1
|
Li Z, Miao L, Zhang T, Thomas AM, Li S. Causal relationship of inflammatory cytokines and serum metabolites in cerebral small vessel disease: a two-step Mendelian randomization study. Eur J Neurol 2024; 31:e16443. [PMID: 39150083 DOI: 10.1111/ene.16443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND AND PURPOSE The aim was to investigate the causal relationships of inflammatory cytokines and serum metabolites in cerebral small vessel disease (CSVD). METHODS Bidirectional Mendelian randomization was first conducted to screen inflammatory cytokines and serum metabolites that were associated with imaging features of CSVD, including white matter hyperintensities, recent small subcortical infarcts, cortical cerebral microinfarcts, cerebral microbleeds, lacunes and enlarged perivascular spaces. Sensitivity analyses were performed to evaluate the robustness and pleiotropy of these results. Subsequently, inflammatory cytokines and serum metabolites that were associated with CSVD were subjected to functional enrichment. Finally, mediation analysis was employed to investigate whether inflammatory cytokines or serum metabolites acted as an intermediary for the other in their causal relationship with CSVD. RESULTS Of the inflammatory cytokines, five were risk factors (e.g., tumour-necrosis-factor-related apoptosis-inducing ligand) and five (e.g., fibroblast growth factor 19) were protective factors for CSVD. Eleven serum metabolites that increased CSVD risk and 13 metabolites that decreased CSVD risk were also identified. The majority of these markers of CSVD susceptibility were lipid metabolites. Natural killer cell receptor sub-type 2B4 was determined to act as a mediating factor of an unidentified metabolite for the enlargement of perivascular spaces. CONCLUSION Several inflammatory cytokines and serum metabolites had causal relationships with imaging features of CSVD. A natural killer cell receptor mediated in part the promotional effect of a metabolite on perivascular space enlargement.
Collapse
Affiliation(s)
- Zidong Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Lu Miao
- Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Tianyi Zhang
- School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Aline M Thomas
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Yang Y, Yao Z, Huo L. Causal effect of circulating cytokines on MRI markers of cerebral small vessel disease: A mendelian randomization study. Cytokine 2024; 182:156713. [PMID: 39079216 DOI: 10.1016/j.cyto.2024.156713] [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: 02/16/2024] [Revised: 04/05/2024] [Accepted: 07/22/2024] [Indexed: 08/25/2024]
Abstract
BACKGROUND Previous observational studies have reported the correlation between circulating inflammatory cytokines and cerebral small vessel disease (CSVD). However, the causality of this association is uncertain. This study used Mendelian randomization to investigate the causal effect of circulating inflammatory cytokines on neuroimaging changes in CSVD. METHODS This study utilized genetic variances of 41 inflammatory cytokines and 3 neuroimaging markers of CSVD from genome-wide association studies to assess the causal effects in a two-sample Mendelian randomization approach. Inverse variance weighted analysis was used as the main analytical method, and sensitivity analysis was used to further validate the robustness of the results. RESULTS Increased IL-18 was associated with increased white matter hyperintensity (WMH) and mean diffusivity (MD) (β = 0.034, 95 % CI 0.002, 0.065, P=0.038, β = 0.157, 95 % CI 0.015, 0.299, P=0.030). However, increased IL-18 was associated with decreased fractional anisotropy (FA) (β = -0.141, 95 % CI -0.279, -0.002, P=0.047). Increased monocyte chemotactic protein-1(MCP-1) was associated with decreased FA (β = -0.278, 95 % CI -0.502, -0.054, P=0.015). Increased IL-10 levels and IL-2ra levels were associated with decreased risks of MD (β = -0.228, 95 % CI -0.448, -0.009, p = 0.041; β = -0.204, 95 % CI=-0.377, -0.031, p = 0.021). CONCLUSIONS This study revealed that increased levels of IL-18 and MCP-1 were associated with white matter microstructural injury, and increased levels of IL-10 and IL-2ra were associated with decreased MD.
Collapse
Affiliation(s)
- Yang Yang
- Central Laboratory, Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing 100038, China
| | - Zhichao Yao
- Central Laboratory, Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing 100038, China
| | - Lirong Huo
- Central Laboratory, Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing 100038, China.
| |
Collapse
|
3
|
Das P, Wang Y, Angom RS, Dredla B, Wang E, Jansen-West K, Badi M, Ross O, Meschia JF, Mukhopadhyay D. Changes in plasma concentrations of novel vascular and inflammatory biomarkers in obstructive sleep apnea patients pre- and post-stroke. Sleep Med 2024; 119:518-525. [PMID: 38805859 DOI: 10.1016/j.sleep.2024.05.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/19/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is increasingly recognized as a common condition in the general population and causes significant OSA-associated morbidities including cardiovascular and cerebrovascular events such as cerebral small vessel disease (CSVD) and stroke. METHODS In this study, using sensitive ELISA immunoassays, we measured subset of endothelial/vascular and inflammatory biomarkers as well as neurofilament light chain (NfL), a sensitive marker for neuroaxonal injury, using plasma from OSA patients post-stroke (Acute Cerebral Infarction (ACI), N = 26) to determine their usefulness as potential prognostic markers in disease progression. RESULTS Our results showed significantly increased plasma TNFα and NfL concentrations and decreased concentrations of platelet derived growth factor (PDGF-AA) in post-stroke OSA patients with more severe white matter hyperintensities (WMHs). And after separating the patients based on sex, compared to females, male post-stroke OSA patients with severe WMHs have increased circulating levels of inflammatory chemokine CXCL10 and cytokine Interleukin-10 (IL-10) and significantly decreased levels of Angiopoietin-1 (Ang-1) an important protein responsible for endothelial/vascular integrity functions. Importantly, in a subset of newly diagnosed OSA patients (without prior history of stroke), significantly increased plasma CXCL10 levels and decreased plasma Ang-1 levels were also readily observed when compared to healthy controls, indicating possible altered endothelial integrity and ongoing vascular inflammation in these newly diagnosed OSA patients. CONCLUSIONS In summary, our study has identified a novel set of plasma biomarkers including PDGF-AA, CXCL10 and Ang-1 for their potential prognostic value for disease outcomes pre- and post-stroke in OSA patients and use as surrogate markers to measure efficacy of treatment modalities.
Collapse
Affiliation(s)
- Pritam Das
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, Jacksonville, FL, USA.
| | - Ying Wang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Brynn Dredla
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Karen Jansen-West
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Mohammed Badi
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Owen Ross
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, USA
| | - James F Meschia
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, Jacksonville, FL, USA.
| |
Collapse
|
4
|
Gonzalez-Ortiz F, Kac PR, Brum WS, Zetterberg H, Blennow K, Karikari TK. Plasma phospho-tau in Alzheimer's disease: towards diagnostic and therapeutic trial applications. Mol Neurodegener 2023; 18:18. [PMID: 36927491 PMCID: PMC10022272 DOI: 10.1186/s13024-023-00605-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/15/2023] [Indexed: 03/18/2023] Open
Abstract
As the leading cause of dementia, Alzheimer's disease (AD) is a major burden on affected individuals, their families and caregivers, and healthcare systems. Although AD can be identified and diagnosed by cerebrospinal fluid or neuroimaging biomarkers that concord with neuropathological evidence and clinical symptoms, challenges regarding practicality and accessibility hinder their widespread availability and implementation. Consequently, many people with suspected cognitive impairment due to AD do not receive a biomarker-supported diagnosis. Blood biomarkers have the capacity to help expand access to AD diagnostics worldwide. One such promising biomarker is plasma phosphorylated tau (p-tau), which has demonstrated specificity to AD versus non-AD neurodegenerative diseases, and will be extremely important to inform on clinical diagnosis and eligibility for therapies that have recently been approved. This review provides an update on the diagnostic and prognostic performances of plasma p-tau181, p-tau217 and p-tau231, and their associations with in vivo and autopsy-verified diagnosis and pathological hallmarks. Additionally, we discuss potential applications and unanswered questions of plasma p-tau for therapeutic trials, given their recent addition to the biomarker toolbox for participant screening, recruitment and during-trial monitoring. Outstanding questions include assay standardization, threshold generation and biomarker verification in diverse cohorts reflective of the wider community attending memory clinics and included in clinical trials.
