1
|
Hu Y, Zhang F, Ikonomovic M, Yang T. The Role of NRF2 in Cerebrovascular Protection: Implications for Vascular Cognitive Impairment and Dementia (VCID). Int J Mol Sci 2024; 25:3833. [PMID: 38612642 PMCID: PMC11012233 DOI: 10.3390/ijms25073833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Vascular cognitive impairment and dementia (VCID) represents a broad spectrum of cognitive decline secondary to cerebral vascular aging and injury. It is the second most common type of dementia, and the prevalence continues to increase. Nuclear factor erythroid 2-related factor 2 (NRF2) is enriched in the cerebral vasculature and has diverse roles in metabolic balance, mitochondrial stabilization, redox balance, and anti-inflammation. In this review, we first briefly introduce cerebrovascular aging in VCID and the NRF2 pathway. We then extensively discuss the effects of NRF2 activation in cerebrovascular components such as endothelial cells, vascular smooth muscle cells, pericytes, and perivascular macrophages. Finally, we summarize the clinical potential of NRF2 activators in VCID.
Collapse
Affiliation(s)
- Yizhou Hu
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA; (Y.H.); (F.Z.); (M.I.)
- Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA 15216, USA
- Department of Internal Medicine, University of Pittsburgh Medical Center (UPMC) McKeesport, McKeesport, PA 15132, USA
| | - Feng Zhang
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA; (Y.H.); (F.Z.); (M.I.)
- Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Milos Ikonomovic
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA; (Y.H.); (F.Z.); (M.I.)
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15216, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| | - Tuo Yang
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA; (Y.H.); (F.Z.); (M.I.)
- Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA 15216, USA
- Department of Internal Medicine, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15216, USA
| |
Collapse
|
2
|
Wu LY, Chai YL, Cheah IK, Chia RSL, Hilal S, Arumugam TV, Chen CP, Lai MKP. Blood-based biomarkers of cerebral small vessel disease. Ageing Res Rev 2024; 95:102247. [PMID: 38417710 DOI: 10.1016/j.arr.2024.102247] [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: 04/10/2023] [Revised: 02/12/2024] [Accepted: 02/22/2024] [Indexed: 03/01/2024]
Abstract
Age-associated cerebral small vessel disease (CSVD) represents a clinically heterogenous condition, arising from diverse microvascular mechanisms. These lead to chronic cerebrovascular dysfunction and carry a substantial risk of subsequent stroke and vascular cognitive impairment in aging populations. Owing to advances in neuroimaging, in vivo visualization of cerebral vasculature abnormities and detection of CSVD, including lacunes, microinfarcts, microbleeds and white matter lesions, is now possible, but remains a resource-, skills- and time-intensive approach. As a result, there has been a recent proliferation of blood-based biomarker studies for CSVD aimed at developing accessible screening tools for early detection and risk stratification. However, a good understanding of the pathophysiological processes underpinning CSVD is needed to identify and assess clinically useful biomarkers. Here, we provide an overview of processes associated with CSVD pathogenesis, including endothelial injury and dysfunction, neuroinflammation, oxidative stress, perivascular neuronal damage as well as cardiovascular dysfunction. Then, we review clinical studies of the key biomolecules involved in the aforementioned processes. Lastly, we outline future trends and directions for CSVD biomarker discovery and clinical validation.
Collapse
Affiliation(s)
- Liu-Yun Wu
- Memory Aging and Cognition Centre, National University Health System, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yuek Ling Chai
- Memory Aging and Cognition Centre, National University Health System, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Irwin K Cheah
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Neurobiology Programme, Centre for Life Sciences, National University of Singapore, Singapore
| | - Rachel S L Chia
- Memory Aging and Cognition Centre, National University Health System, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Saima Hilal
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Kent Ridge, Singapore
| | - Thiruma V Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea; Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Christopher P Chen
- Memory Aging and Cognition Centre, National University Health System, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Mitchell K P Lai
- Memory Aging and Cognition Centre, National University Health System, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
3
|
Pȩczalski K, Sobiech J, Buchner T, Kornack T, Foley E, Janczak D, Jakubowska M, Newby D, Ford N, Zajdel M. Synchronous recording of magnetocardiographic and electrocardiographic signals. Sci Rep 2024; 14:4098. [PMID: 38374368 PMCID: PMC11341780 DOI: 10.1038/s41598-024-54126-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 02/08/2024] [Indexed: 02/21/2024] Open
Abstract
We present a system for simultaneous recording of the electrocardiogram and the magnetocardiogram. The measurement system contained of printed carbon electrodes and SERF magnetometer. The use of this system confirms that the position of the end of the magnetic T wave extends further than the electric T wave, which is an important indicator for the diagnosis of cardiological patients and for drug arrhythmogenicity. We analyze this phenomenon in depth, and demonstrate, that it originates from the fundamental difference between electric and magnetic measurements. The measured value is always bipolar since the electric measurements require two electrodes. We demonstrate how the dual electric and magnetic measuring system adds a new information to the commonly used electrocardiographic diagnosis. The ECG should be interpreted as the spatial asymmetry of the electric cardiac potential, and not as the potential itself. The results seem to prove, that the relation between the magnetic and the electric imaging of neural activities may be broadly applied for the benefit of medical diagnosis in cardiology and many other fields, where the neural activity is measured. This is a pilot study which requires further confirmation at the clinical level.
Collapse
Affiliation(s)
| | - Judyta Sobiech
- Faculty of Physics, Warsaw University of Technology, Warsaw, Poland.
| | - Teodor Buchner
- Faculty of Physics, Warsaw University of Technology, Warsaw, Poland
| | | | | | - Daniel Janczak
- Faculty of Mechanical and Industrial Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Małgorzata Jakubowska
- Faculty of Mechanical and Industrial Engineering, Warsaw University of Technology, Warsaw, Poland
| | | | - Nancy Ford
- Twinleaf LLC, Plainsboro, NJ, 08536, USA
| | - Maryla Zajdel
- Faculty of Physics, Warsaw University of Technology, Warsaw, Poland
| |
Collapse
|
4
|
Huang J, Biessels GJ, de Leeuw FE, Ii Y, Skoog I, Mok V, Chen C, Hilal S. Cerebral microinfarcts revisited: Detection, causes, and clinical relevance. Int J Stroke 2024; 19:7-15. [PMID: 37470314 DOI: 10.1177/17474930231187979] [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: 07/21/2023]
Abstract
Cerebral microinfarcts (CMIs) are small ischemic lesions invisible to the naked eye at brain autopsy, while the larger ones (0.5-4 mm in diameter) have been visualized in-vivo on magnetic resonance imaging (MRI). CMIs can be detected on diffusion-weighted imaging (DWI) as incidental small DWI-positive lesions (ISDPLs) and on structural MRI for those confined to the cortex and in the chronic phase. ISDPLs may evolve into old cortical-CMIs, white matter hyperintensities or disappear depending on their location and size. Novel techniques in neuropathology and neuroimaging facilitate the detection of CMIs, which promotes understanding of these lesions. CMIs have heterogeneous causes, involving both cerebral small- and large-vessel disease as well as heart diseases such as atrial fibrillation and congestive heart failure. The underlying mechanisms incorporate vascular remodeling, inflammation, blood-brain barrier leakage, penetrating venule congestion, cerebral hypoperfusion, and microembolism. CMIs lead to clinical outcomes, including cognitive decline, a higher risk of stroke and mortality, and accelerated neurobehavioral disturbances. It has been suggested that CMIs can impair brain function and connectivity beyond the microinfarct core and are also associated with perilesional and global cortical atrophy. This review aims to summarize recent progress in studies involving both cortical-CMIs and ISDPLs since 2017, including their detection, etiology, risk factors, MRI correlates, and clinical consequences.
Collapse
Affiliation(s)
- Jiannan Huang
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Geert Jan Biessels
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank-Erik de Leeuw
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Yuichiro Ii
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
- Department of Neuroimaging and Pathophysiology, Mie University School of Medicine, Tsu, Japan
| | - Ingmar Skoog
- Institute of Neuroscience and Physiology and Centre for Ageing and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Psychiatry Cognition and Old Age Psychiatry, Sahlgrenska University Hospital, Region Västra Götaland, Mölndal, Sweden
| | - Vincent Mok
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese and Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Christopher Chen
- Memory Aging and Cognition Centre, National University Health System, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Saima Hilal
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
5
|
Ndunge OBA, Shikani HJ, Dai M, Freeman BD, Desruisseaux MS. Effects of anti-tau immunotherapy on reactive microgliosis, cerebral endotheliopathy, and cognitive function in an experimental model of cerebral malaria. J Neurochem 2023; 167:441-460. [PMID: 37814468 PMCID: PMC10596299 DOI: 10.1111/jnc.15972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 10/11/2023]
Abstract
Cerebral malaria (CM), a potentially fatal encephalopathy caused primarily by infection with Plasmodium falciparum, results in long-term adverse neuro-psychiatric sequelae. Neural cell injury contributes to the neurological deficits observed in CM. Abnormal regulation of tau, an axonal protein pathologically associated with the formation of neurofibrillary lesions in neurodegenerative diseases, has been linked to inflammation and cerebral microvascular compromise and has been reported in human and experimental CM (ECM). Immunotherapy with a monoclonal antibody to pathological tau (PHF-1 mAB) in experimental models of neurodegenerative diseases has been reported to mitigate cognitive decline. We investigated whether immunotherapy with PHF-1 mAB prevented cerebral endotheliopathy, neural cell injury, and neuroinflammation during ECM. Using C57BL/6 mice infected with either Plasmodium berghei ANKA (PbA), which causes ECM, Plasmodium berghei NK65 (PbN), which causes severe malaria, but not ECM, or uninfected mice (Un), we demonstrated that when compared to PbN infection or uninfected mice, PbA infection resulted in significant memory impairment at 6 days post-infection, in association with abnormal tau phosphorylation at Ser202 /Thr205 (pSer202 /Thr205 ) and Ser396-404 (pSer396-404 ) in mouse brains. ECM also resulted in significantly higher expression of inflammatory markers, in microvascular congestion, and glial cell activation. Treatment with PHF-1 mAB prevented PbA-induced cognitive impairment and was associated with significantly less vascular congestion, neuroinflammation, and neural cell activation in mice with ECM. These findings suggest that abnormal regulation of tau protein contributes to cerebral vasculopathy and is critical in the pathogenesis of neural cell injury during CM. Tau-targeted therapies may ameliorate the neural cell damage and subsequent neurocognitive impairment that occur during disease.
Collapse
Affiliation(s)
| | - Henry J. Shikani
- Albert Einstein College of Medicine, Department of Pathology, Bronx, NY, USA
| | - Minxian Dai
- Albert Einstein College of Medicine, Department of Pathology, Bronx, NY, USA
| | - Brandi D. Freeman
- Albert Einstein College of Medicine, Department of Pathology, Bronx, NY, USA
| | - Mahalia S. Desruisseaux
- Correspondence and reprint requests: Mahalia S. Desruisseaux, MD, Associate Professor of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, PO Box 208022, TAC S169B, New Haven, CT 06520-8022,
| |
Collapse
|
6
|
Kalyuzhnaya Y, Khaitin A, Demyanenko S. Modeling transient ischemic attack via photothrombosis. Biophys Rev 2023; 15:1279-1286. [PMID: 37974996 PMCID: PMC10643708 DOI: 10.1007/s12551-023-01121-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/18/2023] [Indexed: 11/19/2023] Open
Abstract
The health significance of transient ischemic attacks (TIAs) is largely underestimated. Often, TIAs are not given significant importance, and in vain, because TIAs are a predictor of the development of serious cardiovascular diseases and even death. Because of this, and because of the difficulty in diagnosing the disease, TIAs and related microinfarcts are poorly investigated. Photothrombotic models of stroke and TIA allow reproducing the occlusion of small brain vessels, even single ones. When dosing the concentration of photosensitizer, intensity and irradiation time, it is possible to achieve occlusion of well-defined small vessels with high reproducibility, and with the help of modern methods of blood flow assessment it is possible to achieve spontaneous restoration of blood flow without vessel rupture. In this review, we discuss the features of microinfarcts and the contemporary experimental approaches used to model TIA and microinfarcts, with an emphasis on models using the principle of photothrombosis of brain vessels. We review modern techniques for in vivo detection of blood flow in small brain vessels, as well as biomarkers of microinfarcts.
Collapse
Affiliation(s)
- Y.N. Kalyuzhnaya
- Southern Federal University, Academy of Biology and Biotechnology, Rostov-on-Don, Russia
| | - A.M. Khaitin
- Southern Federal University, Academy of Biology and Biotechnology, Rostov-on-Don, Russia
| | - S.V. Demyanenko
- Southern Federal University, Academy of Biology and Biotechnology, Rostov-on-Don, Russia
| |
Collapse
|
7
|
Zedde M, Grisendi I, Assenza F, Vandelli G, Napoli M, Moratti C, Lochner P, Seiffge DJ, Piazza F, Valzania F, Pascarella R. The Venular Side of Cerebral Amyloid Angiopathy: Proof of Concept of a Neglected Issue. Biomedicines 2023; 11:2663. [PMID: 37893037 PMCID: PMC10604278 DOI: 10.3390/biomedicines11102663] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Small vessel diseases (SVD) is an umbrella term including several entities affecting small arteries, arterioles, capillaries, and venules in the brain. One of the most relevant and prevalent SVDs is cerebral amyloid angiopathy (CAA), whose pathological hallmark is the deposition of amyloid fragments in the walls of small cortical and leptomeningeal vessels. CAA frequently coexists with Alzheimer's Disease (AD), and both are associated with cerebrovascular events, cognitive impairment, and dementia. CAA and AD share pathophysiological, histopathological and neuroimaging issues. The venular involvement in both diseases has been neglected, although both animal models and human histopathological studies found a deposition of amyloid beta in cortical venules. This review aimed to summarize the available information about venular involvement in CAA, starting from the biological level with the putative pathomechanisms of cerebral damage, passing through the definition of the peculiar angioarchitecture of the human cortex with the functional organization and consequences of cortical arteriolar and venular occlusion, and ending to the hypothesized links between cortical venular involvement and the main neuroimaging markers of the disease.
