages and glia, as well as smaller microvessels became mCRP-positive in regions staining for the inflammatory markers CD68 (macrophage), interleukin-1 beta (IL-1β) and nuclear factor kappa B (NFκB), showing evidence of a perpetuation of inflammation. Positive staining for mCRP was seen even in distant hypothalamic regions. In conclusion, brain injury or inflammatory neurodegenerative processes are strongly associated with mCRP localization within the tissue and given our knowledge of its biological properties, it is likely that this protein plays a direct role in promoting tissue damage and supporting progression of AD after injury.
Collapse
Affiliation(s)
- Raid S Al-Baradie
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majma'ah, Saudi Arabia
| | - Shuang Pu
- National Natural Science foundation of China, Beijing, China.,School of Healthcare Science, John Dalton Building, Manchester Metropolitan University, Manchester, United Kingdom
| | - Donghui Liu
- School of Healthcare Science, John Dalton Building, Manchester Metropolitan University, Manchester, United Kingdom
| | - Yasmin Zeinolabediny
- School of Healthcare Science, John Dalton Building, Manchester Metropolitan University, Manchester, United Kingdom
| | - Glenn Ferris
- School of Healthcare Science, John Dalton Building, Manchester Metropolitan University, Manchester, United Kingdom
| | - Coral Sanfeli
- Instituto De Investigaciones Biomedicas De Barcelona, CSIC, Barcelona, Spain
| | - Ruben Corpas
- Instituto De Investigaciones Biomedicas De Barcelona, CSIC, Barcelona, Spain
| | - Elisa Garcia-Lara
- Department of Physical Therapy and Health Rehabilitation, College of Applied Medical Sciences, Majmaah University, Al Majma'ah, Saudi Arabia
| | - Suliman A Alsagaby
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majma'ah, Saudi Arabia
| | - Bader M Alshehri
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majma'ah, Saudi Arabia
| | - Ahmed M Abdel-Hadi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majma'ah, Saudi Arabia
| | - Fuzail Ahmad
- Department of Physical Therapy and Health Rehabilitation, College of Applied Medical Sciences, Majmaah University, Al Majma'ah, Saudi Arabia
| | - Psalm Moatari
- Salford Royal NHS Foundation Trust, Manchester, United Kingdom
| | | | - Mark Slevin
- National Natural Science foundation of China, Beijing, China.,University of Medicine, Pharmacy, Science and Technology, Târgu Mures, Romania
| |
Collapse
|
37
|
Vinters HV, Magaki SD, Williams CK. Neuropathologic Findings in Chronic Kidney Disease (CKD). J Stroke Cerebrovasc Dis 2021; 30:105657. [PMID: 33579545 DOI: 10.1016/j.jstrokecerebrovasdis.2021.105657] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/30/2020] [Accepted: 01/31/2021] [Indexed: 12/24/2022] Open
Abstract
Studying the neuropathologic autopsy findings in subjects with chronic kidney disease (CKD) or chronic renal failure (CRF) is difficult for several reasons: etiology of the CKD may be heterogeneous, affected patients may have one or more major co-morbidities that themselves can cause significant neurologic disease, and agonal events may result in significant findings that were of minimal significance earlier in a patient's life. We studied the constellation of neuropathologic abnormalities in 40 autopsy brains originating from subjects of ages 34-95 years (no children in the study). The most common pathologic change was that of ischemic infarcts (cystic, lacunar and/or microinfarcts), which were seen in over half of subjects. These were associated with both large artery atherosclerosis and arteriolosclerosis (A/S), the latter finding being present in 29/40 subjects. Charcot-Bouchard microaneurysms were present in the brains of three subjects, in one case associated with severe amyloid angiopathy. Microvascular calcinosis (medial sclerosis in the case of arterioles) was seen in the basal ganglia (n=8) and/or endplate region of the hippocampus (n=7) and occasional ischemic infarcts in one brain showed severe calcification. Sequelae of cerebrovascular disease (especially A/S or microvascular disease) are a common neuropathologic substrate for neurologic disability and brain lesions in this complex group of patients. Regulation of calcium metabolism within brain microvessel walls may be worthy of further research in both human brain specimens and animal models.