Collapse
Affiliation(s)
- Fernando Gonzalez-Ortiz
- grid.8761.80000 0000 9919 9582Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- grid.1649.a000000009445082XClinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Przemysław R. Kac
- grid.8761.80000 0000 9919 9582Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Wagner S. Brum
- grid.8761.80000 0000 9919 9582Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- grid.8532.c0000 0001 2200 7498Graduate Program in Biological Sciences: Biochemistry, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil
| | - Henrik Zetterberg
- grid.8761.80000 0000 9919 9582Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- grid.1649.a000000009445082XClinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- grid.83440.3b0000000121901201Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- grid.83440.3b0000000121901201UK Dementia Research Institute at UCL, London, UK
- grid.24515.370000 0004 1937 1450Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- grid.8761.80000 0000 9919 9582Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- grid.1649.a000000009445082XClinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Thomas K. Karikari
- grid.8761.80000 0000 9919 9582Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- grid.21925.3d0000 0004 1936 9000Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA USA
| |
Collapse
|
5
|
Yamamoto Y, Liao YC, Lee YC, Ihara M, Choi JC. Update on the Epidemiology, Pathogenesis, and Biomarkers of Cerebral Autosomal Dominant Arteriopathy With Subcortical Infarcts and Leukoencephalopathy. J Clin Neurol 2023; 19:12-27. [PMID: 36606642 PMCID: PMC9833879 DOI: 10.3988/jcn.2023.19.1.12] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/06/2022] [Accepted: 11/09/2022] [Indexed: 01/04/2023] Open
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common monogenic disorder of the cerebral small blood vessels. It is caused by mutations in the NOTCH3 gene on chromosome 19, and more than 280 distinct pathogenic mutations have been reported to date. CADASIL was once considered a very rare disease with an estimated prevalence of 1.3-4.1 per 100,000 adults. However, recent large-scale genomic studies have revealed a high prevalence of pathogenic NOTCH3 variants among the general population, with the highest risk being among Asians. The disease severity and age at onset vary significantly even among individuals who carry the same NOTCH3 mutations. It is still unclear whether a significant genotype-phenotype correlation is present in CADASIL. The accumulation of granular osmiophilic material in the vasculature is a characteristic feature of CADASIL. However, the exact pathogenesis of CADASIL remains largely unclear despite various laboratory and clinical observations being made. Major hypotheses proposed so far have included aberrant NOTCH3 signaling, toxic aggregation, and abnormal matrisomes. Several characteristic features have been observed in the brain magnetic resonance images of patients with CADASIL, including subcortical lacunar lesions and white matter hyperintensities in the anterior temporal lobe or external capsule, which were useful in differentiating CADASIL from sporadic stroke in patients. The number of lacunes and the degree of brain atrophy were useful in predicting the clinical outcomes of patients with CADASIL. Several promising blood biomarkers have also recently been discovered for CADASIL, which require further research for validation.
Collapse
Affiliation(s)
- Yumi Yamamoto
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yi-Chu Liao
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Chung Lee
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Jay Chol Choi
- Department of Neurology, Jeju National University, Jeju, Korea.,Institute for Medical Science, Jeju National University, Jeju, Korea
| |
Collapse
|
6
|
Zhang W, Li M, Zhou X, Huang C, Wan K, Li C, Yin J, Zhao W, Zhang C, Zhu X, Sun Z. Altered serum amyloid beta and cerebral perfusion and their associations with cognitive function in patients with subcortical ischemic vascular disease. Front Neurosci 2022; 16:993767. [PMID: 36312019 PMCID: PMC9608371 DOI: 10.3389/fnins.2022.993767] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/08/2022] [Indexed: 01/07/2024] Open
Abstract
Subcortical ischemic vascular disease (SIVD) is one of the important causes of cognitive dysfunction, altered amyloid-beta (Aβ) and cerebral perfusion may be involved in the pathophysiological mechanism of SIVD and are closely related to cognitive function. We aimed to investigate altered serum Aβ and cerebral perfusion in patients with SIVD and their correlation with cognitive function. Seventy-four healthy controls (HCs) and 74 SIVD patients, including 38 SIVD patients with no cognitive impairment (SIVD-NCI) and 36 SIVD patients with mild cognitive impairment (SIVD-MCI) underwent the measurement of serum Aβ40 and Aβ42 levels, pseudo-continuous arterial spin labeling MRI scanning, and cognitive evaluation. Compared to the healthy controls (HCs), the level of serum Aβ40 and Aβ40/42 ratio increased and Aβ42 decreased in SIVD patients. The serum Aβ40 level and Aβ40/42 ratio in patients with SIVD-MCI were significantly higher than those in the HCs and SIVD-NCI, and the level of Aβ42 in the SIVD-MCI was lower than the HCs. In addition, the serum Aβ40/42 ratio provided high diagnostic accuracy for SIVD and SIVD-MCI, it was further identified as an independent risk factor for cognitive impairment. Patients with SIVD-NCI and SIVD-MCI exhibited both increased and decreased cerebral blood flow (CBF) in regional. The Aβ40/42 ratio was associated with global CBF, while altered global and regional CBF was associated with cognitive deficits. In addition, white matter hyperintensities volume (WMHV) correlated with Aβ40/42 ratio, CBF, and cognition. The relationship between Aβ40/42 ratio and cognition was partially mediated by altered CBF. Based on these results, we conclude that the serum Aβ40/42 ratio may be a potential biomarker that can complement current methods for the prediction and diagnosis of cognitive impairment in SIVD patients. In addition, serum Aβ may play a role in cognitive function by regulating CBF, which provides new insights into the intervention, treatment, and prevention of cognitive impairment in SIVD.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mingxu Li
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xia Zhou
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chaojuan Huang
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ke Wan
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chenchen Li
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiabin Yin
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wenming Zhao
- Department of Radiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cun Zhang
- Department of Radiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaoqun Zhu
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhongwu Sun
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
7
|
Kim H, Jang B, Kim YJ, Choi JC. Determination of Blood NOTCH3 Extracellular Domain and Jagged-1 Levels in Healthy Subjects. Int J Mol Sci 2022; 23:ijms231810547. [PMID: 36142458 PMCID: PMC9505916 DOI: 10.3390/ijms231810547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 11/23/2022] Open
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common genetic disorder among those responsible for hereditary strokes, and it is caused by a mutation in the NOTCH3 gene on chromosome 19. Blood biomarkers related to the Notch signaling pathway have not been investigated extensively in CADASIL. In this study, we measured the serum and plasma levels of NOTCH3 extracellular domain (N3ECD) and its ligand, Jagged-1, in 279 healthy subjects. The levels of N3ECD and Jagged-1 showed significant correlations in both serum (p < 0.0001, r = 0.2681) and plasma (p < 0.0001, r = 0.4065). The N3ECD levels were significantly higher in the serum than in plasma and tend to increase with age. In contrast, there was no significant difference between the serum and plasma levels of Jagged-1 levels. To summarize, we were able to measure N3ECD and Jagged-1 protein levels in healthy human serum and plasma. Taken together, our findings provide the basis for further studies investigating the clinical use of blood N3ECD and Jagged-1 levels for CADASIL and other Notch signaling-related diseases.