Collapse
Affiliation(s)
- Marialuisa Zedde
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Ilaria Grisendi
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Federica Assenza
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Gabriele Vandelli
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Manuela Napoli
- Neuroradiology Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Claudio Moratti
- Neuroradiology Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Piergiorgio Lochner
- Department of Neurology, Saarland University Medical Center, 66421 Homburg, Germany;
| | - David J. Seiffge
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Fabrizio Piazza
- CAA and AD Translational Research and Biomarkers Laboratory, School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy;
| | - Franco Valzania
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Rosario Pascarella
- Neuroradiology Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| |
Collapse
|
8
|
Iadecola C, Smith EE, Anrather J, Gu C, Mishra A, Misra S, Perez-Pinzon MA, Shih AY, Sorond FA, van Veluw SJ, Wellington CL. The Neurovasculome: Key Roles in Brain Health and Cognitive Impairment: A Scientific Statement From the American Heart Association/American Stroke Association. Stroke 2023; 54:e251-e271. [PMID: 37009740 PMCID: PMC10228567 DOI: 10.1161/str.0000000000000431] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
BACKGROUND Preservation of brain health has emerged as a leading public health priority for the aging world population. Advances in neurovascular biology have revealed an intricate relationship among brain cells, meninges, and the hematic and lymphatic vasculature (the neurovasculome) that is highly relevant to the maintenance of cognitive function. In this scientific statement, a multidisciplinary team of experts examines these advances, assesses their relevance to brain health and disease, identifies knowledge gaps, and provides future directions. METHODS Authors with relevant expertise were selected in accordance with the American Heart Association conflict-of-interest management policy. They were assigned topics pertaining to their areas of expertise, reviewed the literature, and summarized the available data. RESULTS The neurovasculome, composed of extracranial, intracranial, and meningeal vessels, as well as lymphatics and associated cells, subserves critical homeostatic functions vital for brain health. These include delivering O2 and nutrients through blood flow and regulating immune trafficking, as well as clearing pathogenic proteins through perivascular spaces and dural lymphatics. Single-cell omics technologies have unveiled an unprecedented molecular heterogeneity in the cellular components of the neurovasculome and have identified novel reciprocal interactions with brain cells. The evidence suggests a previously unappreciated diversity of the pathogenic mechanisms by which disruption of the neurovasculome contributes to cognitive dysfunction in neurovascular and neurodegenerative diseases, providing new opportunities for the prevention, recognition, and treatment of these conditions. CONCLUSIONS These advances shed new light on the symbiotic relationship between the brain and its vessels and promise to provide new diagnostic and therapeutic approaches for brain disorders associated with cognitive dysfunction.
Collapse
|
9
|
Biswas R, Kawas C, Montine TJ, Bukhari SA, Jiang L, Corrada MM. Superior Global Cognition in Oldest-Old Is Associated with Resistance to Neurodegenerative Pathologies: Results from The 90+ Study. J Alzheimers Dis 2023; 93:561-575. [PMID: 37066908 DOI: 10.3233/jad-221062] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
BACKGROUND Some oldest-old individuals can maintain superior cognition despite advanced age. Little is known about the neuropathological changes in the brains of oldest-old superior cognitive performers. OBJECTIVE Our objective was to examine the associations between Alzheimer's disease (AD) and non-AD neuropathologic features in relation to superior cognitive performance in oldest-old individuals. METHODS We analyzed brain autopsy data from 102 participants with normal cognition from The 90+ Study. Superior global cognitive performers (SGCP) were defined as having Mini-Mental State Examination (MMSE) score ≥28 in the last visit 12 to 2 months before death. To examine the associations between individual and multiple comorbid neuropathologic features with SGCP status we used multiple logistic regression models adjusting for age, sex, and education. RESULTS Alzheimer's disease neuropathological change (ADNC) and low levels of vascular pathologic change were not associated with superior cognition. In contrast, participants with limbic (OR = 8.37; 95% CI: 1.48-47.44) and neocortical (OR = 10.80;95% CI: 1.03-113.82) Lewy body disease (LBD), or with hippocampal sclerosis (HS) (OR = 5.28; 95% CI: 1.10-25.47) were more likely to be non-SGCP. High total burden of multiple comorbid neuropathologic features was associated with a lower likelihood of being SGCP. CONCLUSION Oldest-old superior cognitive performers were resilient to ADNC and low levels of vascular pathologic change and were resistant to non-AD neurodegenerative changes and multiple comorbid neuropathologic features. Understanding the factors underlying the ability of superior cognitive performers to resist these changes might provide useful insights on maintenance of superior cognition despite advanced age.
Collapse
Affiliation(s)
- Roshni Biswas
- Department of Neurology, University of California, Irvine, CA, USA
| | - Claudia Kawas
- Department of Neurology, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Thomas J Montine
- Department of Pathology, Stanford University, Palo Alto, CA, USA
| | - Syed A Bukhari
- Department of Pathology, Stanford University, Palo Alto, CA, USA
| | - Luohua Jiang
- Department of Epidemiology and Biostatistics, University of California, Irvine, CA, USA
| | - Maria M Corrada
- Department of Neurology, University of California, Irvine, CA, USA
- Department of Epidemiology and Biostatistics, University of California, Irvine, CA, USA
| |
Collapse
|
10
|
Cure of Alzheimer's Dementia Requires Addressing All of the Affected Brain Cell Types. J Clin Med 2023; 12:jcm12052049. [PMID: 36902833 PMCID: PMC10004473 DOI: 10.3390/jcm12052049] [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: 02/01/2023] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Multiple genetic, metabolic, and environmental abnormalities are known to contribute to the pathogenesis of Alzheimer's dementia (AD). If all of those abnormalities were addressed it should be possible to reverse the dementia; however, that would require a suffocating volume of drugs. Nevertheless, the problem may be simplified by using available data to address, instead, the brain cells whose functions become changed as a result of the abnormalities, because at least eleven drugs are available from which to formulate a rational therapy to correct those changes. The affected brain cell types are astrocytes, oligodendrocytes, neurons, endothelial cells/pericytes, and microglia. The available drugs include clemastine, dantrolene, erythropoietin, fingolimod, fluoxetine, lithium, memantine, minocycline, pioglitazone, piracetam, and riluzole. This article describes the ways by which the individual cell types contribute to AD's pathogenesis and how each of the drugs corrects the changes in the cell types. All five of the cell types may be involved in the pathogenesis of AD; of the 11 drugs, fingolimod, fluoxetine, lithium, memantine, and pioglitazone, each address all five of the cell types. Fingolimod only slightly addresses endothelial cells, and memantine is the weakest of the remaining four. Low doses of either two or three drugs are suggested in order to minimize the likelihood of toxicity and drug-drug interactions (including drugs used for co-morbidities). Suggested two-drug combinations are pioglitazone plus lithium and pioglitazone plus fluoxetine; a three-drug combination could add either clemastine or memantine. Clinical trials are required to validate that the suggest combinations may reverse AD.
Collapse
|
11
|
Koueik J, Wesley UV, Dempsey RJ. Pathophysiology, cellular and molecular mechanisms of large and small vessel diseases. Neurochem Int 2023; 164:105499. [PMID: 36746322 DOI: 10.1016/j.neuint.2023.105499] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/07/2023]
Abstract
Cerebrovascular disease (CVD) is the second most common cause of cognitive impairment and dementia in aged population. CVD presents in a myriad number of clinical ways based on the functional location of pathology. While primary clinical emphasis has been placed on motor, speech and visual deficits, vascular cognitive decline is a vastly under recognized and devastating condition afflicting millions of Americans. CVD, a disease of the blood vessels that supply blood to brain involves an integration between small and large vessels. Cerebral large vessel diseases (LVD) are associated with atherosclerosis, artery-to-artery embolism, intracardiac embolism and a large vessel stroke leading to substantial functional disability. Cerebral small vessel disease (SVD) is critically involved in stroke, brain hemorrhages, cognitive decline and functional loss in elderly patients. An evolving understanding of cellular and molecular mechanisms emphasizes that inflammatory vascular changes contribute to systemic pathologic conditions of the central nervous systems (CNS), with specific clinical presentations including, cognitive decline. Advances in an understanding of pathophysiology of disease processes and therapeutic interventions may help improve outcomes. This review will focus on large and small vessels diseases and their relationship to vascular cognitive decline, atherosclerosis, stroke, and inflammatory neurodegeneration. We will also emphasize the molecular and cellular mechanisms, as well as genetic and epigenetic factors associated with LVD and SVD.
Collapse
Affiliation(s)
- Joyce Koueik
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53792, USA
| | - Umadevi V Wesley
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53792, USA
| | - Robert J Dempsey
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53792, USA.
| |
Collapse
|
12
|
Zhang D, Chen Y, Shen J, Xie Q, Jing L, Lin L, Wang Q, Wu J. Static and Dynamic Characteristics of Functional Network Connectivity in Neurologically Asymptomatic Patients Undergoing Maintenance Hemodialysis: A Resting-State Functional MRI Study. J Magn Reson Imaging 2023; 57:420-431. [PMID: 35762494 PMCID: PMC10084323 DOI: 10.1002/jmri.28317] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The characteristics of static functional network connectivity (sFNC) and dynamic FNC (dFNC) in neurologically asymptomatic patients undergoing maintenance hemodialysis are unknown. Elucidating these characteristics may improve our understanding of the mechanisms of neuropathological damage in these patients. PURPOSE To explore the static and dynamic characteristics of FNC in neurologically asymptomatic patients undergoing maintenance hemodialysis and the relationship between FNC-related parameters with the neuropsychological scores and blood biomarkers. STUDY TYPE Retrospective. POPULATION A total of 23 neurologically asymptomatic patients undergoing maintenance hemodialysis and 25 healthy controls matched for age, sex, and years of education. FIELD STRENGTH/SEQUENCE A 3.0 T MRI/functional MRI and three-dimensional-T1 structural imaging ASSESSMENT: Independent components; spatial map intensity; sFNC and dFNC strengths; and time attribute parameters (mean dwell time, fractional window, and number of transitions) were determined. Neuropsychological tests were performed. Blood biochemical tests were performed for the patients but not healthy controls. STATISTICAL TESTS Chi-squared test, one-sample t-test, two-sample t-test, partial correlation analysis, and family-wise error and false discovery rate correction. P < 0.05 denoted statistical significance. RESULTS Significant group differences in the strengths of sFNC and dFNC between networks were found. The sFNC strength between the visual and sensorimotor networks was significantly associated with the global cognitive function score (i.e. the Montreal Cognitive Assessment [MoCA]) (r = 0.606). The sFNC strength between the salience and default mode networks was significantly associated with anxiety scores (r = 0.458). In state 1, positive correlations were found between the mean dwell time and backward digital span task score (r = 0.562), fractional window and MoCA score (r = 0.576), and fractional window and backward digital span task score (r = 0.592). DATA CONCLUSION Neurologically asymptomatic patients undergoing maintenance hemodialysis had defective sFNC and dFNC. Our results provide a new perspective on the mechanism of neuropathological damage in patients undergoing maintenance hemodialysis. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Die Zhang
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, People's Republic of China.,Department of Radiology, Shenzhen Third People's Hospital, Longgang District, Shenzhen, Guangdong, People's Republic of China
| | - Yingying Chen
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, People's Republic of China.,Department of Radiology, National Cancer Centre, National Clinical Research Centre for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Shenzhen, People's Republic of China
| | - Jing Shen
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, People's Republic of China
| | - Qing Xie
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, People's Republic of China
| | - Li Jing
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, People's Republic of China
| | - Lin Lin
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, People's Republic of China
| | - Qiong Wang
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, People's Republic of China
| | - Jianlin Wu
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, People's Republic of China
| |
Collapse
|
13
|
Fessel J. Formulating treatment of major psychiatric disorders: algorithm targets the dominantly affected brain cell-types. DISCOVER MENTAL HEALTH 2023; 3:3. [PMID: 37861813 PMCID: PMC10501034 DOI: 10.1007/s44192-022-00029-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 12/21/2022] [Indexed: 10/21/2023]
Abstract
BACKGROUND Pharmacotherapy for most psychiatric conditions was developed from serendipitous observations of benefit from drugs prescribed for different reasons. An algorithmic approach to formulating pharmacotherapy is proposed, based upon which combination of changed activities by brain cell-types is dominant for any particular condition, because those cell-types contain and surrogate for genetic, metabolic and environmental information, that has affected their function. The algorithm performs because functions of some or all the affected cell-types benefit from several available drugs: clemastine, dantrolene, erythropoietin, fingolimod, fluoxetine, lithium, memantine, minocycline, pioglitazone, piracetam, and riluzole PROCEDURES/FINDINGS: Bipolar disorder, major depressive disorder, schizophrenia, Alzheimer's disease, and post-traumatic stress disorder, illustrate the algorithm; for them, literature reviews show that no single combination of altered cell-types accounts for all cases; but they identify, for each condition, which combination occurs most frequently, i.e., dominates, as compared with other possible combinations. Knowing the dominant combination of altered cell-types in a particular condition, permits formulation of therapy with combinations of drugs taken from the above list. The percentage of patients who might benefit from that therapy, depends upon the frequency with which the dominant combination occurs in patients with that particular condition. CONCLUSIONS Knowing the dominant combination of changed cell types in psychiatric conditions, permits an algorithmically formulated, rationally-based treatment. Different studies of the same condition often produce discrepant results; all might be correct, because identical clinical phenotypes result from different combinations of impaired cell-types, thus producing different results. Clinical trials would validate both the proposed concept and choice of drugs.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Department of Medicine, University of California, 2069 Filbert Street, San Francisco, CA, 94123, USA.
| |
Collapse
|
14
|
BYHW Decoction Improves Cognitive Impairments in Rats with Cerebral Microinfarcts via Activation of the PKA/CREB Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4455654. [PMID: 36620084 PMCID: PMC9822752 DOI: 10.1155/2022/4455654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 11/09/2022] [Accepted: 11/16/2022] [Indexed: 01/01/2023]
Abstract
Cerebral microinfarcts (CMIs) are characterized by sporadic obstruction of small vessels leading to neurons death. They are associated with increased risk of cognitive impairments and may have different risk factors compared with macroinfarcts. CMIs have a high incidence and result in heavy social burden; thus, it is essential to provide reasonable treatment in clinical practice. However, there are relatively few researches on the mechanism and treatment of CMIs, and the literature is composed almost exclusively of community-or hospital based on autopsy or imageological studies focusing on elderly patients. The Bu Yang Huan Wu (BYHW) decoction, a traditional Chinese herbal formula, has long been used to treat stroke and stroke-related diseases, including cognitive impairments. We applied microsphere-induced CMI model in rats to investigate the behavioral and molecular consequences of CMIs and to determine how they were ameliorated by BYHW decoction treatment. We then used the Morris water maze, quantitative proteomics, immunohistochemistry, and other molecular assays and found that activation of the PKA/CREB pathway by BYHW decoction treatment may reverse mitochondrial dysfunction, inhibit apoptosis of hippocampal neurons, and ameliorate CMI-induced cognitive impairments in rats. Collectively, these findings confirmed the therapeutic potential of the BYHW decoction in treating cognitive impairments induced by CMIs and demonstrated a viable mechanism for its action.