Collapse
Affiliation(s)
- Harry V Vinters
- Department of Pathology & Laboratory Medicine; Department of Neurology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, 90095-1732, United States.
| | | | | |
Collapse
|
38
|
Blevins BL, Vinters HV, Love S, Wilcock DM, Grinberg LT, Schneider JA, Kalaria RN, Katsumata Y, Gold BT, Wang DJJ, Ma SJ, Shade LMP, Fardo DW, Hartz AMS, Jicha GA, Nelson KB, Magaki SD, Schmitt FA, Teylan MA, Ighodaro ET, Phe P, Abner EL, Cykowski MD, Van Eldik LJ, Nelson PT. Brain arteriolosclerosis. Acta Neuropathol 2021; 141:1-24. [PMID: 33098484 PMCID: PMC8503820 DOI: 10.1007/s00401-020-02235-6] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/04/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022]
Abstract
Brain arteriolosclerosis (B-ASC), characterized by pathologic arteriolar wall thickening, is a common finding at autopsy in aged persons and is associated with cognitive impairment. Hypertension and diabetes are widely recognized as risk factors for B-ASC. Recent research indicates other and more complex risk factors and pathogenetic mechanisms. Here, we describe aspects of the unique architecture of brain arterioles, histomorphologic features of B-ASC, relevant neuroimaging findings, epidemiology and association with aging, established genetic risk factors, and the co-occurrence of B-ASC with other neuropathologic conditions such as Alzheimer's disease and limbic-predominant age-related TDP-43 encephalopathy (LATE). There may also be complex physiologic interactions between metabolic syndrome (e.g., hypertension and inflammation) and brain arteriolar pathology. Although there is no universally applied diagnostic methodology, several classification schemes and neuroimaging techniques are used to diagnose and categorize cerebral small vessel disease pathologies that include B-ASC, microinfarcts, microbleeds, lacunar infarcts, and cerebral amyloid angiopathy (CAA). In clinical-pathologic studies that factored in comorbid diseases, B-ASC was independently associated with impairments of global cognition, episodic memory, working memory, and perceptual speed, and has been linked to autonomic dysfunction and motor symptoms including parkinsonism. We conclude by discussing critical knowledge gaps related to B-ASC and suggest that there are probably subcategories of B-ASC that differ in pathogenesis. Observed in over 80% of autopsied individuals beyond 80 years of age, B-ASC is a complex and under-studied contributor to neurologic disability.
Collapse
Affiliation(s)
- Brittney L Blevins
- Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Harry V Vinters
- Department of Pathology and Laboratory Medicine, David Geffen SOM at UCLA and Ronald Reagan UCLA Medical Center, Los Angeles, CA, 90095-1732, USA
| | - Seth Love
- University of Bristol and Southmead Hospital, Bristol, BS10 5NB, UK
| | - Donna M Wilcock
- Sanders-Brown Center on Aging, Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Lea T Grinberg
- Department of Neurology and Pathology, UCSF, San Francisco, CA, USA
- Global Brain Health Institute, UCSF, San Francisco, CA, USA
- LIM-22, Department of Pathology, University of Sao Paulo Medical School, São Paulo, Brazil
| | - Julie A Schneider
- Departments of Neurology and Pathology, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Rajesh N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging, Department of Biostatistics, University Kentucky, Lexington, KY, 40536, USA
| | - Brian T Gold
- Sanders-Brown Center on Aging, Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Danny J J Wang
- Laboratory of FMRI Technology (LOFT), USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Samantha J Ma
- Laboratory of FMRI Technology (LOFT), USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Lincoln M P Shade
- Sanders-Brown Center on Aging, Department of Biostatistics, University Kentucky, Lexington, KY, 40536, USA
| | - David W Fardo
- Sanders-Brown Center on Aging, Department of Biostatistics, University Kentucky, Lexington, KY, 40536, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, University Kentucky, Lexington, KY, 40536, USA
| | - Gregory A Jicha
- Sanders-Brown Center on Aging, Department of Neurology, University Kentucky, Lexington, KY, 40536, USA
| | | | - Shino D Magaki
- Department of Pathology and Laboratory Medicine, David Geffen SOM at UCLA and Ronald Reagan UCLA Medical Center, Los Angeles, CA, 90095-1732, USA
| | - Frederick A Schmitt
- Sanders-Brown Center on Aging, Department of Neurology, University Kentucky, Lexington, KY, 40536, USA
| | - Merilee A Teylan
- Department of Epidemiology, University Washington, Seattle, WA, 98105, USA
| | | | - Panhavuth Phe
- Sanders-Brown Center on Aging, University Kentucky, Lexington, KY, 40536, USA
| | - Erin L Abner
- Sanders-Brown Center on Aging, Department of Epidemiology, University Kentucky, Lexington, KY, 40536, USA
| | - Matthew D Cykowski
- Departments of Pathology and Genomic Medicine and Neurology, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, Department of Pathology, University of Kentucky, Lexington, KY, 40536, USA.