Collapse
Affiliation(s)
- Hyesung Kim
- Department of Pathology, School of Medicine, Jeju National University, Jeju 63241, Korea
| | - Bogun Jang
- Department of Pathology, School of Medicine, Jeju National University, Jeju 63241, Korea
- Department of Pathology, Jeju National University Hospital, Jeju 62341, Korea
| | - Yang-Ji Kim
- Institute for Medical Science, Jeju National University, Jeju 63241, Korea
| | - Jay Chol Choi
- Institute for Medical Science, Jeju National University, Jeju 63241, Korea
- Department of Neulogy, Jeju National University Hospital, Jeju 62341, Korea
- Department of Neurology, School of Medicine, Jeju National University, Jeju 63241, Korea
- Correspondence:
| |
Collapse
|
8
|
Association of Serum GFAP with Functional and Neurocognitive Outcome in Sporadic Small Vessel Disease. Biomedicines 2022; 10:biomedicines10081869. [PMID: 36009416 PMCID: PMC9405121 DOI: 10.3390/biomedicines10081869] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/27/2022] [Accepted: 07/31/2022] [Indexed: 11/25/2022] Open
Abstract
Cerebrospinal fluid (CSF) and serum biomarkers are critical for clinical decision making in neurological diseases. In cerebral small vessel disease (CSVD), white matter hyperintensities (WMH) are an important neuroimaging biomarker, but more blood-based biomarkers capturing different aspects of CSVD pathology are needed. In 42 sporadic CSVD patients, we prospectively analysed WMH on magnetic resonance imaging (MRI) and the biomarkers neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), chitinase3-like protein 1 (CHI3L1), Tau and Aβ1-42 in CSF and NfL and GFAP in serum. GFAP and CHI3L1 expression was studied in post-mortem brain tissue in additional cases. CSVD cases with higher serum NfL and GFAP levels had a higher modified Rankin Scale (mRS) and NIHSS score and lower CSF Aβ1-42 levels, whereas the CSF NfL and CHI3L1 levels were positively correlated with the WMH load. Moreover, the serum GFAP levels significantly correlated with the neurocognitive functions. Pathological analyses in CSVD revealed a high density of GFAP-immunoreactive fibrillary astrocytic processes in the periventricular white matter and clusters of CHI3L1-immunoreactive astrocytes in the basal ganglia and thalamus. Thus, besides NfL, serum GFAP is a highly promising fluid biomarker of sporadic CSVD, because it does not only correlate with the clinical severity but also correlates with the cognitive function in patients.
Collapse
|
9
|
Kuipers S, Overmars LM, van Es B, de Bresser J, Bron EE, Hoefer IE, Kappelle LJ, Teunissen CE, Biessels GJ, Haitjema S. A cluster of blood-based protein biomarkers reflecting coagulation relates to the burden of cerebral small vessel disease. J Cereb Blood Flow Metab 2022; 42:1282-1293. [PMID: 35086368 PMCID: PMC9207498 DOI: 10.1177/0271678x221077339] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Biological processes underlying cerebral small vessel disease (cSVD) are largely unknown. We hypothesized that identification of clusters of inter-related bood-based biomarkers that are associated with the burden of cSVD provides leads on underlying biological processes. In 494 participants (mean age 67.6 ± 8.7 years; 36% female; 75% cardiovascular diseases; 25% reference participants) we assessed the relation between 92 blood-based biomarkers from the OLINK cardiovascular III panel and cSVD, using cluster-based analyses. We focused particularly on white matter hyperintensities (WMH). Nineteen biomarkers individually correlated with WMH ratio (r range: 0.16-0.27, Bonferroni corrected p-values <0.05), of which sixteen biomarkers formed one biomarker cluster. Pathway analysis showed that this biomarker cluster predominantly reflected coagulation processes. This cluster related also significantly to other cSVD manifestations (lacunar infarcts, microbleeds, and enlarged perivascular spaces), which supports generalizability beyond WMHs. To study possible causal effects of biological processes reflected by the cluster we performed a mediation analysis that showed a mediation effect of the cluster on the relation between age and WMH ratio (proportion mediated 17%), and hypertension and WMH-volume (proportion mediated 21%). In conclusion, we identified a cluster of blood-based biomarkers reflecting coagulation, that is related to manifestations of cSVD, corroborating involvement of coagulation abnormalities in the etiology of cSVD.
Collapse
Affiliation(s)
- Sanne Kuipers
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - L Malin Overmars
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.,Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Bram van Es
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jeroen de Bresser
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Esther E Bron
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Imo E Hoefer
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - L Jaap Kappelle
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, VrijeUniversiteit Amsterdam, Amsterdam, the Netherlands
| | - Geert Jan Biessels
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Saskia Haitjema
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
10
|
Wang Y, Liu G, Song H, Cao C, Ji X, Cao G. Elevated Lipoprotein-Associated Phospholipase A2 Is Associated With Intracranial Atherosclerosis. Front Neurol 2022; 13:858302. [PMID: 35756924 PMCID: PMC9226299 DOI: 10.3389/fneur.2022.858302] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
Background Lipoprotein-associated phospholipase A2 (Lp-PLA2) is an inflammatory factor in the pathogenesis of atherosclerotic plaque and is associated with an increased risk of ischemic stroke. Whether Lp-PLA2 is associated with stenosis subtypes in acute ischemic stroke (AIS) has not been investigated. Methods A total of 126 eligible AIS patients were divided into four groups: (1) no cerebral artery stenosis (NCS); (2) intracranial artery stenosis (ICAS); (3) extracranial artery stenosis (ECAS); and (4) combined intracranial and extracranial artery stenosis (IECS). Associations between serum Lp-PLA2 levels and the stenosis subtypes were assessed. Results The ICAS group had a lower frequency of dyslipidemia as compared to the NCS group and the IECS group (35.3% vs. 70% vs. 71.8%, respectively, p = 0.001) and was more likely to be symptomatic than the ECAS group (76.5% vs. 43.8%, respectively, p = 0.014). Lp-PLA2 levels in the ICAS group were 112.2 ± 66.8 μg/L which are, higher than those in the NCS, ECAS, and IECS groups (81.7 ± 38.5, 106.1 ± 57.8, 89.3 ± 52.2 μg/L, respectively, p = 0.025). In the third and fourth quartiles of Lp-PLA2 levels, stenosis had occurred more frequently in the ICAS group than in the other three groups (third Q: 50.0% vs. 3.1% vs. 28.1% vs. 18.8%, p = 0.002; fourth Q: 48.4% vs. 16.1% vs. 25.8% vs. 9.7%, p = 0.014). Lp-PLA2 levels were higher in patients with more or severe stenosis in the ICAS group. Conclusions Elevated Lp-PLA2 levels were differentially associated with increased risk in AIS patients with ICAS compared to those with ECAS or no stenosis. Lp-PLA2 may be a promising biomarker and potential therapeutic target for ICAS.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Neurology, Xuanwu Hospital, Capital Medicine University, Beijing, China
| | - Gang Liu
- Department of Neurology, Xuanwu Hospital, Capital Medicine University, Beijing, China
| | - Haiqing Song
- Department of Neurology, Xuanwu Hospital, Capital Medicine University, Beijing, China
| | - Catherine Cao
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital Medicine University, Beijing, China
| | - Guodong Cao
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States.,Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, United States
| |
Collapse
|
11
|
Hußler W, Höhn L, Stolz C, Vielhaber S, Garz C, Schmitt FC, Gundelfinger ED, Schreiber S, Seidenbecher CI. Brevican and Neurocan Cleavage Products in the Cerebrospinal Fluid - Differential Occurrence in ALS, Epilepsy and Small Vessel Disease. Front Cell Neurosci 2022; 16:838432. [PMID: 35480959 PMCID: PMC9036369 DOI: 10.3389/fncel.2022.838432] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
The neural extracellular matrix (ECM) composition shapes the neuronal microenvironment and undergoes substantial changes upon development and aging, but also due to cerebral pathologies. In search for potential biomarkers, cerebrospinal fluid (CSF) and serum concentrations of brain ECM molecules have been determined recently to assess ECM changes during neurological conditions including Alzheimer’s disease or vascular dementia. Here, we measured the levels of two signature proteoglycans of brain ECM, neurocan and brevican, in the CSF and serum of 96 neurological patients currently understudied regarding ECM alterations: 16 cases with amyotrophic lateral sclerosis (ALS), 26 epilepsy cases, 23 cerebral small vessel disease (CSVD) patients and 31 controls. Analysis of total brevican and neurocan was performed via sandwich Enzyme-linked immunosorbent assays (ELISAs). Major brevican and neurocan cleavage products were measured in the CSF using semiquantitative immunoblotting. Total brevican and neurocan concentrations in serum and CSF did not differ between groups. The 60 kDa brevican fragment resulting from cleavage by the protease ADAMTS-4 was also found unchanged among groups. The presumably intracellularly generated 150 kDa C-terminal neurocan fragment, however, was significantly increased in ALS as compared to all other groups. This group also shows the highest correlation between cleaved and total neurocan in the CSF. Brevican and neurocan levels strongly correlated with each other across all groups, arguing for a joint but yet unknown transport mechanism from the brain parenchyma into CSF. Conclusively our findings suggest an ALS-specific pattern of brain ECM remodeling and may thus contribute to new diagnostic approaches for this disorder.