Collapse
|
15
|
Schilling LP, Balthazar MLF, Radanovic M, Forlenza OV, Silagi ML, Smid J, Barbosa BJAP, Frota NAF, Souza LCD, Vale FAC, Caramelli P, Bertolucci PHF, Chaves MLF, Brucki SMD, Damasceno BP, Nitrini R. Diagnosis of Alzheimer’s disease: recommendations of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol 2022. [DOI: 10.1590/1980-5764-dn-2022-s102en] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023] Open
Abstract
ABSTRACT This paper presents the consensus of the Scientific Department of Cognitive Neurology and Aging from the Brazilian Academy of Neurology on the diagnostic criteria for Alzheimer’s disease (AD) in Brazil. The authors conducted a literature review regarding clinical and research criteria for AD diagnosis and proposed protocols for use at primary, secondary, and tertiary care levels. Within this clinical scenario, the diagnostic criteria for typical and atypical AD are presented as well as clinical, cognitive, and functional assessment tools and complementary propaedeutics with laboratory and neuroimaging tests. The use of biomarkers is also discussed for both clinical diagnosis (in specific conditions) and research.
Collapse
Affiliation(s)
- Lucas Porcello Schilling
- Pontifícia Universidade do Rio Grande do Sul, Brasil; Pontifícia Universidade do Rio Grande do Sul, Brasil; Pontifícia Universidade do Rio Grande do Sul, Brasil
| | | | | | | | - Marcela Lima Silagi
- Universidade Federal de São Paulo, Brasil; Universidade de São Paulo, Brasil
| | | | - Breno José Alencar Pires Barbosa
- Universidade de São Paulo, Brasil; Universidade Federal de Pernambuco, Brasil; Instituto de Medicina Integral Prof. Fernando Figueira, Brasil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Schilling LP, Balthazar MLF, Radanovic M, Forlenza OV, Silagi ML, Smid J, Barbosa BJAP, Frota NAF, de Souza LC, Vale FAC, Caramelli P, Bertolucci PHF, Chaves MLF, Brucki SMD, Damasceno BP, Nitrini R. Diagnosis of Alzheimer's disease: recommendations of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol 2022; 16:25-39. [PMID: 36533157 PMCID: PMC9745995 DOI: 10.1590/1980-5764-dn-2022-s102pt] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/22/2021] [Accepted: 04/27/2022] [Indexed: 01/25/2023] Open
Abstract
This paper presents the consensus of the Scientific Department of Cognitive Neurology and Aging from the Brazilian Academy of Neurology on the diagnostic criteria for Alzheimer's disease (AD) in Brazil. The authors conducted a literature review regarding clinical and research criteria for AD diagnosis and proposed protocols for use at primary, secondary, and tertiary care levels. Within this clinical scenario, the diagnostic criteria for typical and atypical AD are presented as well as clinical, cognitive, and functional assessment tools and complementary propaedeutics with laboratory and neuroimaging tests. The use of biomarkers is also discussed for both clinical diagnosis (in specific conditions) and research.
Collapse
Affiliation(s)
- Lucas Porcello Schilling
- Pontifícia Universidade do Rio Grande do Sul, Escola de Medicina, Serviço de Neurologia, Porto Alegre RS, Brasil
- Pontifícia Universidade do Rio Grande do Sul, Instituto do Cérebro do Rio Grande do Sul, Porto Alegre RS, Brasil
- Pontifícia Universidade do Rio Grande do Sul, Programa de Pós-Graduação em Gerontologia Biomédica, Porto Alegre RS, Brasil
| | | | - Márcia Radanovic
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Psiquiatria, Laboratório de Neurociências, São Paulo SP, Brasil
| | - Orestes Vicente Forlenza
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Psiquiatria, Laboratório de Neurociências, São Paulo SP, Brasil
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Psiquiatria, São Paulo SP, Brasil
| | - Marcela Lima Silagi
- Universidade Federal de São Paulo, Departamento de Fonoaudiologia, São Paulo SP, Brasil
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
| | - Jerusa Smid
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
| | - Breno José Alencar Pires Barbosa
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
- Universidade Federal de Pernambuco, Centro de Ciências Médicas, Área Acadêmica de Neuropsiquiatria, Recife PE, Brasil
- Instituto de Medicina Integral Prof. Fernando Figueira, Recife PE, Brasil
| | | | - Leonardo Cruz de Souza
- Universidade Federal de Minas Gerais, Departamento de Clínica Médica, Belo Horizonte MG, Brasil
| | - Francisco Assis Carvalho Vale
- Universidade Federal de São Carlos, Centro de Ciências Biológicas e da Saúde, Departamento de Medicina, São Carlos SP, Brasil
| | - Paulo Caramelli
- Universidade Federal de Minas Gerais, Departamento de Clínica Médica, Belo Horizonte MG, Brasil
| | | | - Márcia Lorena Fagundes Chaves
- Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Porto Alegre RS, Brasil
- Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Medicina Interna, Porto Alegre RS, Brasil
| | - Sonia Maria Dozzi Brucki
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
| | - Benito Pereira Damasceno
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Neurologia, Campinas SP, Brasil
| | - Ricardo Nitrini
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
| |
Collapse
|
17
|
Tao W, Cheng Y, Guo W, Kwapong WR, Ye C, Wu B, Zhang S, Liu M. Clinical features and imaging markers of small vessel disease in symptomatic acute subcortical cerebral microinfarcts. BMC Neurol 2022; 22:311. [PMID: 35999494 PMCID: PMC9396904 DOI: 10.1186/s12883-022-02824-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/03/2022] [Indexed: 11/30/2022] Open
Abstract
Background As currently defined, recent small subcortical infarcts (RSSI) do not have a lower size boundary, and the smallest diffusion-weighted imaging (DWI) infarcts, which we term acute subcortical cerebral microinfarcts (As-CMI) with lesion diameter less than 5 mm, might have clinical implications distinct from RSSI. We aimed to investigate the distinct characteristics of As-CMI as compared to the larger size of RSSI regarding vascular risk factors, clinical manifestation, radiological markers of SVD distribution, and outcomes. Methods In a consecutive cohort, patients were selected with a magnetic resonance DWI-confirmed RSSI between January 2010 and November 2020. We measured axial infarct diameter and classified patients into two groups: The As-CMI group (diameter < 5 mm) versus the Larger RSSI group (diameter 5-20 mm). Clinical variables, including vascular risk factors, clinical symptoms/signs, lesion locations, and radiological markers of cerebral small vessel disease (SVD) on MRI were analyzed between the two groups. Patients were followed up for 12 months and functional outcomes were measured by the modified ranking scale (mRS). Results In a total of 584 patients with RSSI, 23 (3.9%) were defined as As-CMI. The most common neurological deficits with As-CMI were hemiparalysis (n = 20), followed by central facial/lingual palsy (n = 10) and hemidysesthesia (n = 10). Most As-CMIs were located in the basal ganglia (n = 11), followed by the thalamus (n = 5) and centrum semiovale (n = 4). No different regional distributions and symptoms/signs frequencies were found between the two groups except for a lower percentage of dysarthria in the As-CMI group (p = 0.008). In a multivariate analysis, patients with As-CMI were independently associated with the presence of lacunes (adjusted odds ratio [aOR] 2.88; 95% confidence interval [CI] 1.21–6.84), multiple lacunes (aOR 3.5, CI 1.29–9.48) and higher total SVD burden (aOR 1.68, CI 1.11–2.53). Patients with As-CMI did not show a better functional outcome after 12 months of follow-up. Conclusions Patients with As-CMI had a non-specific clinical profile but a higher burden of SVD, indicating As-CMI might be s sign of more severe small vascular injury. Whether its vascular features are associated with worse cognitive outcomes requires further investigation.
Collapse
Affiliation(s)
- Wendan Tao
- Center of Cerebrovascular Disease, Department of Neurology, West China Hospital, Sichuan University, Sichuan Province, No. 37 Guo Xue Xiang, Chengdu, 610041, People's Republic of China
| | - Yajun Cheng
- Center of Cerebrovascular Disease, Department of Neurology, West China Hospital, Sichuan University, Sichuan Province, No. 37 Guo Xue Xiang, Chengdu, 610041, People's Republic of China.,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Wen Guo
- Center of Cerebrovascular Disease, Department of Neurology, West China Hospital, Sichuan University, Sichuan Province, No. 37 Guo Xue Xiang, Chengdu, 610041, People's Republic of China
| | - William Robert Kwapong
- Center of Cerebrovascular Disease, Department of Neurology, West China Hospital, Sichuan University, Sichuan Province, No. 37 Guo Xue Xiang, Chengdu, 610041, People's Republic of China
| | - Chen Ye
- Center of Cerebrovascular Disease, Department of Neurology, West China Hospital, Sichuan University, Sichuan Province, No. 37 Guo Xue Xiang, Chengdu, 610041, People's Republic of China
| | - Bo Wu
- Center of Cerebrovascular Disease, Department of Neurology, West China Hospital, Sichuan University, Sichuan Province, No. 37 Guo Xue Xiang, Chengdu, 610041, People's Republic of China
| | - Shuting Zhang
- Center of Cerebrovascular Disease, Department of Neurology, West China Hospital, Sichuan University, Sichuan Province, No. 37 Guo Xue Xiang, Chengdu, 610041, People's Republic of China.
| | - Ming Liu
- Center of Cerebrovascular Disease, Department of Neurology, West China Hospital, Sichuan University, Sichuan Province, No. 37 Guo Xue Xiang, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
18
|
Agrawal S, Schneider JA. Vascular pathology and pathogenesis of cognitive impairment and dementia in older adults. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2022; 3:100148. [PMID: 36324408 PMCID: PMC9616381 DOI: 10.1016/j.cccb.2022.100148] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 05/25/2022] [Accepted: 06/23/2022] [Indexed: 12/24/2022]
Abstract
It is well recognized that brains of older people often harbor cerebrovascular disease pathology including vessel disease and vascular-related tissue injuries and that this is associated with vascular cognitive impairment and contributes to dementia. Here we review vascular pathologies, cognitive impairment, and dementia. We highlight the importance of mixed co-morbid AD/non-AD neurodegenerative and vascular pathology that has been collected in multiple clinical pathologic studies, especially in community-based studies. We also provide an update of vascular pathologies from the Rush Memory and Aging Project and Religious Orders Study cohorts with special emphasis on the differences across age in persons with and without dementia. Finally, we discuss neuropathological perspectives on the interpretation of clinical-pathological studies and emerging data in community-based studies.
Collapse
Affiliation(s)
- Sonal Agrawal
- Rush Alzheimer's Disease Center, Rush University Medical Center, Jelke Building, 1750 W. Harrison Street, Chicago 60612, IL, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Julie A. Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Jelke Building, 1750 W. Harrison Street, Chicago 60612, IL, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
19
|
Ghaznawi R, Zwartbol MHT, de Bresser J, Kuijf HJ, Vincken KL, Rissanen I, Geerlings MI, Hendrikse J. Microinfarcts in the Deep Gray Matter on 7T MRI: Risk Factors, MRI Correlates, and Relation to Cognitive Functioning-The SMART-MR Study. AJNR Am J Neuroradiol 2022; 43:829-836. [PMID: 35618425 DOI: 10.3174/ajnr.a7512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 03/24/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE The clinical relevance of cortical microinfarcts has recently been established; however, studies on microinfarcts in the deep gray matter are lacking. We examined the risk factors and MR imaging correlates of microinfarcts in the deep gray matter on 7T MR imaging and their relation to cognitive functioning. MATERIALS AND METHODS Within the Second Manifestations of ARTerial disease-Magnetic Resonance (SMART-MR) study, 213 patients (mean age, 68 [SD, 8] years) had a risk-factor assessment, 7T and 1.5T brain MR imaging, and a cognitive examination. Microinfarcts on 7T MR imaging were defined as lesions of <5 mm. Regression models were used to examine the age-adjusted associations among risk factors, MR imaging markers, and microinfarcts. Cognitive function was summarized as composite and domain-specific z scores. RESULTS A total of 47 microinfarcts were found in 28 patients (13%), most commonly in the thalamus. Older age, history of stroke, hypertension, and intima-media thickness were associated with microinfarcts. On 1.5T MR imaging, cerebellar infarcts (relative risk = 2.75; 95% CI, 1.4-5.33) and lacunes in the white (relative risk = 3.28; 95% CI, 3.28-6.04) and deep gray matter (relative risk = 3.06; 95% CI, 1.75-5.35) were associated with microinfarcts, and on 7T MR imaging cortical microinfarcts (relative risk = 2.33; 95% CI, 1.32-4.13). Microinfarcts were also associated with poorer global cognitive functioning (mean difference in the global z score between patients with multiple microinfarcts versus none = -0.97; 95% CI, -1.66 to -0.28, P = .006) and across all cognitive domains. CONCLUSIONS Microinfarcts in the deep gray matter on 7T MR imaging were associated with worse cognitive functioning and risk factors and MR imaging markers of small-vessel and large-vessel disease. Our findings suggest that microinfarcts in the deep gray matter may represent a novel imaging marker of vascular brain injury.
Collapse
Affiliation(s)
- R Ghaznawi
- Form the Department of Radiology (R.G., M.H.T.Z., J.H.), University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands.,Julius Center for Health Sciences and Primary Care (R.G., I.R., M.I.G.), University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - M H T Zwartbol
- Form the Department of Radiology (R.G., M.H.T.Z., J.H.), University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - J de Bresser
- Department of Radiology (J.D.B.), Leiden University Medical Center, Leiden, the Netherlands
| | - H J Kuijf
- Image Sciences Institute (H.J.K, K.L.V), University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - K L Vincken
- Image Sciences Institute (H.J.K, K.L.V), University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - I Rissanen
- Julius Center for Health Sciences and Primary Care (R.G., I.R., M.I.G.), University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - M I Geerlings
- Julius Center for Health Sciences and Primary Care (R.G., I.R., M.I.G.), University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | | | | |
Collapse
|
20
|
Association of cerebral microvascular dysfunction and white matter injury in Alzheimer's disease. GeroScience 2022; 44:1-14. [PMID: 35612774 PMCID: PMC9617002 DOI: 10.1007/s11357-022-00585-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/05/2022] [Indexed: 12/23/2022] Open
Abstract
Patients with Alzheimer's disease (AD) often have cerebral white matter (WM) hyperintensities on MRI and microinfarcts of presumed microvascular origin pathologically. Here, we determined if vasodilator dysfunction of WM-penetrating arterioles is associated with pathologically defined WM injury and disturbances in quantitative MRI-defined WM integrity in patients with mixed microvascular and AD pathology. We analyzed tissues from 28 serially collected human brains from research donors diagnosed with varying degrees of AD neuropathologic change (ADNC) with or without cerebral microinfarcts (mVBI). WM-penetrating and pial surface arteriolar responses to the endothelium-dependent agonist bradykinin were quantified ex vivo with videomicroscopy. Vascular endothelial nitric oxide synthase (eNOS) and NAD(P)H-oxidase (Nox1, 2 and 4 isoforms) expression were measured with quantitative PCR. Glial fibrillary acidic protein (GFAP)-labeled astrocytes were quantified by unbiased stereological approaches in regions adjacent to the sites of WM-penetrating vessel collection. Post-mortem diffusion tensor imaging (DTI) was used to measure mean apparent diffusion coefficient (ADC) and fractional anisotropy (FA), quantitative indices of WM integrity. In contrast to pial surface arterioles, white matter-penetrating arterioles from donors diagnosed with high ADNC and mVBI exhibited a significantly reduced dilation in response to bradykinin when compared to the other groups. Expression of eNOS was reduced, whereas Nox1 expression was increased in WM arterioles in AD and mVBI cases. WM astrocyte density was increased in AD and mVBI, which correlated with a reduced vasodilation in WM arterioles. Moreover, in cases with low ADNC, bradykinin-induced WM arteriole dilation correlated with lower ADC and higher FA values. Comorbid ADNC and mVBI appear to synergistically interact to selectively impair bradykinin-induced vasodilation in WM-penetrating arterioles, which may be related to reduced nitric oxide- and excess reactive oxygen species-mediated vascular endothelial dysfunction. WM arteriole vasodilator dysfunction is associated with WM injury, as supported by reactive astrogliosis and MRI-defined disrupted WM microstructural integrity.