- Rm 311 Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone Avenue, Lexington, KY, 40536, USA.
| |
Collapse
|
39
|
Marzoughi S, Banerjee A, Jutzeler CR, Prado MAM, Rosner J, Cragg JJ, Cashman N. Tardive neurotoxicity of anticholinergic drugs: A review. J Neurochem 2020; 158:1334-1344. [PMID: 33222198 DOI: 10.1111/jnc.15244] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/15/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022]
Abstract
The cholinergic system is a complex neurotransmitter system with functional involvement at multiple levels of the nervous system including the cerebral cortex, spinal cord, autonomic nervous system, and neuromuscular junction. Anticholinergic medications are among the most prescribed medications, making up one-third to one-half of all medications prescribed for seniors. Recent evidence has linked long-term use of anticholinergic medications and dementia. Emerging evidence implicates the cholinergic system in the regulation of cerebral vasculature as well as neuroinflammation, suggesting that anticholinergic medications may contribute to absolute risk and progression of neurodegenerative diseases. In this review, we explore the involvement of the cholinergic system in various neurodegenerative diseases and the possible detrimental effects of anticholinergic medications on the onset and progression of these disorders. We identified references by searching the PubMed and Cochrane database between January 1990 and September 2019 for English-language animal and human studies including randomized clinical trials (RCTs), meta-analyses, systematic reviews, and observational studies. In addition, we conducted a manual search of reference lists from retrieved studies. Long-term anticholinergic medication exposure may have detrimental consequences beyond well-documented short-term cognitive effects, through a variety of mechanisms either directly impacting cholinergic neurotransmission or through receptors expressed on the vasculature or immune cells, providing a pathophysiological framework for complex interactions across the entire neuroaxis.
Collapse
Affiliation(s)
- Sina Marzoughi
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Ankur Banerjee
- Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Catherine R Jutzeler
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Marco A M Prado
- Robarts Research Institute, The University of Western Ontario, London, ON, Canada
| | - Jan Rosner
- Collaboration for Outcomes Research and Evaluation (CORE), Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jacquelyn J Cragg
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Neil Cashman
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
40
|
Zhang X, Trebak F, Souza LAC, Shi J, Zhou T, Kehoe PG, Chen Q, Feng Earley Y. Small RNA modifications in Alzheimer's disease. Neurobiol Dis 2020; 145:105058. [PMID: 32835860 PMCID: PMC7572745 DOI: 10.1016/j.nbd.2020.105058] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/11/2020] [Accepted: 08/18/2020] [Indexed: 12/12/2022] Open
Abstract
Background While significant advances have been made in uncovering the aetiology of Alzheimer's disease and related dementias at the genetic level, molecular events at the epigenetic level remain largely undefined. Emerging evidence indicates that small non-coding RNAs (sncRNAs) and their associated RNA modifications are important regulators of complex physiological and pathological processes, including aging, stress responses, and epigenetic inheritance. However, whether small RNAs and their modifications are altered in dementia is not known. Methods We performed LC-MS/MS-based, high-throughput assays of small RNA modifications in post-mortem samples of the prefrontal lobe cortices of Alzheimer's disease (AD) and control individuals. We noted that some of the AD patients has co-occurring vascular cognitive impairment-related pathology (VaD). Findings We report altered small RNA modifications in AD samples compared with normal controls. The 15-25-nucleotide (nt) RNA fraction of these samples was enriched for microRNAs, whereas the 30-40-nt RNA fraction was enriched for tRNA-derived small RNAs (tsRNAs), rRNA-derived small RNAs (rsRNAs), and YRNA-derived small RNAs (ysRNAs). Interestingly, most of these altered RNA modifications were detected both in the AD and AD with co-occurring vascular dementia subjects. In addition, sequencing of small RNA in the 30-40-nt fraction from AD cortices revealed reductions in rsRNA-5S, tsRNA-Tyr, and tsRNA-Arg. Interpretation These data suggest that sncRNAs and their associated modifications are novel signals that may be linked to the pathogenesis and development of Alzheimer's disease. Fund NIH grants (R01HL122770, R01HL091905, 1P20GM130459, R01HD092431, P50HD098593, GM103440), AHA grant (17IRG33370128), Sigmund Gestetner Foundation Fellowship to P Kehoe.