Collapse
Affiliation(s)
- Wilhelm Hußler
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany
- Department of Neurology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Lukas Höhn
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany
- Department of Neurology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | | | - Stefan Vielhaber
- Department of Neurology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Cornelia Garz
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany
- Department of Neurology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Friedhelm C. Schmitt
- Department of Neurology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Eckart D. Gundelfinger
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Institute for Pharmacology and Toxicology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Stefanie Schreiber
- Department of Neurology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Constanze I. Seidenbecher
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- *Correspondence: Constanze I. Seidenbecher,
| |
Collapse
|
12
|
Anastassiadis C, Vasilevskaya A, Gumus M, Santos A, Tartaglia MC. Fluid biomarkers of white matter hyperintensities in cerebrovascular disease and neurodegeneration: a systematic review protocol. JBI Evid Synth 2021; 19:2464-2473. [PMID: 33993148 DOI: 10.11124/jbies-20-00210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE The goal of this systematic review is to evaluate the association between fluid biomarkers and white matter hyperintensities (WMH) in cerebrovascular disease and neurodegenerative disorders. While previous research has examined the etiology of WMH in specific diseases, we propose a comprehensive framework encompassing WMH of both vascular and non-vascular origin. INTRODUCTION Although WMH have been mostly described in aging populations with cerebrovascular disease, extensive lesions also occur in non-vascular diseases. Such lesions are traditionally treated as a separate pathological entity from vascular ones, but recent work has challenged the appropriateness of that framework when probing WMH etiology. Comparing biomarkers associated with WMH across various pathologies may improve our understanding of their etiology. INCLUSION CRITERIA The review will focus on cerebrovascular disease and neurodegenerative disorders and exclude infectious, metabolic, drug-induced, or radiation-induced white matter diseases. Original, peer-reviewed research on the relationship of WMH on magnetic resonance imaging with blood/cerebrospinal fluid biomarkers will be considered for inclusion. Postmortem studies will guide the selection of biomarkers of interest and the interpretation of our findings. Genomic markers will be excluded. METHODS The review will be conducted in accordance with PRISMA and JBI guidelines. English articles of interest published between 2000 and 2020 will be identified in MEDLINE and Embase. Two reviewers will perform abstract and full-text screening, standardized data extraction, and quality assessments of the selected studies. The relationship between each biomarker and WMH burden will be meta-analyzed, if possible, with subgroup or meta-regression analyses to assess differences between diseases. SYSTEMATIC REVIEW REGISTRATION NUMBER PROSPERO CRD42020218298.
Collapse
Affiliation(s)
- Chloe Anastassiadis
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Tanz Center for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Anna Vasilevskaya
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Tanz Center for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada.,Memory Clinic, Division of Neurology, University Health Network (UHN), Toronto, ON, Canada
| | - Melisa Gumus
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Tanz Center for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Alexandra Santos
- Tanz Center for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Maria Carmela Tartaglia
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Tanz Center for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada.,Memory Clinic, Division of Neurology, University Health Network (UHN), Toronto, ON, Canada.,Division of Brain, Imaging and Behaviour - Systems Neuroscience, Krembil Research Institute, UHN, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
13
|
Che Mohd Nassir CMN, Hashim S, Wong KK, Abdul Halim S, Idris NS, Jayabalan N, Guo D, Mustapha M. COVID-19 Infection and Circulating Microparticles-Reviewing Evidence as Microthrombogenic Risk Factor for Cerebral Small Vessel Disease. Mol Neurobiol 2021; 58:4188-4215. [PMID: 34176095 PMCID: PMC8235918 DOI: 10.1007/s12035-021-02457-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 06/16/2021] [Indexed: 02/08/2023]
Abstract
Severe acute respiratory syndrome corona virus-2 (SARS-CoV-2) due to novel coronavirus disease 2019 (COVID-19) has affected the global society in numerous unprecedented ways, with considerable morbidity and mortality. Both direct and indirect consequences from COVID-19 infection are recognized to give rise to cardio- and cerebrovascular complications. Despite current limited knowledge on COVID-19 pathogenesis, inflammation, endothelial dysfunction, and coagulopathy appear to play critical roles in COVID-19-associated cerebrovascular disease (CVD). One of the major subtypes of CVD is cerebral small vessel disease (CSVD) which represents a spectrum of pathological processes of various etiologies affecting the brain microcirculation that can trigger subsequent neuroinflammation and neurodegeneration. Prevalent with aging, CSVD is a recognized risk factor for stroke, vascular dementia, and Alzheimer's disease. In the background of COVID-19 infection, the heightened cellular activations from inflammations and oxidative stress may result in elevated levels of microthrombogenic extracellular-derived circulating microparticles (MPs). Consequently, MPs could act as pro-coagulant risk factor that may serve as microthrombi for the vulnerable microcirculation in the brain leading to CSVD manifestations. This review aims to appraise the accumulating body of evidence on the plausible impact of COVID-19 infection on the formation of microthrombogenic MPs that could lead to microthrombosis in CSVD manifestations, including occult CSVD which may last well beyond the pandemic era.
Collapse
Affiliation(s)
- Che Mohd Nasril Che Mohd Nassir
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Sabarisah Hashim
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
- Hospital Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Kah Keng Wong
- Hospital Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Sanihah Abdul Halim
- Hospital Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
- Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Nur Suhaila Idris
- Hospital Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
- Department of Family Medicine, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Nanthini Jayabalan
- Translational Neuroscience Lab, UQ Centre for Clinical Research, the University of Queensland, Herston, Brisbane, 4029, Australia
| | - Dazhi Guo
- Department of Hyperbaric Oxygen, The Sixth Medical Center of PLA General Hospital, 6 Fucheng Rd, Beijing, 100048, China
| | - Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia.
- Hospital Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
14
|
Jiménez-Balado J, Pizarro J, Riba-Llena I, Penalba A, Faura J, Palà E, Montaner J, Hernández-Guillamon M, Delgado P. New candidate blood biomarkers potentially associated with white matter hyperintensities progression. Sci Rep 2021; 11:14324. [PMID: 34253757 PMCID: PMC8275657 DOI: 10.1038/s41598-021-93498-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/18/2021] [Indexed: 11/09/2022] Open
Abstract
We aimed to discover blood biomarkers associated with longitudinal changes in white matter hyperintensities (WMH). This study was divided into a discovery phase and a replication phase. Subjects in both studies were patients with hypertension, aged 50-70, who underwent two magnetic resonance imaging (MRI) sessions and blood extractions over a 4-year follow-up period. In the discovery phase, we screened 1305 proteins in 12 subjects with WMH progression and in 12 matched control subjects. We found that 41 proteins were differentially expressed: 13 were upregulated and 28 were downregulated. We subsequently selected three biomarkers for replication in baseline and follow-up samples in 80 subjects with WMH progression and in 80 control subjects. The selected protein candidates for the replication were MMP9 (matrix metalloproteinase-9), which was higher in cases, MET (hepatocyte growth factor receptor) and ASAH2 (neutral ceramidase), which were both lower in cases of WMH progression. Baseline biomarker concentrations did not predict WMH progression. In contrast, patients with WMH progression presented a steeper decline in MET over time. Furthermore, cases showed higher MMP9 and lower ASAH2 levels than controls at the follow-up. These results indicate that MMP9, MET, and ASAH2 are potentially associated with the progression of WMH, and could therefore be interesting candidates to validate in future studies.