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
|
Weil EL, Noseworthy PA, Lopez CL, Rabinstein AA, Friedman PA, Attia ZI, Yao X, Siontis KC, Kremers WK, Christopoulos G, Mielke MM, Vemuri P, Jack CR, Gersh BJ, Machulda MM, Knopman DS, Petersen RC, Graff-Radford J. Artificial Intelligence-Enabled Electrocardiogram for Atrial Fibrillation Identifies Cognitive Decline Risk and Cerebral Infarcts. Mayo Clin Proc 2022; 97:871-880. [PMID: 35512882 PMCID: PMC9179015 DOI: 10.1016/j.mayocp.2022.01.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/01/2021] [Accepted: 01/11/2022] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To investigate whether artificial intelligence-enabled electrocardiogram (AI-ECG) assessment of atrial fibrillation (AF) risk predicts cognitive decline and cerebral infarcts. PATIENTS AND METHODS This population-based study included sinus-rhythm ECG participants seen from November 29, 2004 through July 13, 2020, and a subset with brain magnetic resonance imaging (MRI) (October 10, 2011, through November 2, 2017). The AI-ECG score of AF risk calculated for participants was 0-1. To determine the AI-ECG-AF relationship with baseline cognitive dysfunction, we compared linear mixed-effects models with global and domain-specific cognitive z-scores from longitudinal neuropsychological assessments. The AI-ECG-AF score was logit transformed and modeled with cubic splines. For the brain-MRI subset, logistic regression evaluated correlation of the AI-ECG-AF score and the high-threshold, dichotomized AI-ECG-AF score with infarcts. RESULTS Participants (N=3729; median age, 74.1 years) underwent cognitive analysis. Adjusting for age, sex, education, and APOE ɛ4-carrier status, the AI-ECG-AF score correlated with lower baseline and faster decline in global-cognitive z-scores (P=.009 and P=.01, respectively, non-linear-based spline-models tests) and attention z-scores (P<.001 and P=.01, respectively). Sinus-rhythm-ECG participants (n=1373) underwent MRI. As a continuous measure, the AI-ECG-AF score correlated with infarcts but not after age and sex adjustment (P=.52). For dichotomized analysis, an AI-ECG-AF score greater than 0.5 correlated with infarcts (OR, 4.61; 95% CI, 2.45-8.55; P<.001); even after age and sex adjustment (OR, 2.09; 95% CI, 1.06-4.07; P=.03). CONCLUSION The AI-ECG-AF score correlated with worse baseline cognition and gradual global cognition and attention decline. High AF probability by AI-ECG-AF score correlated with MRI cerebral infarcts. However, most infarcts observed in our cohort were subcortical, suggesting that AI-ECG not only predicts AF but also detects other non-AF cardiac disease markers and correlates with small vessel cerebrovascular disease and cognitive decline.
Collapse
Affiliation(s)
- Erika L Weil
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Camden L Lopez
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | - Paul A Friedman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Zachi I Attia
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Xiaoxi Yao
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA; Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN, USA
| | | | - Walter K Kremers
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | - Michelle M Mielke
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Bernard J Gersh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | | | | | | |
Collapse
|
23
|
Tao W, Wang Z, Liu J, Li J, Deng Y, Guo W, Wei W, Wu B, Liu M. Acute Cerebral Microinfarcts in Acute Ischemic Stroke: Imaging and Clinical Significance. Cerebrovasc Dis 2022; 51:755-763. [PMID: 35490673 DOI: 10.1159/000524021] [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: 12/23/2021] [Accepted: 02/18/2022] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Limited data exist on the significance of acute cerebral microinfarcts (A-CMIs) in the context of acute ischemic stroke (AIS). We aimed to determine the profile and prognostic significance of A-CMIs on magnetic resonance imaging (MRI) in patients presenting with AIS. METHODS A prospective single-center series of patients with AIS who had 3T MRIs between March 2013 and December 2019. The presence, number, and location of A-CMIs on diffusion-weighted imaging, and markers of cerebral small vessel disease (CSVD), macroinfarcts features, and etiology were classified as cardioembolism (CE) or large artery atherosclerosis (LAA) or none. RESULTS Among 273 patients, A-CMIs were detected in 130 patients (47.6%), of whom cortical A-CMIs were found in 95 (73.0%) patients. Patients with A-CMIs were significantly older, less likely to have diabetes mellitus, and more likely to have atrial fibrillation and an embolic source (CE or LAA) compared to other patients. Patients with A-CMI had a higher frequency of macroinfarcts (diameter >20 mm), more often multiple and distributed in single or multiple vessel territories than other patients. An embolic source (LAA or CE) was independently associated with cortical A-CMIs (LAA adjusted odds ratio [aOR] 4.0 95% confidence interval [CI] 1.6-9.5; CE aOR 2.5, 95% CI 1.1-5.6), whereas lacunes were independently related to subcortical A-CMIs (aOR 2.6, 95% CI 1.2-5.8). CONCLUSIONS We have shown A-CMIs occur in cortical and subcortical regions in nearly half of AIS patients, where microembolism and CSVD are, respectively, the key presumed etiological mechanism.
Collapse
Affiliation(s)
- Wendan Tao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China,
| | - Zhetao Wang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Junfeng Liu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Li
- Department of Neurology, Deyang People's Hospital, Deyang, China
| | - Yilun Deng
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Wen Guo
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Wei
- Department of Radiology, Chengdu First People's Hospital, Chengdu, China
| | - Bo Wu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Ming Liu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Lee J, Kim JG, Hong S, Kim YS, Ahn S, Kim R, Chun H, Park KD, Jeong Y, Kim DE, Lee CJ, Ku T, Kim P. Longitudinal intravital imaging of cerebral microinfarction reveals a dynamic astrocyte reaction leading to glial scar formation. Glia 2022; 70:975-988. [PMID: 35106851 DOI: 10.1002/glia.24151] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/24/2021] [Accepted: 01/17/2022] [Indexed: 01/25/2023]
Abstract
Cerebral microinfarct increases the risk of dementia. But how microscopic cerebrovascular disruption affects the brain tissue in cellular-level are mostly unknown. Herein, with a longitudinal intravital imaging, we serially visualized in vivo dynamic cellular-level changes in astrocyte, pericyte and neuron as well as microvascular integrity after the induction of cerebral microinfarction for 1 month in mice. At day 2-3, it revealed a localized edema with acute astrocyte loss, neuronal death, impaired pericyte-vessel coverage and extravascular leakage of 3 kDa dextran (but not 2 MDa dextran) indicating microinfarction-related blood-brain barrier (BBB) dysfunction for small molecules. At day 5, the local edema disappeared with the partial restoration of microcirculation and recovery of pericyte-vessel coverage and BBB integrity. But brain tissue continued to shrink with persisted loss of astrocyte and neuron in microinfarct until 30 days, resulting in a collagen-rich fibrous scar surrounding the microinfarct. Notably, reactive astrocytes expressing glial fibrillary acidic protein (GFAP) appeared at the peri-infarct area early at day 2 and thereafter accumulated in the peri-infarct until 30 days, inducing glial scar formation in cerebral cortex. Our longitudinal intravital imaging of serial microscopic neurovascular pathophysiology in cerebral microinfarction newly revealed that astrocytes are critically susceptible to the acute microinfarction and their reactive response leads to the fibrous glial scar formation.
Collapse
Affiliation(s)
- Jingu Lee
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Joon-Goon Kim
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Sujung Hong
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Young Seo Kim
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Soyeon Ahn
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ryul Kim
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Heejung Chun
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Ki Duk Park
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,Division of Bio-Med Science & Technology, KIST School, University of Science and Technology (UST), Seoul, South Korea
| | - Yong Jeong
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Dong-Eog Kim
- Department of Neurology, Dongguk University College of Medicine, Dongguk University Ilsan Hospital, Goyang, South Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Taeyun Ku
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Pilhan Kim
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| |
Collapse
|
25
|
Lecordier S, Pons V, Rivest S, ElAli A. Multifocal Cerebral Microinfarcts Modulate Early Alzheimer’s Disease Pathology in a Sex-Dependent Manner. Front Immunol 2022; 12:813536. [PMID: 35173711 PMCID: PMC8841345 DOI: 10.3389/fimmu.2021.813536] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/17/2021] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease (AD) constitutes a major cause of dementia, affecting more women than men. It is characterized by amyloid-β (Aβ) deposition and neurofibrillary tangles (NFTs) formation, associated with a progressive cognitive decline. Evidence indicates that AD onset increases the prevalence of cerebral microinfarcts caused by vascular pathologies, which occur in approximately in half of AD patients. In this project, we postulated that multifocal cerebral microinfarcts decisively influence early AD-like pathology progression in a sex dependent manner in young APP/PS1 mice. For this purpose, we used a novel approach to model multifocal microinfarcts in APP/PS1 mice via the sporadic occlusions of the microvasculature. Our findings indicate that microinfarcts reduced Aβ deposits without affecting soluble Aβ levels in the brain of male and female APP/PS1 mice, while causing rapid and prolonged cognitive deficits in males, and a mild and transient cognitive decline in females. In male APP/PS1 mice, microinfarcts triggered an acute hypoperfusion followed by a chronic hyperperfusion. Whereas in female APP/PS1 mice, microinfarcts caused an acute hypoperfusion, which was recovered in the chronic phase. Microinfarcts triggered a robust microglial activation and recruitment of peripheral monocytes to the lesion sites and Aβ plaques more potently in female APP/PS1 mice, possibly accounting for the reduced Aβ deposition. Finally, expression of Dickkopf-1 (DKK1), which plays a key role in mediating synaptic and neuronal dysfunction in AD, was strongly induced at the lesion sites of male APP/PS1 mice, while its expression was reduced in females. Our findings suggest that multifocal microinfarcts accelerate AD pathology more potently in young males compared to young females independently upon Aβ pathology via modulation of neurovascular coupling, inflammatory response, and DKK1 expression. Our results suggest that the effects of microinfarcts should be taken into consideration in AD diagnosis, prognosis, and therapies.
Collapse
Affiliation(s)
- Sarah Lecordier
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Quebec City, QC, Canada
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Vincent Pons
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Quebec City, QC, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Serge Rivest
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Quebec City, QC, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Ayman ElAli
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Quebec City, QC, Canada
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- *Correspondence: Ayman ElAli,
| |
Collapse
|
26
|
Poh L, Sim WL, Jo DG, Dinh QN, Drummond GR, Sobey CG, Chen CLH, Lai MKP, Fann DY, Arumugam TV. The role of inflammasomes in vascular cognitive impairment. Mol Neurodegener 2022; 17:4. [PMID: 35000611 PMCID: PMC8744307 DOI: 10.1186/s13024-021-00506-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 12/02/2021] [Indexed: 12/11/2022] Open
Abstract
There is an increasing prevalence of Vascular Cognitive Impairment (VCI) worldwide, and several studies have suggested that Chronic Cerebral Hypoperfusion (CCH) plays a critical role in disease onset and progression. However, there is a limited understanding of the underlying pathophysiology of VCI, especially in relation to CCH. Neuroinflammation is a significant contributor in the progression of VCI as increased systemic levels of the proinflammatory cytokine interleukin-1β (IL-1β) has been extensively reported in VCI patients. Recently it has been established that CCH can activate the inflammasome signaling pathways, involving NLRP3 and AIM2 inflammasomes that critically regulate IL-1β production. Given that neuroinflammation is an early event in VCI, it is important that we understand its molecular and cellular mechanisms to enable development of disease-modifying treatments to reduce the structural brain damage and cognitive deficits that are observed clinically in the elderly. Hence, this review aims to provide a comprehensive insight into the molecular and cellular mechanisms involved in the pathogenesis of CCH-induced inflammasome signaling in VCI.
Collapse
Affiliation(s)
- Luting Poh
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei Liang Sim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Quynh Nhu Dinh
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| | - Grant R. Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| | - Christopher G. Sobey
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| | - Christopher Li-Hsian Chen
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mitchell K. P. Lai
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Y. Fann
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Healthy Longevity, National University Health System (NUHS), Singapore, Singapore
| | - Thiruma V. Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| |
Collapse
|
27
|
Ezeala-Adikaibe B, Oti BB, Ohaegbulam S, Ndubuisi C, Okwudili O. Pattern of structural magnetic resonance imaging lesions in the patients with progressive cognitive decline: A single-center study in Southeast Nigeria. WEST AFRICAN JOURNAL OF RADIOLOGY 2022. [DOI: 10.4103/wajr.wajr_23_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
28
|
Luo X, Hong H, Wang S, Li K, Zeng Q, Hong L, Liu X, Li Z, Fu Y, Jiaerken Y, Xu X, Yu X, Huang P, Zhang M. Exploration of the Mechanism Underlying the Association of Incident Microinfarct and Motor Deficit: A Preliminary Functional MRI Study. J Alzheimers Dis 2021; 85:1545-1554. [PMID: 34958031 DOI: 10.3233/jad-215227] [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: 11/15/2022]
Abstract
BACKGROUND Cerebral microinfarcts (CMIs) might cause measurable disruption to brain connections and are associated with cognitive decline, but the association between CMIs and motor impairment is still unclear. OBJECTIVE To assess the CMIs effect on motor function in vivo and explore the potential neuropathological mechanism based on graph-based network method. METHODS We identified 133 non-demented middle-aged and elderly participants who underwent MRI scanning, cognitive, and motor assessment. The short physical performance battery (SPPB) assessed motor function, including balance, walking speed, and chair stand. We grouped participants into 34 incident CMIs carriers and 99 non-CMIs carriers as controls, depending on diffusion-weighted imaging. Then we assessed the independent CMIs effects on motor function and explored neural mechanisms of CMIs on motor impairment via mapping of degree centrality (DC) and eigenvector centrality (EC). RESULTS CMIs carriers had worse motor function than non-carriers. Linear regression analyses showed that CMIs independently contributed to motor function. CMIs carriers had decreased EC in the precuneus, while increased DC and EC in the middle temporal gyrus and increased DC in the inferior frontal gyrus compared to controls (p < 0.05, corrected). Correlation analyses showed that EC of precuneus was related to SPPB (r = 0.25) and balance (r = 0.27); however, DC (r = -0.25) and EC (r = -0.25) of middle temporal gyrus was related with SPPB in all participants (p < 0.05, corrected). CONCLUSION CMIs represent an independent risk factor for motor dysfunction. The relationship between CMIs and motor function may be attributed to suppression of functional hub region and compensatory activation of motor-related regions.