Collapse
Affiliation(s)
- Xudong Zhang
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, USA
| | - Fatima Trebak
- Departments of Pharmacology, Physiology & Cell Biology, Center for Molecular & Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno, School of Medicine, Reno, NV, USA
| | - Lucas A C Souza
- Departments of Pharmacology, Physiology & Cell Biology, Center for Molecular & Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno, School of Medicine, Reno, NV, USA
| | - Junchao Shi
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, USA
| | - Tong Zhou
- Departments of Physiology & Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, USA
| | - Patrick G Kehoe
- Dementia Research Group, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Qi Chen
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, USA.
| | - Yumei Feng Earley
- Departments of Pharmacology, Physiology & Cell Biology, Center for Molecular & Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno, School of Medicine, Reno, NV, USA.
| |
Collapse
|
41
|
Choi HI, Ryu CW, Kim S, Rhee HY, Jahng GH. Changes in Microvascular Morphology in Subcortical Vascular Dementia: A Study of Vessel Size Magnetic Resonance Imaging. Front Neurol 2020; 11:545450. [PMID: 33192974 PMCID: PMC7658467 DOI: 10.3389/fneur.2020.545450] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/23/2020] [Indexed: 12/22/2022] Open
Abstract
Background: Cerebral small vessel disease is the most common cause of subcortical vascular dementia (SVaD). Unfortunately, conventional imaging techniques do not always demonstrate the microvascular pathology that is associated with small vessel disease. The purpose of this study was to evaluate the changes in the microvascular structure of SVaD and to identify how the microvascular changes in vessel size, detected with imaging, affect the gray matter. Methods: Ten SVaD patients and 12 healthy controls underwent vessel size imaging with gradient-echo and spin-echo sequences before and after contrast agent injection. Four microvessel index maps, including total blood volume fraction (BVf), mean vessel density (Q), mean vessel diameter (mVD), and vessel size index (VSI) were calculated. ROI value of each microvessel parameter was compared between SVaD patients and controls. Voxel-wise comparison of microvessel parameters was also performed to assess the regional difference. The relationship between the microvessel parameters in white matter and total gray matter volume (TGV) were assessed. Results: Both mVD and VSI were significantly different between the SVaD and controls in the ROI-based comparisons (unpaired t-test, p < 0.05). mVD and VSI were significantly increased in the SVaD group at the subcortical, periventricular white matter, basal ganglia, and thalami compared with the controls (FDR corrected, p < 0.05). VSI in the white matter areas were significantly negatively correlated with TGV (r = −0.446, p < 0.05). Conclusions: The increase of mVD and VSI in SVaD patients reflects the damage of the microvessels in the white matter, and these changes may lead to the damage of the gray matter.