Collapse
Affiliation(s)
- Joan Jiménez-Balado
- Neurovascular Research Lab. Vall D'Hebron Research Institute, Universitat Autònoma de Barcelona, Edifici Mediterrània, Planta 1ª, Laboratori 123, Passeig Vall d'Hebron 119-129, 08035, Barcelona, CP, Spain
| | - Jesús Pizarro
- Neurovascular Research Lab. Vall D'Hebron Research Institute, Universitat Autònoma de Barcelona, Edifici Mediterrània, Planta 1ª, Laboratori 123, Passeig Vall d'Hebron 119-129, 08035, Barcelona, CP, Spain
| | - Iolanda Riba-Llena
- Neurovascular Research Lab. Vall D'Hebron Research Institute, Universitat Autònoma de Barcelona, Edifici Mediterrània, Planta 1ª, Laboratori 123, Passeig Vall d'Hebron 119-129, 08035, Barcelona, CP, Spain
| | - Anna Penalba
- Neurovascular Research Lab. Vall D'Hebron Research Institute, Universitat Autònoma de Barcelona, Edifici Mediterrània, Planta 1ª, Laboratori 123, Passeig Vall d'Hebron 119-129, 08035, Barcelona, CP, Spain
| | - Júlia Faura
- Neurovascular Research Lab. Vall D'Hebron Research Institute, Universitat Autònoma de Barcelona, Edifici Mediterrània, Planta 1ª, Laboratori 123, Passeig Vall d'Hebron 119-129, 08035, Barcelona, CP, Spain
| | - Elena Palà
- Neurovascular Research Lab. Vall D'Hebron Research Institute, Universitat Autònoma de Barcelona, Edifici Mediterrània, Planta 1ª, Laboratori 123, Passeig Vall d'Hebron 119-129, 08035, Barcelona, CP, Spain
| | - Joan Montaner
- Neurovascular Research Lab. Vall D'Hebron Research Institute, Universitat Autònoma de Barcelona, Edifici Mediterrània, Planta 1ª, Laboratori 123, Passeig Vall d'Hebron 119-129, 08035, Barcelona, CP, Spain.,Institute de Biomedicine of Seville, IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville & Department of Neurology, Hospital Universitario Virgen Macarena, Seville, Spain
| | - Mar Hernández-Guillamon
- Neurovascular Research Lab. Vall D'Hebron Research Institute, Universitat Autònoma de Barcelona, Edifici Mediterrània, Planta 1ª, Laboratori 123, Passeig Vall d'Hebron 119-129, 08035, Barcelona, CP, Spain
| | - Pilar Delgado
- Neurovascular Research Lab. Vall D'Hebron Research Institute, Universitat Autònoma de Barcelona, Edifici Mediterrània, Planta 1ª, Laboratori 123, Passeig Vall d'Hebron 119-129, 08035, Barcelona, CP, Spain. .,Vall D'Hebron University Hospital, Universitat Autònoma de Barcelona, Dementia Unit, Neurology Service, Barcelona, Spain.
| |
Collapse
|
15
|
Ariton DM, Jiménez-Balado J, Maisterra O, Pujadas F, Soler MJ, Delgado P. Diabetes, Albuminuria and the Kidney-Brain Axis. J Clin Med 2021; 10:2364. [PMID: 34072230 PMCID: PMC8198842 DOI: 10.3390/jcm10112364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/19/2021] [Accepted: 05/24/2021] [Indexed: 01/02/2023] Open
Abstract
Cognitive decline and kidney disease are significant public health problems that share similar characteristics and risk factors. The pathophysiology of the kidney-brain axis is not completely understood, and studies analysing the relationship between the biomarkers of kidney damage and cognitive impairment show different results. This article focuses on the epidemiological and clinical aspects concerning the association of albuminuria, a marker for endothelial dysfunction and microvascular disease, and cognitive impairment in patients with chronic kidney disease, diabetic kidney disease and end-stage kidney disease. Most studies show a positive relationship between albuminuria and cognitive impairment in all groups, but evidence in type 2 diabetes (T2D) patients is limited. We briefly discuss the mechanisms underlying these associations, such as damage to the microvascular circulation, leading to hypoperfusion and blood pressure fluctuations, as well as increased inflammation and oxidative stress, both in the brain and in the kidneys. Further clinical and epidemiological studies developed to understand the interplay between the kidneys and brain diseases will hopefully lead to a reduction in cognitive impairment in these patients.
Collapse
Affiliation(s)
- Diana Maria Ariton
- Neurology Department, Hospital Vall d’Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (D.M.A.); (J.J.-B.); (O.M.); (F.P.)
| | - Joan Jiménez-Balado
- Neurology Department, Hospital Vall d’Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (D.M.A.); (J.J.-B.); (O.M.); (F.P.)
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Olga Maisterra
- Neurology Department, Hospital Vall d’Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (D.M.A.); (J.J.-B.); (O.M.); (F.P.)
| | - Francesc Pujadas
- Neurology Department, Hospital Vall d’Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (D.M.A.); (J.J.-B.); (O.M.); (F.P.)
| | - María José Soler
- Nephrology Department, Hospital Vall d’Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
| | - Pilar Delgado
- Neurology Department, Hospital Vall d’Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (D.M.A.); (J.J.-B.); (O.M.); (F.P.)
| |
Collapse
|
16
|
Wilcock D, Jicha G, Blacker D, Albert MS, D’Orazio LM, Elahi FM, Fornage M, Hinman JD, Knoefel J, Kramer J, Kryscio RJ, Lamar M, Moghekar A, Prestopnik J, Ringman JM, Rosenberg G, Sagare A, Satizabal CL, Schneider J, Seshadri S, Sur S, Tracy RP, Yasar S, Williams V, Singh H, Mazina L, Helmer KG, Corriveau RA, Schwab K, Kivisäkk P, Greenberg SM. MarkVCID cerebral small vessel consortium: I. Enrollment, clinical, fluid protocols. Alzheimers Dement 2021; 17:704-715. [PMID: 33480172 PMCID: PMC8122220 DOI: 10.1002/alz.12215] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/22/2020] [Indexed: 01/04/2023]
Abstract
The concept of vascular contributions to cognitive impairment and dementia (VCID) derives from more than two decades of research indicating that (1) most older individuals with cognitive impairment have post mortem evidence of multiple contributing pathologies and (2) along with the preeminent role of Alzheimer's disease (AD) pathology, cerebrovascular disease accounts for a substantial proportion of this contribution. Contributing cerebrovascular processes include both overt strokes caused by etiologies such as large vessel occlusion, cardioembolism, and embolic infarcts of unknown source, and frequently asymptomatic brain injuries caused by diseases of the small cerebral vessels. Cerebral small vessel diseases such as arteriolosclerosis and cerebral amyloid angiopathy, when present at moderate or greater pathologic severity, are independently associated with worse cognitive performance and greater likelihood of dementia, particularly in combination with AD and other neurodegenerative pathologies. Based on this evidence, the US National Alzheimer's Project Act explicitly authorized accelerated research in vascular and mixed dementia along with frontotemporal and Lewy body dementia and AD itself. Biomarker development has been consistently identified as a key step toward translating scientific advances in VCID into effective prevention and treatment strategies. Validated biomarkers can serve a range of purposes in trials of candidate interventions, including (1) identifying individuals at increased VCID risk, (2) diagnosing the presence of cerebral small vessel disease or specific small vessel pathologies, (3) stratifying study participants according to their prognosis for VCID progression or treatment response, (4) demonstrating an intervention's target engagement or pharmacodynamic mechanism of action, and (5) monitoring disease progression during treatment. Effective biomarkers allow academic and industry investigators to advance promising interventions at early stages of development and discard interventions with low success likelihood. The MarkVCID consortium was formed in 2016 with the goal of developing and validating fluid- and imaging-based biomarkers for the cerebral small vessel diseases associated with VCID. MarkVCID consists of seven project sites and a central coordinating center, working with the National Institute of Neurologic Diseases and Stroke and National Institute on Aging under cooperative agreements. Through an internal selection process, MarkVCID has identified a panel of 11 candidate biomarker "kits" (consisting of the biomarker measure and the clinical and cognitive data used to validate it) and established a range of harmonized procedures and protocols for participant enrollment, clinical and cognitive evaluation, collection and handling of fluid samples, acquisition of neuroimaging studies, and biomarker validation. The overarching goal of these protocols is to generate rigorous validating data that could be used by investigators throughout the research community in selecting and applying biomarkers to multi-site VCID trials. Key features of MarkVCID participant enrollment, clinical/cognitive testing, and fluid biomarker procedures are summarized here, with full details in the following text, tables, and supplemental material, and a description of the MarkVCID imaging biomarker procedures in a companion paper, "MarkVCID Cerebral small vessel consortium: II. Neuroimaging protocols." The procedures described here address a range of challenges in MarkVCID's design, notably: (1) acquiring all data under informed consent and enrollment procedures that allow unlimited sharing and open-ended analyses without compromising participant privacy rights; (2) acquiring the data in a sufficiently wide range of study participants to allow assessment of candidate biomarkers across the various patient groups who might ultimately be targeted in VCID clinical trials; (3) defining a common dataset of clinical and cognitive elements that contains all the key outcome markers and covariates for VCID studies and is realistically