Collapse
Affiliation(s)
- Xiao Luo
- Department of Radiology, The 2nd Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Hong
- Department of Radiology, The 2nd Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shuyue Wang
- Department of Radiology, The 2nd Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Kaicheng Li
- Department of Radiology, The 2nd Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Qingze Zeng
- Department of Radiology, The 2nd Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Luwei Hong
- Department of Radiology, The 2nd Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaocao Liu
- Department of Radiology, The 2nd Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zheyu Li
- Department of Neurology, The 2nd Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yanv Fu
- Department of Neurology, The 2nd Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yeerfan Jiaerken
- Department of Radiology, The 2nd Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - XiaoPei Xu
- Department of Radiology, The 2nd Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xinfeng Yu
- Department of Radiology, The 2nd Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Peiyu Huang
- Department of Radiology, The 2nd Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Minming Zhang
- Department of Radiology, The 2nd Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
29
|
Ferro DA, Kuijf HJ, Hilal S, van Veluw SJ, van Veldhuizen D, Venketasubramanian N, Tan BY, Biessels GJ, Chen C. Association Between Cerebral Cortical Microinfarcts and Perilesional Cortical Atrophy on 3T MRI. Neurology 2021; 98:e612-e622. [PMID: 34862322 DOI: 10.1212/wnl.0000000000013140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 11/16/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Cerebral cortical microinfarcts (CMIs) are a novel MRI-marker of cerebrovascular disease (CeVD) that predicts accelerated cognitive decline. Presence of CMIs is known to be associated with global cortical atrophy, although the mechanism linking the two is unclear. Our primary objective was to examine the relation between CMIs and cortical atrophy and establish possible perilesional atrophy surrounding CMIs. Our secondary objective was to examine the role of cortical atrophy in CMI-associated cognitive impairment. METHODS Patients were recruited from two Singapore memory clinics between December 2010 and September 2013 and included if they received the diagnosis no objective cognitive impairment, cognitive impairment (with or without a history of stroke) or Alzheimer's or vascular dementia. Cortical thickness, chronic cortical microinfarcts and MRI-markers of CeVD were assessed on 3T MRI. Patients underwent cognitive testing. Cortical thickness was compared globally between patients with and without CMIs, regionally within individual patients with CMIs comparing brain regions with CMIs to the corresponding contralateral region without CMIs and locally within individuals patients in a 50 mm radius of CMIs. Global cortical thickness was analyzed as mediator in the relation between CMI and cognitive performance. RESULTS Of the 238 patients (mean age 72.5 SD 9.1 years) enrolled, 75 had ≥1 CMIs. Patient with CMIs had a 2.1% lower global cortical thickness (B=-.049 mm, 95% CI [.091; -.007] p=.022) compared to patients without CMIs, after correction for age, sex, education and intracranial volume. In patients with CMIs, cortical thickness in brain regions with CMIs was 2.2 % lower than in contralateral regions without CMIs (B=-.048 mm [-.071; -.026] p<.001). In a 20 mm radius area surrounding the CMI-core, cortical thickness was lower than in the area 20-50 mm from the CMI-core (Mean difference -.06 mm 95% CI [-.10; -.02] p=.002). Global cortical thickness was a significant mediator in the relationship between CMI presence and cognitive performance as measure with the Mini-Mental State Examination (B=-.12 [-.22; -.01] p=.025). DISCUSSION We found cortical atrophy surrounding CMIs, suggesting a perilesional effect in a cortical area many times larger than the CMI-core. Our findings support the notion that CMIs affect brain structure beyond the actual lesion site.
Collapse
Affiliation(s)
- Doeschka A Ferro
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Hugo J Kuijf
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Saima Hilal
- Memory Aging and Cognition Centre, Department of Pharmacology, National University of Singapore, Singapore
| | - Susanne J van Veluw
- Department of Neurology, J.P.K. Stroke Research Center, Massachusetts General Hospital, Boston, MA, USA
| | | | | | | | - Geert Jan Biessels
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Christopher Chen
- Memory Aging and Cognition Centre, Department of Pharmacology, National University of Singapore, Singapore
| |
Collapse
|
30
|
Wiegertjes K, Chan KS, Telgte AT, Gesierich B, Norris DG, Klijn CJ, Duering M, Tuladhar AM, Marques JP, Leeuw FED. Assessing cortical cerebral microinfarcts on iron-sensitive MRI in cerebral small vessel disease. J Cereb Blood Flow Metab 2021; 41:3391-3399. [PMID: 34415209 PMCID: PMC8669205 DOI: 10.1177/0271678x211039609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Recent studies suggest that a subset of cortical microinfarcts may be identifiable on T2* but invisible on T1 and T2 follow-up images. We aimed to investigate whether cortical microinfarcts are associated with iron accumulation after the acute stage. The RUN DMC - InTENse study is a serial MRI study including individuals with cerebral small vessel disease (SVD). 54 Participants underwent 10 monthly 3 T MRIs, including diffusion-weighted imaging, quantitative R1 (=1/T1), R2 (=1/T2), and R2* (=1/T2*) mapping, from which MRI parameters within areas corresponding to microinfarcts and control region of interests (ROIs) were retrieved within 16 participants. Finally, we compared pre- and post-lesional values with repeated measures ANOVA and post-hoc paired t-tests using the mean difference between lesion and control ROI values. We observed 21 acute cortical microinfarcts in 7 of the 54 participants (median age 69 years [IQR 66-74], 63% male). R2* maps demonstrated an increase in R2* values at the moment of the last available follow-up MRI (median [IQR], 5 [5-14] weeks after infarction) relative to prelesional values (p = .08), indicative of iron accumulation. Our data suggest that cortical microinfarcts are associated with increased R2* values, indicative of iron accumulation, possibly due to microhemorrhages, neuroinflammation or neurodegeneration, awaiting histopathological verification.
Collapse
Affiliation(s)
- Kim Wiegertjes
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Kwok-Shing Chan
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, Nijmegen, the Netherlands
| | - Annemieke Ter Telgte
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Benno Gesierich
- Institute for Stroke and Dementia Research (ISD), University Hospital LMU Munich, Munich, Germany
| | - David G Norris
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, Nijmegen, the Netherlands.,Erwin L. Hahn Institute for Magnetic Resonance Imaging, University of Duisburg-Essen, Essen, Germany.,MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Catharina Jm Klijn
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marco Duering
- Institute for Stroke and Dementia Research (ISD), University Hospital LMU Munich, Munich, Germany.,Medical Image Analysis Center (MIAC AG), Basel and qbig, Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Anil M Tuladhar
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - José P Marques
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, Nijmegen, the Netherlands
| | - Frank-Erik de Leeuw
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
31
|
Lecordier S, Manrique-Castano D, El Moghrabi Y, ElAli A. Neurovascular Alterations in Vascular Dementia: Emphasis on Risk Factors. Front Aging Neurosci 2021; 13:727590. [PMID: 34566627 PMCID: PMC8461067 DOI: 10.3389/fnagi.2021.727590] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/05/2021] [Indexed: 12/25/2022] Open
Abstract
Vascular dementia (VaD) constitutes the second most prevalent cause of dementia in the world after Alzheimer’s disease (AD). VaD regroups heterogeneous neurological conditions in which the decline of cognitive functions, including executive functions, is associated with structural and functional alterations in the cerebral vasculature. Among these cerebrovascular disorders, major stroke, and cerebral small vessel disease (cSVD) constitute the major risk factors for VaD. These conditions alter neurovascular functions leading to blood-brain barrier (BBB) deregulation, neurovascular coupling dysfunction, and inflammation. Accumulation of neurovascular impairments over time underlies the cognitive function decline associated with VaD. Furthermore, several vascular risk factors, such as hypertension, obesity, and diabetes have been shown to exacerbate neurovascular impairments and thus increase VaD prevalence. Importantly, air pollution constitutes an underestimated risk factor that triggers vascular dysfunction via inflammation and oxidative stress. The review summarizes the current knowledge related to the pathological mechanisms linking neurovascular impairments associated with stroke, cSVD, and vascular risk factors with a particular emphasis on air pollution, to VaD etiology and progression. Furthermore, the review discusses the major challenges to fully elucidate the pathobiology of VaD, as well as research directions to outline new therapeutic interventions.
Collapse
Affiliation(s)
- Sarah Lecordier
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Daniel Manrique-Castano
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Yara El Moghrabi
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Ayman ElAli
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| |
Collapse
|
32
|
Shen Y, Yao MJ, Su YX, Xu DS, Wang J, Wang GR, Cui JJ, Zhang JL, Bai WZ. Histochemistry of microinfarcts in the mouse brain after injection of fluorescent microspheres into the common carotid artery. Neural Regen Res 2021; 17:832-837. [PMID: 34472483 PMCID: PMC8530124 DOI: 10.4103/1673-5374.322470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The mouse model of multiple cerebral infarctions, established by injecting fluorescent microspheres into the common carotid artery, is a recent development in animal models of cerebral ischemia. To investigate its effectiveness, mouse models of cerebral infarction were created by injecting fluorescent microspheres, 45–53 µm in diameter, into the common carotid artery. Six hours after modeling, fluorescent microspheres were observed directly through a fluorescence stereomicroscope, both on the brain surface and in brain sections. Changes in blood vessels, neurons and glial cells associated with microinfarcts were examined using fluorescence histochemistry and immunohistochemistry. The microspheres were distributed mainly in the cerebral cortex, striatum and hippocampus ipsilateral to the side of injection. Microinfarcts were found in the brain regions where the fluorescent microspheres were present. Here the lodged microspheres induced vascular and neuronal injury and the activation of astroglia and microglia. These histopathological changes indicate that this animal model of multiple cerebral infarctions effectively simulates the changes of various cell types observed in multifocal microinfarcts. This model is an effective, additional tool to study the pathogenesis of ischemic stroke and could be used to evaluate therapeutic interventions. This study was approved by the Animal Ethics Committee of the Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences (approval No. D2021-03-16-1) on March 16, 2021.
Collapse
Affiliation(s)
- Yi Shen
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ming-Jiang Yao
- Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences; Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Beijing, China
| | - Yu-Xin Su
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dong-Sheng Xu
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jia Wang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guang-Rui Wang
- Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences; Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Beijing, China
| | - Jing-Jing Cui
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jian-Liang Zhang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wan-Zhu Bai
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
33
|
Quintana DD, Anantula Y, Garcia JA, Engler-Chiurazzi EB, Sarkar SN, Corbin DR, Brown CM, Simpkins JW. Microvascular degeneration occurs before plaque onset and progresses with age in 3xTg AD mice. Neurobiol Aging 2021; 105:115-128. [PMID: 34062487 PMCID: PMC9703920 DOI: 10.1016/j.neurobiolaging.2021.04.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 01/06/2023]
Abstract
Heart disease and vascular disease positively correlate with the incidence of Alzheimer's disease (AD). Although there is ostensible involvement of dysfunctional cerebrovasculature in AD pathophysiology, the characterization of the specific changes and development of vascular injury during AD remains unclear. In the present study, we established a time-course for the structural changes and degeneration of the angioarchitecture in AD. We used cerebrovascular corrosion cast and µCT imaging to evaluate the geometry, topology, and complexity of the angioarchitecture in the brain of wild type and 3xTg AD mice. We hypothesized that changes to the microvasculature occur early during the disease, and these early identifiable aberrations would be more prominent in the brain subregions implicated in the cognitive decline of AD. Whole-brain analysis of the angioarchitecture indicated early morphological abnormalities and degeneration of microvascular networks in 3xTg AD mice. Our analysis of the hippocampus and cortical subregions revealed microvascular degeneration with onset and progression that was subregion dependent.
Collapse
Affiliation(s)
- Dominic D Quintana
- Department of Neuroscience, Center of Basic and Translational Stroke Research Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV
| | - Yamini Anantula
- Department of Neuroscience, Center of Basic and Translational Stroke Research Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV
| | - Jorge A Garcia
- Department of Neuroscience, Center of Basic and Translational Stroke Research Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV
| | - Elizabeth B Engler-Chiurazzi
- Department of Neuroscience, Center of Basic and Translational Stroke Research Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV
| | - Saumyendra N Sarkar
- Department of Neuroscience, Center of Basic and Translational Stroke Research Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV
| | - Deborah R Corbin
- Department of Neuroscience, Center of Basic and Translational Stroke Research Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV
| | - Candice M Brown
- Department of Neuroscience, Center of Basic and Translational Stroke Research Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV
| | - James W Simpkins
- Department of Neuroscience, Center of Basic and Translational Stroke Research Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV.
| |
Collapse
|
34
|
Localizing Microemboli within the Rodent Brain through Block-Face Imaging and Atlas Registration. eNeuro 2021; 8:ENEURO.0216-21.2021. [PMID: 34272259 PMCID: PMC8342264 DOI: 10.1523/eneuro.0216-21.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/27/2021] [Accepted: 07/07/2021] [Indexed: 12/01/2022] Open
Abstract
Brain microinfarcts are prevalent in humans, however because of the inherent difficulty of identifying and localizing individual microinfarcts, brain-wide quantification is impractical. In mice, microinfarcts have been created by surgically introducing microemboli into the brain, but a major limitation of this model is the absence of automated methods to identify and localize individual occlusions. We present a novel and semi-automated workflow to identify the anatomic location of fluorescent emboli (microspheres) within the mouse brain through histologic processing and atlas registration. By incorporating vibratome block-face imaging with the QuickNII brain registration tool, we show that the anatomic location of microspheres can be accurately registered to brain structures within the Allen mouse brain (AMB) atlas (e.g., somatomotor areas, hippocampal region, visual areas, etc.). Compared with registering images of slide mounted sections to the AMB atlas, microsphere location was more accurately determined when block-face images were used. As a proof of principle, using this workflow we compared the distribution of microspheres within the brains of mice that were either perfused or immersion fixed. No significant effect of perfusion on total microsphere number or location was detected. In general, microspheres were distributed brain-wide, with the largest density found in the thalamus. In sum, our block-face imaging workflow enables efficient characterization of the widespread distribution of fluorescent microemboli, facilitating future investigation into the impact of microinfarct load and location on brain health.