Collapse
Affiliation(s)
- Hyeon-Il Choi
- Department of Radiology, Neurology, Kyung Hee University Hospital at Gangdong, Seoul, South Korea
| | - Chang-Woo Ryu
- Department of Radiology, Neurology, Kyung Hee University Hospital at Gangdong, Seoul, South Korea.,College of Medicine, Kyung Hee University, Seoul, South Korea
| | - Songvin Kim
- Department of Radiology, Neurology, Kyung Hee University Hospital at Gangdong, Seoul, South Korea
| | - Hak Young Rhee
- Department of Neurology, Kyung Hee University Hospital at Gangdong, Seoul, South Korea
| | - Geon-Ho Jahng
- Department of Radiology, Neurology, Kyung Hee University Hospital at Gangdong, Seoul, South Korea
| |
Collapse
|
42
|
Abstract
ABSTRACT Cerebral small vessel disease (SVD) is a common global brain disease that causes cognitive impairment, ischemic or hemorrhagic stroke, problems with mobility, and neuropsychiatric symptoms. The brain damage, seen as focal white and deep grey matter lesions on brain magnetic resonance imaging (MRI) or computed tomography (CT), typically accumulates "covertly" and may reach an advanced state before being detected incidentally on brain scanning or causing symptoms. Patients have typically presented to different clinical services or been recruited into research focused on one clinical manifestation, perhaps explaining a lack of awareness, until recently, of the full range and complexity of SVD.In this review, we discuss the varied clinical presentations, established and emerging risk factors, relationship to SVD features on MRI or CT, and the current state of knowledge on the effectiveness of a wide range of pharmacological and lifestyle interventions. The core message is that effective assessment and clinical management of patients with SVD, as well as future advances in diagnosis, care, and treatment, will require a more "joined-up"' approach. This approach should integrate clinical expertise in stroke neurology, cognitive, and physical dysfunctions. It requires more clinical trials in order to improve pharmacological interventions, lifestyle and dietary modifications. A deeper understanding of the pathophysiology of SVD is required to steer the identification of novel interventions. An essential prerequisite to accelerating clinical trials is to improve the consistency, and standardization of clinical, cognitive and neuroimaging endpoints.
Collapse
|
43
|
Langer C, Adukauskaite A, Plank F, Feuchtner G, Cartes-Zumelzu F. Cerebral Autosomal Dominant Arteriopathy (CADASIL) with cardiac involvement (ANOCA) and subcortical leukencephalopathy. J Cardiovasc Comput Tomogr 2020; 14:e1-e6. [PMID: 30197288 DOI: 10.1016/j.jcct.2018.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/19/2018] [Accepted: 08/30/2018] [Indexed: 11/24/2022]
Abstract
We report a rare case of a CADASIL-syndrome with cardiac involvement presenting as ANOCA (angina in the absence of obstructive coronary artery disease). Our case highlights the added value of non-invasive fractional flow reserve (FFR)CT over coronary CT angiography (CTA), and recommends CTA as useful indication in CADASIL patients for screening of occult coronary artery disease (CAD).
Collapse
Affiliation(s)
| | - Agne Adukauskaite
- Department of Internal Medicine III- Cardiology, Innsbruck Medical University, Austria
| | - Fabian Plank
- Department of Internal Medicine III- Cardiology, Innsbruck Medical University, Austria
| | | | | |
Collapse
|
44
|
Vinters HV. The 'ACCIDENTAL NEUROPATHOLOGIST'-PERSPECTIVES on 40 years in Neuropathology. FREE NEUROPATHOLOGY 2020; 1. [PMID: 34291231 DOI: 10.17879/freeneuropathology-2020-2956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Harry V Vinters
- Distinguished Professor Emeritus, Depts. of Pathology & Laboratory Medicine & Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA; formerly Chief of Neuropathology, David Geffen School of Medicine at UCLA (1993-2016)
| |
Collapse
|
45
|
Castellazzi G, Cuzzoni MG, Cotta Ramusino M, Martinelli D, Denaro F, Ricciardi A, Vitali P, Anzalone N, Bernini S, Palesi F, Sinforiani E, Costa A, Micieli G, D'Angelo E, Magenes G, Gandini Wheeler-Kingshott CAM. A Machine Learning Approach for the Differential Diagnosis of Alzheimer and Vascular Dementia Fed by MRI Selected Features. Front Neuroinform 2020; 14:25. [PMID: 32595465 PMCID: PMC7300291 DOI: 10.3389/fninf.2020.00025] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/24/2020] [Indexed: 12/28/2022] Open
Abstract