obtainable during a practical study visit; (4) instituting best fluid-handling practices for minimizing avoidable sources of variability; and (5) establishing rigorous procedures for testing the reliability of candidate fluid-based biomarkers across replicates, assay runs, sites, and time intervals (collectively defined as the biomarker's instrumental validity). Participant Enrollment Project sites enroll diverse study cohorts using site-specific inclusion and exclusion criteria so as to provide generalizable validation data across a range of cognitive statuses, risk factor profiles, small vessel disease severities, and racial/ethnic characteristics representative of the diverse patient groups that might be enrolled in a future VCID trial. MarkVCID project sites include both prospectively enrolling centers and centers providing extant data and samples from preexisting community- and population-based studies. With approval of local institutional review boards, all sites incorporate MarkVCID consensus language into their study documents and informed consent agreements. The consensus language asks prospectively enrolled participants to consent to unrestricted access to their data and samples for research analysis within and outside MarkVCID. The data are transferred and stored as a de-identified dataset as defined by the Health Insurance Portability and Accountability Act Privacy Rule. Similar human subject protection and informed consent language serve as the basis for MarkVCID Research Agreements that act as contracts and data/biospecimen sharing agreements across the consortium. Clinical and Cognitive Data Clinical and cognitive data are collected across prospectively enrolling project sites using common MarkVCID instruments. The clinical data elements are modified from study protocols already in use such as the Alzheimer's Disease Center program Uniform Data Set Version 3 (UDS3), with additional focus on VCID-related items such as prior stroke and cardiovascular disease, vascular risk factors, focal neurologic findings, and blood testing for vascular risk markers and kidney function including hemoglobin A1c, cholesterol subtypes, triglycerides, and creatinine. Cognitive assessments and rating instruments include the Clinical Dementia Rating Scale, Geriatric Depression Scale, and most of the UDS3 neuropsychological battery. The cognitive testing requires ≈60 to 90 minutes. Study staff at the prospectively recruiting sites undergo formalized training in all measures and review of their first three UDS3 administrations by the coordinating center. Collection and Handling of Fluid Samples Fluid sample types collected for MarkVCID biomarker kits are serum, ethylenediaminetetraacetic acid-plasma, platelet-poor plasma, and cerebrospinal fluid (CSF) with additional collection of packed cells to allow future DNA extraction and analyses. MarkVCID fluid guidelines to minimize variability include fasting morning fluid collections, rapid processing, standardized handling and storage, and avoidance of CSF contact with polystyrene. Instrumental Validation for Fluid-Based Biomarkers Instrumental validation of MarkVCID fluid-based biomarkers is operationally defined as determination of intra-plate and inter-plate repeatability, inter-site reproducibility, and test-retest repeatability. MarkVCID study participants both with and without advanced small vessel disease are selected for these determinations to assess instrumental validity across the full biomarker assay range. Intra- and inter-plate repeatability is determined by repeat assays of single split fluid samples performed at individual sites. Inter-site reproducibility is determined by assays of split samples distributed to multiple sites. Test-retest repeatability is determined by assay of three samples acquired from the same individual, collected at least 5 days apart over a 30-day period and assayed on a single plate. The MarkVCID protocols are designed to allow direct translation of the biomarker validation results to multicenter trials. They also provide a template for outside groups to perform analyses using identical methods and therefore allow direct comparison of results across studies and centers. All MarkVCID protocols are available to the biomedical community and intended to be shared. In addition to the instrumental validation procedures described here, each of the MarkVCID kits will undergo biological validation to determine whether the candidate biomarker measures important aspects of VCID such as cognitive function. Analytic methods and results of these validation studies for the 11 MarkVCID biomarker kits will be published separately. The results of this rigorous validation process will ultimately determine each kit's potential usefulness for multicenter interventional trials aimed at preventing or treating small vessel disease related VCID.
Collapse
Affiliation(s)
- Donna Wilcock
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY 40504, USA
| | - Gregory Jicha
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY 40504, USA
| | - Deborah Blacker
- Department of Epidemiology, Harvard T.H Chan School of Public Health and Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Marilyn S. Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Lina M. D’Orazio
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Fanny M. Elahi
- Center for Memory and Aging, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School and Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jason D. Hinman
- David Geffen School of Medicine, Department of Neurology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Janice Knoefel
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Joel Kramer
- David Geffen School of Medicine, Department of Neurology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Richard J. Kryscio
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY 40504, USA
| | - Melissa Lamar
- Rush Alzheimer’s Disease Center, Rush University, Chicago, IL, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jillian Prestopnik
- Center for Memory and Aging, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - John M. Ringman
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Gary Rosenberg
- Center for Memory and Aging, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Abhay Sagare
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Claudia L. Satizabal
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Julie Schneider
- Rush Alzheimer’s Disease Center, Rush University, Chicago, IL, USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Sandeepa Sur
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine, University of Vermont Larner College of Medicine, Burlington, VT 05405, USA
| | - Sevil Yasar
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Victoria Williams
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Herpreet Singh
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Lidiya Mazina
- Neurological Clinical Research Institute, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Karl G. Helmer
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Kristin Schwab
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Pia Kivisäkk
- Alzheimer’s Clinical and Translational Research Unit, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Steven M. Greenberg
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | | |
Collapse
|
17
|
Qu Y, Tan CC, Shen XN, Li HQ, Cui M, Tan L, Dong Q, Yu JT. Association of Plasma Neurofilament Light With Small Vessel Disease Burden in Nondemented Elderly: A Longitudinal Study. Stroke 2021; 52:896-904. [PMID: 33517704 DOI: 10.1161/strokeaha.120.030302] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Neurofilament light chain (NfL) is a promising predictive biomarker of active axonal injury and neuronal degeneration diseases. We aimed to evaluate if an increase in plasma NfL levels could play a monitoring role in the progression of cerebral small vessel disease (CSVD) among the nondemented elders, which are highly prevalent in elderly individuals and associated with an increased risk of stroke and dementia. METHODS The study included 496 nondemented participants from the Alzheimer disease neuroimaging initiative database. All participants underwent plasma NfL measurements and 3.0-Tesla magnetic resonance imaging of the brain; 387 (78.0%) underwent longitudinal measurements. The number of cerebral microbleeds, lacunar infarcts, and volumetric white matter hyperintensities, as well as Fazekas scores, were measured. Cross-sectional and longitudinal associations between CSVD burden and NfL levels were evaluated using multivariable-adjusted models. RESULTS Plasma NfL was higher in the moderate-severe CSVD burden group (45.2±16.0 pg/mL) than in the nonburden group (34.3±15.1 pg/mL; odds ratio [OR]=1.71 [95% CI, 1.24-2.35]) at baseline. NfL was positively associated with the presence of cerebral microbleeds (OR=1.29 [95% CI, 1.01-1.64]), lacunar infarcts (OR=1.43 [95% CI, 1.06-1.93]), and moderate-severe white matter hyperintensities (OR=1.67 [95% CI, 1.24-2.25]). Longitudinally, a higher change rate of NfL could predict more progression of CSVD burden (OR=1.38 [95% CI, 1.08-1.76]), white matter hyperintensities (OR=1.41 [95% CI, 1.10-1.79]), and lacunar infarcts (OR=1.99 [95% CI, 1.42-2.77]). CONCLUSIONS Plasma NfL level is a valuable noninvasive biomarker that supplements magnetic resonance imaging scans and possibly reflects the severity of CSVD burden. Furthermore, high plasma NfL levels tend to represent an increased CSVD risk, and dynamic increases in NfL levels might predict a greater progression of CSVD.