Collapse
|
35
|
Benenati S, Canale C, De Marzo V, Della Bona R, Rosa GM, Porto I. Atrial fibrillation and Alzheimer's disease: A conundrum. Eur J Clin Invest 2021; 51:e13451. [PMID: 33219514 DOI: 10.1111/eci.13451] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/04/2020] [Accepted: 11/15/2020] [Indexed: 12/01/2022]
Abstract
During ageing, the prevalence of Alzheimer's disease (AD) and of cardiovascular disease CVD) increases. Our aim is to investigate the relationship between AD and CVD and its risk factors, with a view to explaining the underlying mechanisms of this association. This review is based on the material obtained via MEDLINE (PubMed), Embase and Clinical Trials databases, from January 1980 until May 2019. The search term used was "Alzheimer's disease", combined with "cardiovascular disease", "hypertension", "dyslipidaemia", "diabetes mellitus", "atrial fibrillation", "coronary artery disease", "heart valve disease", "heart failure". Out of the 1328 papers initially retrieved, 431 duplicates and 216 records in languages other than English were removed; thus, only 98 papers were included in our research material. We have found that AD and CVD are frequently associated, while both of them, alone may be considered deleterious to health, the study of their combination constitutes a clinical challenge. Further research will help to clarify the real impact of CVD and its risk factors on AD, in order to better comprehend the effects of subclinical and clinical cardiovascular diseases on the brain. It may be hypothesized that there are various mechanisms underlying the association between AD and CVD, the main ones being: hypoperfusion and emboli, atherosclerosis, furthermore in both the heart and brain of AD patients, amyloid deposits may be present, thus causing damage to these organs. We need to clarify the real impact of these underlying hypothesized mechanisms and to investigate gender issues.
Collapse
Affiliation(s)
- Stefano Benenati
- Cardiovascular Disease Unit, IRCCS Policlinic Hospital San Martino, IRCCS Italian Cardiovascular Network, Genova, Italy.,Department of Internal Medicine, University of Genoa, Genova, Italy
| | - Claudia Canale
- Cardiovascular Disease Unit, IRCCS Policlinic Hospital San Martino, IRCCS Italian Cardiovascular Network, Genova, Italy.,Department of Internal Medicine, University of Genoa, Genova, Italy
| | - Vincenzo De Marzo
- Cardiovascular Disease Unit, IRCCS Policlinic Hospital San Martino, IRCCS Italian Cardiovascular Network, Genova, Italy.,Department of Internal Medicine, University of Genoa, Genova, Italy
| | - Roberta Della Bona
- Cardiovascular Disease Unit, IRCCS Policlinic Hospital San Martino, IRCCS Italian Cardiovascular Network, Genova, Italy
| | - Gian Marco Rosa
- Cardiovascular Disease Unit, IRCCS Policlinic Hospital San Martino, IRCCS Italian Cardiovascular Network, Genova, Italy.,Department of Internal Medicine, University of Genoa, Genova, Italy
| | - Italo Porto
- Cardiovascular Disease Unit, IRCCS Policlinic Hospital San Martino, IRCCS Italian Cardiovascular Network, Genova, Italy.,Department of Internal Medicine, University of Genoa, Genova, Italy
| |
Collapse
|
36
|
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
|
37
|
Humphreys CA, Smith C, Wardlaw JM. Correlations in post-mortem imaging-histopathology studies of sporadic human cerebral small vessel disease: A systematic review. Neuropathol Appl Neurobiol 2021; 47:910-930. [PMID: 34037264 DOI: 10.1111/nan.12737] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/29/2021] [Accepted: 05/02/2021] [Indexed: 11/30/2022]
Abstract
AIMS Sporadic human cerebral small vessel disease (SVD) commonly causes stroke and dementia but its pathogenesis is poorly understood. There are recognised neuroimaging and histopathological features. However, relatively few studies have examined the relationship between the radiological and pathological correlates of SVD; better correlation would promote greater insight into the underlying biological changes. METHODS We performed a systematic review to collate and appraise the information derived from studies that correlated histological with neuroimaging-defined SVD lesions. We searched for studies describing post-mortem imaging and histological tissue examination in adults, extracted data from published studies, categorised the information and compiled this narrative. RESULTS We identified 38 relevant studies, including at least 1146 subjects, 342 of these with SVD: 29 studies focussed on neuroradiological white matter lesions (WML), six on microinfarcts and three on dilated perivascular spaces (PVS) and lacunes. The histopathology terminology was diverse with few robust definitions. Reporting and methodology varied widely between studies, precluding formal meta-analysis. PVS and 'oedema' were frequent findings in WML, being described in at least 94 and 18 radiological WML, respectively, in addition to myelin pallor. Histopathological changes extended beyond the radiological lesion margins in at least 33 radiological WML. At least 43 radiological lesions not seen pathologically and at least 178 histological lesions were not identified on imaging. CONCLUSIONS Histopathological assessment of human SVD is hindered by inconsistent methodological approaches and unstandardised definitions. The data from this systematic review will help to develop standardised definitions to promote consistency in human SVD research.
Collapse
Affiliation(s)
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK.,Row Fogo Centre for Research into Ageing and the Brain, Edinburgh, UK
| |
Collapse
|
38
|
Schmid F, Conti G, Jenny P, Weber B. The severity of microstrokes depends on local vascular topology and baseline perfusion. eLife 2021; 10:60208. [PMID: 34003107 PMCID: PMC8421069 DOI: 10.7554/elife.60208] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 05/17/2021] [Indexed: 01/26/2023] Open
Abstract
Cortical microinfarcts are linked to pathologies like cerebral amyloid angiopathy and dementia. Despite their relevance for disease progression, microinfarcts often remain undetected and the smallest scale of blood flow disturbance has not yet been identified. We employed blood flow simulations in realistic microvascular networks from the mouse cortex to quantify the impact of single-capillary occlusions. Our simulations reveal that the severity of a microstroke is strongly affected by the local vascular topology and the baseline flow rate in the occluded capillary. The largest changes in perfusion are observed in capillaries with two inflows and two outflows. This specific topological configuration only occurs with a frequency of 8%. The majority of capillaries have one inflow and one outflow and is likely designed to efficiently supply oxygen and nutrients. Taken together, microstrokes bear potential to induce a cascade of local disturbances in the surrounding tissue, which might accumulate and impair energy supply locally.
Collapse
Affiliation(s)
- Franca Schmid
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Institute of Fluid Dynamics, ETH Zurich, Zurich, Switzerland
| | - Giulia Conti
- Institute of Fluid Dynamics, ETH Zurich, Zurich, Switzerland
| | - Patrick Jenny
- Institute of Fluid Dynamics, ETH Zurich, Zurich, Switzerland
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
39
|
Kitamura E, Iizuka T, Mamorita N, Nagai T, Usui R, Yanagida A, Nakamura M, Ishima D, Kaneko J, Akutsu T, Kanazawa N, Nishiyama K. Asymptomatic acute ischemic lesions in intracerebral hemorrhage: Its frequency, MRI features, and risk factors. J Neurol Sci 2021; 426:117472. [PMID: 33962325 DOI: 10.1016/j.jns.2021.117472] [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: 02/11/2021] [Revised: 04/22/2021] [Accepted: 04/27/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Asymptomatic acute ischemic lesions (AIL) may be coincidentally found on brain magnetic resonance imaging (MRI) obtained during the acute phase of intracerebral hemorrhage, but its clinical significance has yet to be determined. The objective of this study is to determine the frequency of asymptomatic AIL, its characteristic features of brain MRI and risk factors in patients with acute intracerebral hemorrhage. METHODS We retrospectively reviewed the clinical information of 108 patients with intracerebral hemorrhage who underwent brain MRIs within 30 days of hospitalization between April 2013 and January 2018. We determined the frequency of asymptomatic AIL, its brain MRI features, and risk factors. RESULTS AIL was found in 26 of 108 patients; symptomatic in 2 and asymptomatic in 24 (22.2%). Asymptomatic AIL were small, multiple, mainly distributed to the white matter in the anterior circulation (22/24, 91.7%), and occasionally seen in deep watershed areas (15/24, 62.5%). Only 2 patients had severe major vessel stenosis. Asymptomatic AIL was associated with high mean blood pressure (BP) on admission (> 145 mmHg), excessive drug-induced reduction in mean BP (≥ 55 mmHg), and large hemorrhage (> 31 mL in volume). CONCLUSIONS Asymptomatic AIL were found in 22.2% of patients with intracerebral hemorrhage within 30 days of hospitalization. Asymptomatic AIL were often small, multiple and occasionally developed in deep watershed areas despite the absence of major vessel stenosis. High mean BP on admission, excessive drug-induced BP reduction, and larger hemorrhage may be a risk factor for development of asymptomatic AIL.
Collapse
Affiliation(s)
- Eiji Kitamura
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan.
| | - Takahiro Iizuka
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Noritaka Mamorita
- Department of Medical Informatics, Kitasato University School of Allied Health Sciences, Sagamihara, Japan
| | - Toshiyuki Nagai
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Ryo Usui
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Atsuko Yanagida
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Masaaki Nakamura
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Daisuke Ishima
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Juntaro Kaneko
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Tsugio Akutsu
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Naomi Kanazawa
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kazutoshi Nishiyama
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| |
Collapse
|
40
|
Scheffler M, Salomir R, Maturana E, Montandon ML, Kövari EV, Haller S. Identification of hippocampal cortical microinfarcts on postmortem 3-T magnetic resonance imaging. Neuroradiology 2021; 63:1569-1573. [PMID: 33909116 PMCID: PMC8376696 DOI: 10.1007/s00234-021-02717-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/12/2021] [Indexed: 11/29/2022]
Abstract
Cortical microinfarcts (CMI) are increasingly recognized in the neurological community as a biomarker related to cognitive impairment and dementia. If their radiological depiction has been largely described in experimental settings using ultra-high-field magnetic resonance imaging (MRI), less is known about their visibility on routinely used 3-T MRI. In this radiologic-pathologic correlation study, using 3-T post-mortem MRI, we searched for hippocampal CMI, in a double-blinded fashion, and found that only 4/36, or 11%, were clearly demonstrated on both radiological and histopathological exams.
Collapse
Affiliation(s)
- Max Scheffler
- Division of Radiology, Geneva University Hospitals, Chemin du Pont-Bochet 3, 1226, Thonex, Switzerland.
| | - Rares Salomir
- Image Guided Interventions Laboratory, University of Geneva, Rue Gabrielle-Perret-Gentil 4, 1205, Geneva, Switzerland.,Division of Radiology, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 4, 1205, Geneva, Switzerland
| | - Enrique Maturana
- Division of Radiology, Geneva University Hospitals, Chemin du Pont-Bochet 3, 1226, Thonex, Switzerland
| | - Marie-Louise Montandon
- Department of Rehabilitation and Geriatrics, Geneva University Hospitals, Route de Loëx 151, 1233, Bernex, Switzerland.,Department of Psychiatry, Geneva University Hospitals, Chemin du Petit Bel-Air 2, 1225, Chene-Bourg, Switzerland
| | - Enikö V Kövari
- Department of Psychiatry, Geneva University Hospitals, Chemin du Petit Bel-Air 2, 1225, Chene-Bourg, Switzerland
| | - Sven Haller
- CIMC - Centre d'Imagerie Médicale de Cornavin, Place de Cornavin 18, 1201, Geneva, Switzerland.,Radiology, Department of Surgical Sciences, Uppsala University, 75236, Sjukhusvägen, Sweden
| |
Collapse
|
41
|
Kwapong WR, Yan Y, Hao Z, Wu B. Reduced Superficial Capillary Density in Cerebral Infarction Is Inversely Correlated With the NIHSS Score. Front Aging Neurosci 2021; 13:626334. [PMID: 33716714 PMCID: PMC7947804 DOI: 10.3389/fnagi.2021.626334] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/02/2021] [Indexed: 02/05/2023] Open
Abstract
Purpose: The retina and the brain share similar neuronal and microvascular features, therein we aimed to assess the structural and microvascular changes in the macula and choriocapillaris (CC) in patients with cerebral infarction when compared with healthy controls using optical coherence tomography angiography (OCTA). Methods: OCTA was used to image and measure the capillary density in the radial peripapillary capillaries (RPC), superficial capillary plexus (SCP), deep capillary plexus (DCP), choriocapillaris (CC), and mean area of the foveal avascular zone (FAZ) in all participants. Twenty-two cerebral infarction patients based on their magnetic resonance imaging (MRI) and 25 healthy controls were included in our study. Results: Density of the RPC (P < 0.001), SCP (P = 0.001), DCP (P < 0.001) and CC (P < 0.001) were significantly reduced in cerebral infarction patients when compared with healthy controls, respectively. Retinal thickness measurements (P < 0.05) were significantly reduced in cerebral infarction patients when compared with healthy controls. The mean FAZ area was significantly larger (P = 0.012) in cerebral infarction patients when compared with healthy controls. National Institute of HealthStroke Scale (NIHSS) inversely correlated with SCP density in cerebral infarction patients (Rho = −0.409, P = 0.001). Receiver operating characteristics curve analysis showed that the blood flow of the choriocapillaris had the highest index [area under the receiver operatingcharacteristic (AUROC) = 0.964] to discriminate cerebral infarction patients from the healthy controls. Conclusions: Our study suggests that cerebral microcirculation dysfunction which occurs in cerebral infarction is mirrored in the macula and choroidal microcirculation. OCTA has the potential to non-invasively characterize the macula and choroidal changes in cerebral infarction in vivo.