Among dementia-like diseases, Alzheimer disease (AD) and vascular dementia (VD) are two of the most frequent. AD and VD may share multiple neurological symptoms that may lead to controversial diagnoses when using conventional clinical and MRI criteria. Therefore, other approaches are needed to overcome this issue. Machine learning (ML) combined with magnetic resonance imaging (MRI) has been shown to improve the diagnostic accuracy of several neurodegenerative diseases, including dementia. To this end, in this study, we investigated, first, whether different kinds of ML algorithms, combined with advanced MRI features, could be supportive in classifying VD from AD and, second, whether the developed approach might help in predicting the prevalent disease in subjects with an unclear profile of AD or VD. Three ML categories of algorithms were tested: artificial neural network (ANN), support vector machine (SVM), and adaptive neuro-fuzzy inference system (ANFIS). Multiple regional metrics from resting-state fMRI (rs-fMRI) and diffusion tensor imaging (DTI) of 60 subjects (33 AD, 27 VD) were used as input features to train the algorithms and find the best feature pattern to classify VD from AD. We then used the identified VD–AD discriminant feature pattern as input for the most performant ML algorithm to predict the disease prevalence in 15 dementia patients with a “mixed VD–AD dementia” (MXD) clinical profile using their baseline MRI data. ML predictions were compared with the diagnosis evidence from a 3-year clinical follow-up. ANFIS emerged as the most efficient algorithm in discriminating AD from VD, reaching a classification accuracy greater than 84% using a small feature pattern. Moreover, ANFIS showed improved classification accuracy when trained with a multimodal input feature data set (e.g., DTI + rs-fMRI metrics) rather than a unimodal feature data set. When applying the best discriminant pattern to the MXD group, ANFIS achieved a correct prediction rate of 77.33%. Overall, results showed that our approach has a high discriminant power to classify AD and VD profiles. Moreover, the same approach also showed potential in predicting earlier the prevalent underlying disease in dementia patients whose clinical profile is uncertain between AD and VD, therefore suggesting its usefulness in supporting physicians' diagnostic evaluations.
Collapse
Affiliation(s)
- Gloria Castellazzi
- NMR Research Unit, Department of Neuroinflammation, Faculty of Brain Sciences, Queen Square MS Centre, UCL Queen Square Institute of Neurology, London, United Kingdom.,Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy.,Brain MRI 3T Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | | | - Matteo Cotta Ramusino
- Laboratory of Neuropsychology and Unit of Behavioral Neurology, IRCCS Mondino Foundation, Pavia, Italy.,Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Daniele Martinelli
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Headache Center, IRCCS Mondino Foundation, Pavia, Italy
| | | | - Antonio Ricciardi
- NMR Research Unit, Department of Neuroinflammation, Faculty of Brain Sciences, Queen Square MS Centre, UCL Queen Square Institute of Neurology, London, United Kingdom.,Department of Medical Physics and Biomedical Engineering, Centre for Medical Image Computing, University College London, London, United Kingdom
| | - Paolo Vitali
- Brain MRI 3T Research Center, IRCCS Mondino Foundation, Pavia, Italy.,Radiology Unit, IRCCS Policlinico San Donato, Milan, Italy
| | - Nicoletta Anzalone
- Scientific Institute H.S. Raffaele Vita e Salute University, Milan, Italy
| | - Sara Bernini
- Laboratory of Neuropsychology and Unit of Behavioral Neurology, IRCCS Mondino Foundation, Pavia, Italy
| | - Fulvia Palesi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Elena Sinforiani
- Laboratory of Neuropsychology and Unit of Behavioral Neurology, IRCCS Mondino Foundation, Pavia, Italy
| | - Alfredo Costa
- Laboratory of Neuropsychology and Unit of Behavioral Neurology, IRCCS Mondino Foundation, Pavia, Italy.,Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Giuseppe Micieli
- Department of Emergency Neurology, IRCCS Mondino Foundation, Pavia, Italy
| | - Egidio D'Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Brain Connectivity Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Giovanni Magenes
- Brain MRI 3T Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Claudia A M Gandini Wheeler-Kingshott
- NMR Research Unit, Department of Neuroinflammation, Faculty of Brain Sciences, Queen Square MS Centre, UCL Queen Square Institute of Neurology, London, United Kingdom.,Brain MRI 3T Research Center, IRCCS Mondino Foundation, Pavia, Italy.,Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
46
|