Collapse
Affiliation(s)
- Yi Qu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, China (Y.Q., C.-C.T., L.T.)
| | - Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, China (Y.Q., C.-C.T., L.T.)
| | - Xue-Ning Shen
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, China (X.-N.S., H.-Q.L., M.C., Q.D., J.-T.Y.)
| | - Hong-Qi Li
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, China (X.-N.S., H.-Q.L., M.C., Q.D., J.-T.Y.)
| | - Mei Cui
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, China (X.-N.S., H.-Q.L., M.C., Q.D., J.-T.Y.)
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, China (Y.Q., C.-C.T., L.T.)
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, China (X.-N.S., H.-Q.L., M.C., Q.D., J.-T.Y.)
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, China (X.-N.S., H.-Q.L., M.C., Q.D., J.-T.Y.)
| | | |
Collapse
|
18
|
Zhang J, Sun P, Zhou C, Zhang X, Ma F, Xu Y, Hamblin MH, Yin K. Regulatory microRNAs and vascular cognitive impairment and dementia. CNS Neurosci Ther 2020; 26:1207-1218. [PMID: 33459504 PMCID: PMC7702235 DOI: 10.1111/cns.13472] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular cognitive impairment and dementia (VCID) is defined as a progressive dementia disease related to cerebrovascular injury and often occurs in aged populations. Despite decades of research, effective treatment for VCID is still absent. The pathological processes of VCID are mediated by the molecular mechanisms that are partly modulated at the post-transcriptional level. As small endogenous non-coding RNAs, microRNAs (miRs) can regulate target gene expression through post-transcriptional gene silencing. miRs have been reported to play an important role in the pathology of VCID and have recently been suggested as potential novel pharmacological targets for the development of new diagnosis and treatment strategies in VCID. In this review, we summarize the current understanding of VCID, the possible role of miRs in the regulation of VCID and attempt to envision future therapeutic strategies. Since manipulation of miR levels by either pharmacological or genetic approaches has shown therapeutic effects in experimental VCID models, we also emphasize the potential therapeutic value of miRs in clinical settings.
Collapse
Affiliation(s)
- Jing Zhang
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Ping Sun
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Chao Zhou
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Xuejing Zhang
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Feifei Ma
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Yang Xu
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Milton H. Hamblin
- Department of PharmacologyTulane University School of MedicineNew OrleansLAUSA
| | - Ke‐Jie Yin
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
- Geriatric ResearchEducation and Clinical CenterVeterans Affairs Pittsburgh Healthcare SystemPittsburghPAUSA
| |
Collapse
|
19
|
Jiménez-Balado J, Riba-Llena I, Maisterra O, Pizarro J, Palasí A, Pujadas F, Mundet X, Vinyoles E, Delgado P. Ambulatory Blood Pressure Levels in the Prediction of Progression of Cerebral Small Vessel Disease. J Am Geriatr Soc 2020; 68:2232-2239. [PMID: 32511756 DOI: 10.1111/jgs.16568] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/30/2020] [Accepted: 05/03/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVES We aimed to study the value of ambulatory blood pressure monitoring (ABPM) in predicting the global progression of cerebral small vessel disease (cSVD). DESIGN Longitudinal cohort study. SETTING Data from the population-based Investigating Silent Strokes in Hypertensives study. PARTICIPANTS Individuals with hypertension who were 50 to 70 years of age and stroke free at baseline. In baseline and follow-up visits, patients underwent magnetic resonance imaging and ABPM. MEASUREMENTS Ambulatory systolic blood pressure (SBP) and diastolic blood pressure (DBP) levels were studied as continuous variables and dichotomized according to good or poor control on the basis of 125/75 (24 hours), 130/80 (day), and 110/65 (night) mm Hg cutoff values. Whole cSVD progression was qualitatively scored with 1 point when an incident lesion (incident lacunar infarcts, deep cerebral microbleeds, white matter hyperintensities, and basal ganglia enlarged perivascular spaces) was detected. The score ranged from 0 to 4. RESULTS We followed up 233 participants with a median age of 65 years within 4 years. A total of 61 (26.2%) and 23 (9.9%) subjects showed cSVD progression in one and two or more markers, respectively. Baseline ambulatory SBP and DBP and nighttime pulse pressure (PP) values were positively correlated with the number of incident cSVD lesions. Interestingly, patients without incident lesions showed greater differences between office and ambulatory BP, thus suggesting an increased white coat effect. Poor DBP control, nighttime PP, and DBP white coat effect were independently associated with cSVD progression. The inclusion of these metrics in a clinical model resulted in a significant increase in the prediction of incident lesions (integrated discrimination improvement = 9.09%; P value <.001). CONCLUSION ABPM may help assess cSVD risk of progression, especially by the identification of poor BP control, masked hypertension, and increased nighttime PP. J Am Geriatr Soc 68:2232-2239, 2020.
Collapse
Affiliation(s)
- Joan Jiménez-Balado
- Neurovascular Research Lab, Vall Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Iolanda Riba-Llena
- Neurovascular Research Lab, Vall Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Olga Maisterra
- Neurovascular Research Lab, Vall Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain.,Dementia Unit, Neurology Service, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jesús Pizarro
- Neurovascular Research Lab, Vall Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Antoni Palasí
- Dementia Unit, Neurology Service, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francesc Pujadas
- Dementia Unit, Neurology Service, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Xavier Mundet
- Primary Healthcare University Research Institute IDIAP Jordi Gol, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ernest Vinyoles
- Primary Healthcare University Research Institute IDIAP Jordi Gol, CAP La Mina, Universitat de Barcelona, Barcelona, Spain
| | - Pilar Delgado
- Neurovascular Research Lab, Vall Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain.,Dementia Unit, Neurology Service, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
20
|
Neurovascular unit dysregulation, white matter disease, and executive dysfunction: the shared triad of vascular cognitive impairment and Alzheimer disease. GeroScience 2020; 42:445-465. [PMID: 32002785 DOI: 10.1007/s11357-020-00164-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 01/22/2020] [Indexed: 01/07/2023] Open
Abstract
Executive dysfunction is the most important predictor for loss of independence in dementia. As executive function involves the coordination of distributed cerebral functions, executive function requires healthy white matter. However, white matter is highly vulnerable to cerebrovascular insults, with executive dysfunction being a core feature of vascular cognitive impairment (VCI). At the same time, cerebrovascular pathology, white matter disease, and executive dysfunction are all increasingly recognized as features of Alzheimer disease (AD). Recent studies have characterized the crucial role of glial cells in the pathological changes observed in both VCI and AD. In comorbid VCI and AD, the glial cells of the neurovascular unit (NVU) emerge as important therapeutic targets for the preservation of white matter integrity and executive function. Our synthesis from current research identifies dysregulation of the NVU, white matter disease, and executive dysfunction as a fundamental triad that is common to both VCI and AD. Further study of this triad will be critical for advancing the prevention and management of dementia.
Collapse
|
21
|
Mustapha M, Nassir CMNCM, Aminuddin N, Safri AA, Ghazali MM. Cerebral Small Vessel Disease (CSVD) - Lessons From the Animal Models. Front Physiol 2019; 10:1317. [PMID: 31708793 PMCID: PMC6822570 DOI: 10.3389/fphys.2019.01317] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 09/30/2019] [Indexed: 12/28/2022] Open
Abstract
Cerebral small vessel disease (CSVD) refers to a spectrum of clinical and imaging findings resulting from pathological processes of various etiologies affecting cerebral arterioles, perforating arteries, capillaries, and venules. Unlike large vessels, it is a challenge to visualize small vessels in vivo, hence the difficulty to directly monitor the natural progression of the disease. CSVD might progress for many years during the early stage of the disease as it remains asymptomatic. Prevalent among elderly individuals, CSVD has been alarmingly reported as an important precursor of full-blown stroke and vascular dementia. Growing evidence has also shown a significant association between CSVD's radiological manifestation with dementia and Alzheimer's disease (AD) pathology. Although it remains contentious as to whether CSVD is a cause or sequelae of AD, it is not far-fetched to posit that effective therapeutic measures of CSVD would mitigate the overall burden of dementia. Nevertheless, the unifying theory on the pathomechanism of the disease remains elusive, hence the lack of effective therapeutic approaches. Thus, this chapter consolidates the contemporary insights from numerous experimental animal models of CSVD, to date: from the available experimental animal models of CSVD and its translational research value; the pathomechanical aspects of the disease; relevant aspects on systems biology; opportunities for early disease biomarkers; and finally, converging approaches for future therapeutic directions of CSVD.