Collapse
Affiliation(s)
| | - Yuying Yan
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Zilong Hao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Wu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
42
|
Ter Telgte A, Wiegertjes K, Gesierich B, Baskaran BS, Marques JP, Kuijf HJ, Norris DG, Tuladhar AM, Duering M, de Leeuw FE. Temporal Dynamics of Cortical Microinfarcts in Cerebral Small Vessel Disease. JAMA Neurol 2021; 77:643-647. [PMID: 32065609 DOI: 10.1001/jamaneurol.2019.5106] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Importance Neuropathology studies show a high prevalence of cortical microinfarcts (CMIs) in aging individuals, especially in patients with cerebrovascular disease and dementia. However, most, are invisible on T1- and T2-weighted magnetic resonance imaging (MRI), raising the question of how to explain this mismatch. Studies on small acute infarcts, detected on diffusion-weighted imaging (DWI), suggest that infarcts are largest in their acute phase and reduce in size thereafter. Therefore, we hypothesized that a subset of the CMI that are invisible on MRI can be detected on MRI in their acute phase. However, to our knowledge, a serial imaging study investigating the temporal dynamics of acute CMI (A-CMI) is lacking. Objective To determine the prevalence of chronic CMI (C-CMI) and the cumulative incidence and temporal dynamics of A-CMI in individuals with cerebral small vessel disease (SVD). Design, Setting, Participants and Exposures The RUN DMC-Intense study is a single-center hospital-based prospective cohort study on SVD performed between March 2016 and November 2017 and comprising 10 monthly 3-T MRI scans, including high-resolution DWI, 3-dimensional T1, 3-dimensional fluid-attenuated inversion recovery, and T2. One hundred six individuals from the previous longitudinal RUN DMC study were recruited based on the presence of progression of white matter hyperintensities on MRI between 2006 and 2015 and exclusion of causes of cerebral ischemia other than SVD. Fifty-four individuals (50.9%) participated. The median total follow-up duration was 39.5 weeks (interquartile range, 37.8-40.3). Statistical data analysis was performed between May and October 2019. Main Outcomes and Measures We determined the prevalence of C-CMI using the baseline T1, fluid-attenuated inversion recovery, and T2 scans. Monthly high-resolution DWI scans (n = 472) were screened to determine the cumulative incidence of A-CMI. The temporal dynamics of A-CMI were determined based on the MRI scans collected during the first follow-up visit after A-CMI onset and the last available follow-up visit. Results The median age of the cohort at baseline MRI was 69 years (interquartile range, 66-74 years) and 34 participants (63%) were men. The prevalence of C-CMI was 35% (95% CI, 0.24-0.49). Monthly DWI detected 21 A-CMI in 7 of 54 participants, resulting in a cumulative incidence of 13% (95% CI, 0.06-0.24). All A-CMI disappeared on follow-up MRI. Conclusions and Relevance Acute CMI never evolved into chronically MRI-detectable lesions. We suggest that these A-CMI underlie part of the submillimeter C-CMI encountered on neuropathological examination and thereby provide a source for the high CMI burden on neuropathology.
Collapse
Affiliation(s)
- Annemieke Ter Telgte
- Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Kim Wiegertjes
- Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Benno Gesierich
- Institute for Stroke and Dementia Research (ISD), University Hospital LMU Munich, Munich, Germany
| | - Brendon Sri Baskaran
- Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - José P Marques
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, Nijmegen, the Netherlands
| | - Hugo J Kuijf
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands
| | - David G Norris
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, Nijmegen, the Netherlands
| | - Anil M Tuladhar
- Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marco Duering
- Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Radboud University Medical Center, Nijmegen, the Netherlands.,Institute for Stroke and Dementia Research (ISD), University Hospital LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Frank-Erik de Leeuw
- Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
43
|
Hilal S, Doolabi A, Vrooman H, Ikram MK, Ikram MA, Vernooij MW. Clinical Relevance of Cortical Cerebral Microinfarcts on 1.5T Magnetic Resonance Imaging in the Late-Adult Population. Stroke 2021; 52:922-930. [PMID: 33535785 DOI: 10.1161/strokeaha.120.032085] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Cortical cerebral microinfarcts (CMIs) have been linked with dementia and impaired cognition in cross-sectional studies. However, the clinical relevance of CMIs in a large population-based setting is lacking. We examine the association of cortical CMIs detected on 1.5T magnetic resonance imaging with cardiovascular risk factors, cerebrovascular disease, and brain tissue volumes. We further explore the association between cortical CMIs with cognitive decline and risk of stroke, dementia, and mortality in the general population. METHODS Two thousand one hundred fifty-six participants (age: 75.7±5.9 years, women: 55.6%) with clinical history and baseline magnetic resonance imaging (January 2009-December 2013) were included from the Rotterdam Study. Cortical CMIs were graded based on a previously validated method. Markers of cerebrovascular disease and brain tissue volumes were assessed on magnetic resonance imaging. Cognition was assessed using a detailed neuropsychological test at baseline and at 5 years of follow-up. Data on incident stroke, dementia, and mortality were included until January 2016. RESULTS Two hundred twenty-seven individuals (10.5%) had ≥1 cortical CMIs. The major risk factors of cortical CMIs were male sex, current smoking, history of heart disease, and stroke. Furthermore, presence of cortical CMIs was associated with infarcts and smaller brain volume. Persons with cortical CMIs showed cognitive decline in Stroop tests (color-naming and interference subtasks; β for color-naming, 0.18 [95% CI, 0.04-0.33], P interaction ≤0.001 and β for interference subtask, 1.74, [95% CI, 0.66-2.82], P interaction ≤0.001). During a mean follow-up of 5.2 years, 73 (4.3%) individuals developed incident stroke, 95 (5.1%) incident dementia, and 399 (19.2%) died. People with cortical CMIs were at an increased risk of stroke (hazard ratio, 1.18 [95% CI, 1.09-1.28]) and mortality (hazard ratio, 1.09 [95% CI, 1.00-1.19]). CONCLUSIONS Cortical CMIs are highly prevalent in a population-based setting and are associated with cardiovascular disease, cognitive decline, and increased risk of stroke and mortality. Future investigations will have to show whether cortical CMIs are a useful biomarker to intervene upon to reduce the burden of stroke.
Collapse
Affiliation(s)
- Saima Hilal
- Department of Radiology and Nuclear Medicine (S.H., M.A.I., M.W.V.), Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Epidemiology (S.H., A.D., M.K.I., M.A.I., M.W.V.), Erasmus Medical Center, Rotterdam, the Netherlands.,Saw Swee Hock School of Public Health, National University of Singapore and National University Health System (S.H.)
| | - Arwin Doolabi
- Department of Epidemiology (S.H., A.D., M.K.I., M.A.I., M.W.V.), Erasmus Medical Center, Rotterdam, the Netherlands
| | - Henri Vrooman
- Departments of Radiology and Medical Informatics (H.V.), Erasmus Medical Center, Rotterdam, the Netherlands
| | - M Kamran Ikram
- Department of Epidemiology (S.H., A.D., M.K.I., M.A.I., M.W.V.), Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Neurology (M.K.I.), Erasmus Medical Center, Rotterdam, the Netherlands
| | - M Arfan Ikram
- Department of Radiology and Nuclear Medicine (S.H., M.A.I., M.W.V.), Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Epidemiology (S.H., A.D., M.K.I., M.A.I., M.W.V.), Erasmus Medical Center, Rotterdam, the Netherlands
| | - Meike W Vernooij
- Department of Radiology and Nuclear Medicine (S.H., M.A.I., M.W.V.), Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Epidemiology (S.H., A.D., M.K.I., M.A.I., M.W.V.), Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
44
|
Zhang L, Biessels GJ, Hilal S, Chong JSX, Liu S, Shim HY, Xu X, Chong EJY, Wong ZX, Loke YM, Venketasubramanian N, Yeow TB, Chen CLH, Zhou JH. Cerebral microinfarcts affect brain structural network topology in cognitively impaired patients. J Cereb Blood Flow Metab 2021; 41:105-115. [PMID: 31986957 PMCID: PMC7747167 DOI: 10.1177/0271678x20902187] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cerebral microinfarcts (CMIs), a novel cerebrovascular marker, are prevalent in Alzheimer's disease (AD) and associated with cognitive impairment. Nonetheless, the underlying mechanism of how CMIs influence cognition remains uncertain. We hypothesized that cortical-CMIs disrupted structural connectivity in the higher-order cognitive networks, leading to cognitive impairment. We analyzed diffusion-MRI data of 92 AD (26 with cortical-CMIs) and 110 cognitive impairment no dementia patients (CIND, 28 with cortical-CMIs). We compared structural network topology between groups with and without cortical-CMIs in AD/CIND, and tested whether structural connectivity mediated the association between cortical-CMIs and cognition. Cortical-CMIs correlated with impaired structural network topology (i.e. lower efficiency/degree centrality in the executive control/dorsal attention networks in CIND, and lower clustering coefficient in the default mode/dorsal attention networks in AD), which mediated the association of cortical-CMIs with visuoconstruction dysfunction. Our findings provide the first in vivo human evidence that cortical-CMIs impair cognition in elderly via disrupting structural connectivity.
Collapse
Affiliation(s)
- Liwen Zhang
- Centre for Cognitive Neuroscience, Neuroscience and Behavioural Disorders Program, Duke-National University of Singapore Medical School, Singapore.,Department of Pharmacology, National University of Singapore, Singapore.,Memory Ageing and Cognition Centre, National University Health System, Singapore
| | - Geert Jan Biessels
- Department of Neurology, Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Saima Hilal
- Department of Pharmacology, National University of Singapore, Singapore.,Memory Ageing and Cognition Centre, National University Health System, Singapore
| | - Joanna Su Xian Chong
- Centre for Cognitive Neuroscience, Neuroscience and Behavioural Disorders Program, Duke-National University of Singapore Medical School, Singapore.,Centre for Sleep and Cognition, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Siwei Liu
- Centre for Cognitive Neuroscience, Neuroscience and Behavioural Disorders Program, Duke-National University of Singapore Medical School, Singapore.,Centre for Sleep and Cognition, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Hee Youn Shim
- Centre for Cognitive Neuroscience, Neuroscience and Behavioural Disorders Program, Duke-National University of Singapore Medical School, Singapore
| | - Xin Xu
- Department of Pharmacology, National University of Singapore, Singapore.,Memory Ageing and Cognition Centre, National University Health System, Singapore
| | - Eddie Jun Yi Chong
- Department of Pharmacology, National University of Singapore, Singapore.,Memory Ageing and Cognition Centre, National University Health System, Singapore
| | - Zi Xuen Wong
- Department of Pharmacology, National University of Singapore, Singapore.,Memory Ageing and Cognition Centre, National University Health System, Singapore
| | - Yng Miin Loke
- Centre for Cognitive Neuroscience, Neuroscience and Behavioural Disorders Program, Duke-National University of Singapore Medical School, Singapore
| | | | | | - Christopher Li-Hsian Chen
- Department of Pharmacology, National University of Singapore, Singapore.,Memory Ageing and Cognition Centre, National University Health System, Singapore
| | - Juan Helen Zhou
- Centre for Cognitive Neuroscience, Neuroscience and Behavioural Disorders Program, Duke-National University of Singapore Medical School, Singapore.,Centre for Sleep and Cognition, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Clinical Imaging Research Centre, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
45
|
Polymeris AA, Coslovksy M, Aeschbacher S, Sinnecker T, Benkert P, Kobza R, Beer J, Rodondi N, Fischer U, Moschovitis G, Monsch AU, Springer A, Schwenkglenks M, Wuerfel J, De Marchis GM, Lyrer PA, Kühne M, Osswald S, Conen D, Kuhle J, Bonati LH. Serum neurofilament light in atrial fibrillation: clinical, neuroimaging and cognitive correlates. Brain Commun 2021; 2:fcaa166. [PMID: 33381755 PMCID: PMC7753055 DOI: 10.1093/braincomms/fcaa166] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/09/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022] Open
Abstract
Emerging evidence suggests that atrial fibrillation is associated with cognitive dysfunction independently of stroke, but the underlying mechanisms remain unclear. In this cross-sectional analysis from the Swiss-atrial fibrillation Study (NCT02105844), we investigated the association of serum neurofilament light protein, a neuronal injury biomarker, with (i) the CHA2DS2-VASc score (congestive heart failure, hypertension, age 65–74 or >75 years, diabetes mellitus, stroke or transient ischaemic attack, vascular disease, sex), clinical and neuroimaging parameters and (ii) cognitive measures in atrial fibrillation patients. We measured neurofilament light in serum using an ultrasensitive single-molecule array assay in a sample of 1379 atrial fibrillation patients (mean age, 72 years; female, 27%). Ischaemic infarcts, small vessel disease markers and normalized brain volume were assessed on brain MRI. Cognitive testing included the Montreal cognitive assessment, trail-making test, semantic verbal fluency and digit symbol substitution test, which were summarized using principal component analysis. Results were analysed using univariable and multivariable linear regression. Neurofilament light was associated with the CHA2DS2-VASc score, with an average 19.2% [95% confidence interval (17.2%, 21.3%)] higher neurofilament per unit CHA2DS2-VASc increase. This association persisted after adjustment for age and MRI characteristics. In multivariable analyses, clinical parameters associated with neurofilament light were higher age [32.5% (27.2%, 38%) neurofilament increase per 10 years], diabetes mellitus, heart failure and peripheral artery disease [26.8% (16.8%, 37.6%), 15.7% (8.1%, 23.9%) and 19.5% (6.8%, 33.7%) higher neurofilament, respectively]. Mean arterial pressure showed a curvilinear association with neurofilament, with evidence for both an inverse linear and a U-shaped association. MRI characteristics associated with neurofilament were white matter lesion volume and volume of large non-cortical or cortical infarcts [4.3% (1.8%, 6.8%) and 5.5% (2.5%, 8.7%) neurofilament increase per unit increase in log-volume of the respective lesion], as well as normalized brain volume [4.9% (1.7%, 8.1%) higher neurofilament per 100 cm3 smaller brain volume]. Neurofilament light was inversely associated with all cognitive measures in univariable analyses. The effect sizes diminished after adjusting for clinical and MRI variables, but the association with the first principal component was still evident. Our results suggest that in atrial fibrillation patients, neuronal loss measured by serum neurofilament light is associated with age, diabetes mellitus, heart failure, blood pressure and vascular brain lesions, and inversely correlates with normalized brain volume and cognitive function.