Ferrer I, Andrés-Benito P. White matter alterations in Alzheimer's disease without concomitant pathologies. Neuropathol Appl Neurobiol 2020; 46:654-672. [PMID: 32255227 PMCID: PMC7754505 DOI: 10.1111/nan.12618] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 03/23/2020] [Indexed: 12/14/2022]
Abstract
Aims Most individuals with AD neuropathological changes have co‐morbidities which have an impact on the integrity of the WM. This study analyses oligodendrocyte and myelin markers in the frontal WM in a series of AD cases without clinical or pathological co‐morbidities. Methods From a consecutive autopsy series, 206 cases had neuropathological changes of AD; among them, only 33 were AD without co‐morbidities. WM alterations were first evaluated in coronal sections of the frontal lobe in every case. Then, RT‐qPCR and immunohistochemistry were carried out in the frontal WM of AD cases without co‐morbidities to analyse the expression of selected oligodendrocyte and myelin markers. Results WM demyelination was more marked in AD with co‐morbidities when compared with AD cases without co‐morbidities. Regarding the later, mRNA expression levels of MBP, PLP1, CNP, MAG, MAL, MOG and MOBP were preserved at stages I–II/0–A when compared with middle‐aged (MA) individuals, but significantly decreased at stages III–IV/0–C. This was accompanied by reduced expression of NG2 and PDGFRA mRNA, reduced numbers of NG2‐, Olig2‐ and HDAC2‐immunoreactive cells and reduced glucose transporter immunoreactivity. Partial recovery of some of these markers occurred at stages V–VI/B–C. Conclusions The present observations demonstrate that co‐morbidities have an impact on WM integrity in the elderly and in AD, and that early alterations in oligodendrocytes and transcription of genes linked to myelin proteins in WM occur in AD cases without co‐morbidities. These are followed by partial recovery attempts at advanced stages. These observations suggest that oligodendrocytopathy is part of AD.
Collapse
Affiliation(s)
- I Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain.,Bellvitge University Hospital, Barcelona, Spain.,Ministry of Economy and Competitiveness, CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - P Andrés-Benito
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain.,Ministry of Economy and Competitiveness, CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| |
Collapse
|
47
|
Lu J, Liu W, Zhao H. Headache in cerebrovascular diseases. Stroke Vasc Neurol 2020; 5:205-210. [PMID: 32606088 PMCID: PMC7337362 DOI: 10.1136/svn-2020-000333] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/03/2020] [Accepted: 03/09/2020] [Indexed: 12/19/2022] Open
Abstract
Headache is a common accompanying symptom of cerebrovascular diseases. The most common patterns of headache for different cerebrovascular disorders, aetiology and pathogenesis and diagnostic workup are reviewed with emphasis on distinguishing characteristics. It will be a clinical guide for physicians who treat patients with headache or cerebral vascular disease.
Collapse
Affiliation(s)
- Jiajie Lu
- Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Wei Liu
- Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hongru Zhao
- Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
48
|
Perivascular spaces in the brain: anatomy, physiology and pathology. Nat Rev Neurol 2020; 16:137-153. [PMID: 32094487 DOI: 10.1038/s41582-020-0312-z] [Citation(s) in RCA: 391] [Impact Index Per Article: 97.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2020] [Indexed: 02/06/2023]
Abstract
Perivascular spaces include a variety of passageways around arterioles, capillaries and venules in the brain, along which a range of substances can move. Although perivascular spaces were first identified over 150 years ago, they have come to prominence recently owing to advances in knowledge of their roles in clearance of interstitial fluid and waste from the brain, particularly during sleep, and in the pathogenesis of small vessel disease, Alzheimer disease and other neurodegenerative and inflammatory disorders. Experimental advances have facilitated in vivo studies of perivascular space function in intact rodent models during wakefulness and sleep, and MRI in humans has enabled perivascular space morphology to be related to cognitive function, vascular risk factors, vascular and neurodegenerative brain lesions, sleep patterns and cerebral haemodynamics. Many questions about perivascular spaces remain, but what is now clear is that normal perivascular space function is important for maintaining brain health. Here, we review perivascular space anatomy, physiology and pathology, particularly as seen with MRI in humans, and consider translation from models to humans to highlight knowns, unknowns, controversies and clinical relevance.