Collapse
Affiliation(s)
- Alexandros A Polymeris
- Department of Neurology and Stroke Center, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Michael Coslovksy
- Clinical Trial Unit, Department of Clinical Research, University Hospital Basel and University of Basel, Switzerland.,Cardiovascular Research Institute Basel (CRIB), Basel, Switzerland
| | - Stefanie Aeschbacher
- Cardiovascular Research Institute Basel (CRIB), Basel, Switzerland.,Cardiology Division, Department of Medicine, University Hospital Basel, Basel, Switzerland
| | - Tim Sinnecker
- Department of Neurology and Stroke Center, University Hospital Basel and University of Basel, Basel, Switzerland.,Medical Image Analysis Center (MIAC) AG and Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Pascal Benkert
- Clinical Trial Unit, Department of Clinical Research, University Hospital Basel and University of Basel, Switzerland
| | - Richard Kobza
- Department of Cardiology, Cantonal Hospital Lucerne, Lucerne, Switzerland
| | - Jürg Beer
- Department of Medicine, Cantonal Hospital Baden, Baden, Switzerland.,Molecular Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Nicolas Rodondi
- Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland.,Department of General Internal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Urs Fischer
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Giorgio Moschovitis
- Cardiology Division, Department of Medicine, EOC Ospedale Regionale di Lugano, Lugano, Switzerland
| | - Andreas U Monsch
- Memory Clinic, University Department of Geriatric Medicine Felix Platter and University of Basel, Basel, Switzerland
| | - Anne Springer
- Cardiovascular Research Institute Basel (CRIB), Basel, Switzerland.,Cardiology Division, Department of Medicine, University Hospital Basel, Basel, Switzerland
| | - Matthias Schwenkglenks
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland.,Institute of Pharmaceutical Medicine (ECPM), University of Basel, Basel, Switzerland
| | - Jens Wuerfel
- Medical Image Analysis Center (MIAC) AG and Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Gian Marco De Marchis
- Department of Neurology and Stroke Center, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Philippe A Lyrer
- Department of Neurology and Stroke Center, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Michael Kühne
- Cardiovascular Research Institute Basel (CRIB), Basel, Switzerland.,Cardiology Division, Department of Medicine, University Hospital Basel, Basel, Switzerland
| | - Stefan Osswald
- Cardiovascular Research Institute Basel (CRIB), Basel, Switzerland.,Cardiology Division, Department of Medicine, University Hospital Basel, Basel, Switzerland
| | - David Conen
- Cardiology Division, Department of Medicine, University Hospital Basel, Basel, Switzerland.,Population Health Research Institute, McMaster University, Hamilton, Canada
| | - Jens Kuhle
- Department of Neurology and Stroke Center, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Leo H Bonati
- Department of Neurology and Stroke Center, University Hospital Basel and University of Basel, Basel, Switzerland
| | | |
Collapse
|
46
|
Fujioka H, Urasaki E, Soejima Y, Harada H, Yamashita K. Combination of Single- and Paired-Pulse Somatosensory Evoked Potentials in Ischemic Monitoring: Preliminary Investigation in Carotid Endarterectomy. Cureus 2020; 12:e12206. [PMID: 33489615 PMCID: PMC7815265 DOI: 10.7759/cureus.12206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Introduction Severe ischemia induces cerebral excitability imbalance before completion of infarct. To investigate the clinical availability of this imbalance with ischemic monitoring, paired-pulse somatosensory evoked potentials (SEPs) were performed in conjunction with conventional SEPs during carotid endarterectomy. Methods For carotid endarterectomy patients with hemodynamic deficits of the middle cerebral artery area (n = 34), the excitability imbalances (Q) were measured by paired-pulse SEPs, wherein the second response (A2) was divided by the first (A1; Q = A2/A1). Regional cerebral saturation (rSO2) was also measured. Occlusion was performed twice using shunting. Results Each carotid occlusion induced a significant decrease in mean A1 and rSO2, and an increase in mean Q values (p < 0.001), which returned to the baseline level after occlusion. While neuronal imbalances were mostly transient, persistently increased Q values were observed in four cases (11.8%), all indicating postoperative abnormalities in diffusion-weighted magnetic resonance imaging (100%). Meanwhile, A1 detected the postoperative abnormality in only one case (25%). Preoperative Q values at the time of surgery were significantly higher in symptomatic patients having the upper limb deficits than those without (p < 0.01), indicating persistent or permanent imbalances. Conclusion Paired-pulse SEPs reliably identified transient, persistent or permanent neuronal imbalances, depending on the ischemic severity. These preliminary results indicated that paired-pulse SEPs, in combination with conventional SEPs (A1), may offer better ischemic monitoring.
Collapse
Affiliation(s)
- Hiroshi Fujioka
- Neurosurgery, Nagasaki Yurino Hospital, Nagasaki, JPN.,Neurosurgery, Kanmon Medical Center, National Hospital Organization (NHO), Shimonoseki, JPN.,Neurosurgery, Cognitive and Molecular Research Institute of Brain Diseases, Kurume University, Fukuoka, JPN
| | | | - Yoshiteru Soejima
- Neurosurgery, Kanmon Medical Center, National Hospital Organization (NHO), Shimonoseki, JPN
| | - Hideki Harada
- Anaesthesiology, Cognitive and Molecular Research Institute of Brain Diseases, Kurume University, Fukuoka, JPN
| | - Katsuhiro Yamashita
- Neurosurgery, Kanmon Medical Center, National Hospital Organization (NHO), Shimonoseki, JPN
| |
Collapse
|
47
|
Ii Y, Ishikawa H, Shindo A, Matsuyama H, Matsuura K, Matsuda K, Yoshimaru K, Satoh M, Kogue R, Umino M, Maeda M, Tomimoto H. Association between cortical microinfarcts and total small vessel disease burden in cerebral amyloid angiopathy on 3-Tesla magnetic resonance imaging. Eur J Neurol 2020; 28:794-799. [PMID: 33098163 DOI: 10.1111/ene.14610] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/19/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND PURPOSE Cortical microinfarcts (CMIs) are frequently found in the brains of patients with advanced cerebral amyloid angiopathy (CAA) at autopsy. The small vessel disease (SVD) score for CAA (i.e., the CAA-SVD score) has been proposed to evaluate the severity of CAA-associated vasculopathic changes by a combination of magnetic resonance imaging (MRI) markers. The aim of this study was to examine the association between total CAA-SVD score and features of CMIs on in vivo 3-Tesla MRI. METHODS Eighty patients with probable CAA were retrospectively analyzed. Lobar cerebral microbleeds, cortical superficial siderosis, enlargement of perivascular space in the centrum semiovale and white matter hyperintensity were collectively assessed, and the total CAA-SVD score was calculated. The presence of CMI was also examined. RESULTS Of the 80 patients, 13 (16.25%) had CMIs. CMIs were detected more frequently in the parietal and occipital lobes. A positive correlation was found between total CAA-SVD score and prevalence of CMI (ρ = 0.943; p = 0.005). Total CAA-SVD score was significantly higher in patients with CMIs than in those without (p = 0.009). In a multivariable logistic regression analysis, the presence of CMIs was significantly associated with total CAA-SVD score (odds ratio 2.318 [95% confidence interval 1.228-4.376]; p = 0.01, per each additional point). CONCLUSIONS The presence of CMIs with a high CAA-SVD score could be an indicator of more severe amyloid-associated vasculopathic changes in patients with probable CAA.
Collapse
Affiliation(s)
- Yuichiro Ii
- Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan
| | - Hidehiro Ishikawa
- Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan
| | - Hirofumi Matsuyama
- Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan
| | - Keita Matsuura
- Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan
| | - Kana Matsuda
- Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan
| | - Kimiko Yoshimaru
- Department of Dementia Prevention and Therapeutics, Mie University Graduate School of Medicine, Mie, Japan
| | - Masayuki Satoh
- Department of Dementia Prevention and Therapeutics, Mie University Graduate School of Medicine, Mie, Japan
| | - Ryota Kogue
- Department of Radiology, Mie University Graduate School of Medicine, Mie, Japan
| | - Maki Umino
- Department of Radiology, Mie University Graduate School of Medicine, Mie, Japan
| | - Masayuki Maeda
- Department of Neuroradiology, Mie University Graduate School of Medicine, Mie, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Mie, Japan
| |
Collapse
|
48
|
Cerebral micro-infarcts; the hidden missing link to vascular cognitive decline. J Neurol Sci 2020; 420:117171. [PMID: 33268082 DOI: 10.1016/j.jns.2020.117171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 11/23/2022]
|
49
|
Wei Y, Pu Y, Pan Y, Nie X, Duan W, Liu D, Yan H, Lu Q, Zhang Z, Yang Z, Wen M, Gu W, Hou X, Ma N, Leng X, Miao Z, Liu L. Cortical Microinfarcts Associated With Worse Outcomes in Patients With Acute Ischemic Stroke Receiving Endovascular Treatment. Stroke 2020; 51:2742-2751. [PMID: 32811382 DOI: 10.1161/strokeaha.120.030895] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE We aimed to evaluate the impact of cortical microinfarcts (CMIs) on functional outcome after endovascular treatment in patients with acute ischemic stroke. METHODS In a multicenter registration study for RESCUE-RE (a registration study for Critical Care of Acute Ischemic Stroke After Recanalization), eligible patients with large vessel occlusion stroke receiving endovascular treatment, who had undergone 3T magnetic resonance imaging on admission or within 24 hours after endovascular treatment were analyzed. We evaluated the presence and numbers of CMIs with assessment of axial T1, T2-weighted images, and fluid-attenuated inversion recovery images. The primary outcome was functional dependence or death defined as modified Rankin Scale scores of 3 to 6 at 90 days. Secondary outcomes included early neurological improvement, any intracranial hemorrhage, symptomatic intracranial hemorrhage, and mortality. We investigated the independent associations of CMIs with the outcomes using multivariable logistic regression in overall patients and in subgroups. RESULTS Among 414 patients (enrolled from July 2018 to May 2019) included in the analyses, 96 (23.2%) patients had at least one CMI (maximum 6). Patients with CMI(s) were more likely to be functionally dependent or dead at 90 days, compared with those without (55.2% versus 37.4%; P<0.01). In multivariable logistic regression analyses, presence of CMI(s) (adjusted odds ratio, 1.78 [95% CI, 1.04-3.07]; P=0.04) and multiple CMIs (CMIs ≥2; adjusted odds ratio, 7.41 [95% CI, 2.48-22.17]; P<0.001) were independently, significantly associated with the primary outcome. There was no significant difference between subgroups in the associations between CMI presence and the primary outcome. CONCLUSIONS Acute large vessel occlusion stroke patients receiving endovascular treatment with CMI(s) were more likely to have a poor functional outcome at 90 days, independent of patients' characteristics. Such associations may be dose-dependent. Registration: URL: http://www.chictr.org.cn; Unique identifier: ChiCTR1900022154.
Collapse
Affiliation(s)
- Yufei Wei
- Department of Neurology (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.), Beijing Tiantan Hospital, Capital Medical University, China.,China National Clinical Research Center for Neurological Diseases, Beijing (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.)
| | - Yuehua Pu
- Department of Neurology (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.), Beijing Tiantan Hospital, Capital Medical University, China.,China National Clinical Research Center for Neurological Diseases, Beijing (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.)
| | - Yuesong Pan
- Department of Neurology (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.), Beijing Tiantan Hospital, Capital Medical University, China.,China National Clinical Research Center for Neurological Diseases, Beijing (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.)
| | - Ximing Nie
- Department of Neurology (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.), Beijing Tiantan Hospital, Capital Medical University, China.,China National Clinical Research Center for Neurological Diseases, Beijing (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.)
| | - Wanying Duan
- Department of Neurology (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.), Beijing Tiantan Hospital, Capital Medical University, China.,China National Clinical Research Center for Neurological Diseases, Beijing (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.)
| | - Dacheng Liu
- Department of Neurology (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.), Beijing Tiantan Hospital, Capital Medical University, China.,China National Clinical Research Center for Neurological Diseases, Beijing (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.)
| | - Hongyi Yan
- Department of Neurology (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.), Beijing Tiantan Hospital, Capital Medical University, China.,China National Clinical Research Center for Neurological Diseases, Beijing (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.)
| | - Qixuan Lu
- Department of Neurology (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.), Beijing Tiantan Hospital, Capital Medical University, China.,China National Clinical Research Center for Neurological Diseases, Beijing (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.)
| | - Zhe Zhang
- Department of Neurology (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.), Beijing Tiantan Hospital, Capital Medical University, China.,China National Clinical Research Center for Neurological Diseases, Beijing (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.)
| | - Zhonghua Yang
- Department of Neurology (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.), Beijing Tiantan Hospital, Capital Medical University, China.,China National Clinical Research Center for Neurological Diseases, Beijing (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.)
| | - Miao Wen
- Department of Neurology (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.), Beijing Tiantan Hospital, Capital Medical University, China.,China National Clinical Research Center for Neurological Diseases, Beijing (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.)
| | - Weibin Gu
- Department of Radiology (W.G., X.H.), Beijing Tiantan Hospital, Capital Medical University, China
| | - Xinyi Hou
- Department of Radiology (W.G., X.H.), Beijing Tiantan Hospital, Capital Medical University, China
| | - Ning Ma
- Department of Interventional Neurology (N.M., Z.M.), Beijing Tiantan Hospital, Capital Medical University, China
| | - Xinyi Leng
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong SAR, China (X.L.)
| | - Zhongrong Miao
- Department of Interventional Neurology (N.M., Z.M.), Beijing Tiantan Hospital, Capital Medical University, China
| | - Liping Liu
- Department of Neurology (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.), Beijing Tiantan Hospital, Capital Medical University, China.,China National Clinical Research Center for Neurological Diseases, Beijing (Y.W., Y. Pu, Y. Pan, X.N., W.D., D.L., H.Y., Q.L., Z.Z., Z.Y., M.W., L.L.)
| | | |
Collapse
|
50
|
Joseph CR. Novel MRI Techniques Identifying Vascular Leak and Paravascular Flow Reduction in Early Alzheimer Disease. Biomedicines 2020; 8:biomedicines8070228. [PMID: 32698354 PMCID: PMC7400582 DOI: 10.3390/biomedicines8070228] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/12/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
With beta amyloid and tau antibody treatment trial failures, avenues directed to other facets of the disease pathophysiology are being explored to treat in the preclinical or early clinical state. Clear evidence of blood–brain barrier (BBB) breakdown occurring early in the AD process has recently been established. Likewise, the glymphatic system regulating water and solute inflow and outflow in parallel with the vascular system is affected causing delayed clearance of fluid waste. Its dysfunction as a component of AD along with BBB leak are reasonable candidates to explore for future treatments. Ideally, human medication trials require a minimally invasive method of quantifying both improvements in BBB integrity and glymphatic fluid clearance correlated with clinical outcomes. We will review the known physiology and anatomy of the BBB system, and its relationship to the glymphatic system and the microglial surveillance system. Dysfunction of this tripart system occurring in preclinical Alzheimer disease (AD) will be reviewed along with existing MRI tools for identifying altered flow dynamics useful for monitoring improved functionality with future treatments. High-resolution dynamic contrast enhanced MRI imaging demonstrating BBB leak and the recently reported non-invasive 3D PASL MRI pilot study demonstrating significant delay in glymphatic clearance in AD subjects appear to be the best candidates.
Collapse
Affiliation(s)
- Charles R Joseph
- Department of Internal Medicine, Liberty University College of Osteopathic Medicine, Lynchburg, VA 24502, USA
| |
Collapse
|