Collapse
|
49
|
Abrahamson EE, Ikonomovic MD. Brain injury-induced dysfunction of the blood brain barrier as a risk for dementia. Exp Neurol 2020; 328:113257. [PMID: 32092298 DOI: 10.1016/j.expneurol.2020.113257] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/31/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023]
Abstract
The blood-brain barrier (BBB) is a complex and dynamic physiological interface between brain parenchyma and cerebral vasculature. It is composed of closely interacting cells and signaling molecules that regulate movement of solutes, ions, nutrients, macromolecules, and immune cells into the brain and removal of products of normal and abnormal brain cell metabolism. Dysfunction of multiple components of the BBB occurs in aging, inflammatory diseases, traumatic brain injury (TBI, severe or mild repetitive), and in chronic degenerative dementing disorders for which aging, inflammation, and TBI are considered risk factors. BBB permeability changes after TBI result in leakage of serum proteins, influx of immune cells, perivascular inflammation, as well as impairment of efflux transporter systems and accumulation of aggregation-prone molecules involved in hallmark pathologies of neurodegenerative diseases with dementia. In addition, cerebral vascular dysfunction with persistent alterations in cerebral blood flow and neurovascular coupling contribute to brain ischemia, neuronal degeneration, and synaptic dysfunction. While the idea of TBI as a risk factor for dementia is supported by many shared pathological features, it remains a hypothesis that needs further testing in experimental models and in human studies. The current review focusses on pathological mechanisms shared between TBI and neurodegenerative disorders characterized by accumulation of pathological protein aggregates, such as Alzheimer's disease and chronic traumatic encephalopathy. We discuss critical knowledge gaps in the field that need to be explored to clarify the relationship between TBI and risk for dementia and emphasize the need for longitudinal in vivo studies using imaging and biomarkers of BBB dysfunction in people with single or multiple TBI.
Collapse
Affiliation(s)
- Eric E Abrahamson
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Milos D Ikonomovic
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
50
|
Haffner C. Proteostasis in Cerebral Small Vessel Disease. Front Neurosci 2019; 13:1142. [PMID: 31798396 PMCID: PMC6874119 DOI: 10.3389/fnins.2019.01142] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/10/2019] [Indexed: 01/02/2023] Open
Abstract
Maintaining the homeostasis of proteins (proteostasis) by controlling their synthesis, folding and degradation is a central task of cells and tissues. The gradual decline of the capacity of the various proteostasis machineries, frequently in combination with their overload through mutated, aggregation-prone proteins, is increasingly recognized as an important catalyst of age-dependent pathologies in the brain, most prominently neurodegenerative disorders. A dysfunctional proteostasis might also contribute to neurovascular disease as indicated by the occurrence of excessive protein accumulation or massive extracellular matrix expansion within vessel walls in conditions such as cerebral small vessel disease (SVD), a major cause of ischemic stroke, and cerebral amyloid angiopathy. Recent advances in brain vessel isolation techniques and mass spectrometry methodology have facilitated the analysis of cerebrovascular proteomes and fueled efforts to determine the proteomic signatures associated with neurovascular disease. In several studies in humans and mice considerable differences between healthy and diseased vessel proteomes were observed, emphasizing the critical contribution of an impaired proteostasis to disease pathogenesis. These findings highlight the important role of a balanced proteostasis for cerebrovascular health.
Collapse
Affiliation(s)
- Christof Haffner
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|