Collapse
Affiliation(s)
- Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | | | - Niferiti Aminuddin
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
- Department of Basic Medical Sciences, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan, Malaysia
| | - Amanina Ahmad Safri
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Mazira Mohamad Ghazali
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
22
|
Liu Y, Chen H, Zhao K, He W, Lin S, He J. High levels of plasma fibrinogen are related to post-stroke cognitive impairment. Brain Behav 2019; 9:e01391. [PMID: 31475471 PMCID: PMC6790326 DOI: 10.1002/brb3.1391] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 06/30/2019] [Accepted: 07/28/2019] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION Studies have shown that high levels of the fibrinogen (FIB) are related to cognitive deficits. However, the relationship between fibrinogen and cognitive deficit after stroke remains unclear. Therefore, we explored the relationship between plasma fibrinogen and post-stroke cognitive impairment (PSCI). METHODS This study is carried out in the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China. A total of 210 patients with acute ischemic stroke were enrolled in this study. Ultimately, 134 patients completed 3-month follow-up. Blood samples were collected at hospital admission. Cognitive function was evaluated 3 months after stroke. All patients underwent the Mini-Mental State Examination (MMSE) after 3 months. RESULTS Higher levels of fibrinogen were observed in patients with post-stroke cognitive impairment compared with the non-PSCI group (p < .001). Additionally, elevated plasma fibrinogen levels were independently associated with PSCI (odds ratio [OR] = 2.000, 95% CI 1.062-3.770 p = .032). The plasma fibrinogen levels were negatively correlated with the 3-month MMSE scores (r = -.171, p = .048). In a multivariate linear regression, FIB was negatively associated with the 3-month MMSE scores after adjustment for the other variables (β = -0.782, p = .035). CONCLUSION High levels of plasma fibrinogen were associated with the presence and severity of PSCI.
Collapse
Affiliation(s)
- Yuntao Liu
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huijun Chen
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kai Zhao
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weilei He
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shasha Lin
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jincai He
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
23
|
Azizkhanian I, Sheth SA, Iavarone AT, Lee S, Kakarla V, Hinman JD. Plasma Lipid Profiling Identifies Biomarkers of Cerebral Microvascular Disease. Front Neurol 2019; 10:950. [PMID: 31555203 PMCID: PMC6727208 DOI: 10.3389/fneur.2019.00950] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 08/16/2019] [Indexed: 12/14/2022] Open
Abstract
Brain-specific sphingolipids (SLs) may serve as effective biomarkers of white matter hyperintensities (WMH). Here, we investigate the efficacy of SLs as a novel fluid-based biomarker to identify WMH reflective of chronic ischemia. Patients presenting to our stroke center for evaluation of acute neurological deficits were enrolled in the Advanced Serum Profiling in Recent Stroke (ASPIRE) study. From this cohort of 202 individuals, 58 patients who underwent an MRI and did not have a clinical stroke event were included in this study. Plasma samples were collected at the time of MRI, and targeted SL profiling was performed by HPLC/tandem mass spectrometry. T2 FLAIR imaging was evaluated for WMH and scored according to the Fazekas scoring (FS) method and manually quantified. Twenty two SLs were definitively identified, consisting of ceramide (Cer) and sphingomyelin (SM) species. Of these, two sphingolipids, SM 38:1 and Cer 34:1, significantly correlated with high FS (r = 0.287, p = 0.029, and r = 0.356, p = 0.006, respectively) and were used in subsequent analysis. SM 38:1 (OR 1.129, 95% CI 1.032, 1.236, p = 0.008) and Cer 34:1 (OR 1.118, 95% CI 1.031, 1.212, p = 0.007), accurately differentiated between FS 0–2 vs. 2.5–6 in regression analysis. A combined lipid score demonstrated fair discrimination in ROC analysis (AUC = 0.729, p = 0.003) and was cross-validated using leave-one-out analysis. Plasma levels of brain-specific SLs may serve as effective biomarkers of subacute white matter disease.
Collapse
Affiliation(s)
- Ida Azizkhanian
- New York Medical College School of Medicine, Valhalla, NY, United States
| | - Sunil A Sheth
- Department of Neurology, UT Health McGovern School of Medicine, Houston, TX, United States
| | - Anthony T Iavarone
- QB3/Chemistry Mass Spectrometry Facility, University of California, Berkeley, Berkeley, CA, United States
| | - Songmi Lee
- Department of Neurology, UT Health McGovern School of Medicine, Houston, TX, United States
| | - Visesha Kakarla
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jason D Hinman
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
24
|
Remote ischemic conditioning improves cognition in patients with subcortical ischemic vascular dementia. BMC Neurol 2019; 19:206. [PMID: 31443692 PMCID: PMC6706912 DOI: 10.1186/s12883-019-1435-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/18/2019] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Subcortical ischemic vascular dementia (SIVD) is very common among the older people, but has no approved treatment. Preclinical trials show that remote ischemic conditioning (RIC) reduces recurrence of ischemic stroke. We hypothesize that RIC may also be an effective therapy for patients with SIVD. METHODS Thirty-seven consecutive SIVD cases were enrolled in this randomized control study. Eighteen RIC patients underwent five brief cycles of conditioning (bilateral upper limb compression at 200 mmHg) followed by reperfusion twice daily over 6 consecutive months. Nineteen control patients underwent the same process, but at a pressure of 60 mmHg which caused no restriction on the blood flow of the upper limb. The primary outcome measures were changes in neuropsychological assessments. The secondary outcomes included the changes in high-sensitive C-reactive protein (hs-CRP) concentration, white matter lesion volume (WMLV), diffusion tension imaging (DTI) metrics of white matter. All data were collected at baseline and follow-up. RESULTS A significant treatment difference favoring RIC at 6 months was observed on performance of Hopkins Verbal Learning Test-Revised (HVLT-R), Controlled Oral Word Association Test (COWAT), Trail Making Test A and B (TMT-A & TMT-B), and Judgment of Line Orientation (JLO) (p < 0.05). The control group did not show much improvement after the treatment, and only with a slight change in HVLT-R and TMT-R (p < 0.05). Covariance analysis of efficacy between the two groups suggested that RIC patients performed better on JLO than control patients at the 6-month follow-up (RIC 23.10 vs. control 18.56; p = 0.013). Although DTI metrics were comparable, Hs-CRP levels and WMLV in RIC patients showed a declining trend. CONCLUSIONS Over the 6-month treatment period, we found that RIC was safe and effective for improving cognitive function in SIVD patients. TRIAL REGISTRATION Clinical Trial Registration ( http://www.clinicaltrials.gov ), Unique identifier: NCT03022149; Retrospectively registered; Date of registration: January 16, 2017.
Collapse
|
25
|
Bahrani AA, Al-Janabi OM, Abner EL, Bardach SH, Kryscio RJ, Wilcock DM, Smith CD, Jicha GA. Post-acquisition processing confounds in brain volumetric quantification of white matter hyperintensities. J Neurosci Methods 2019; 327:108391. [PMID: 31408649 DOI: 10.1016/j.jneumeth.2019.108391] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 08/03/2019] [Accepted: 08/03/2019] [Indexed: 01/18/2023]
Abstract
BACKGROUND Disparate research sites using identical or near-identical magnetic resonance imaging (MRI) acquisition techniques often produce results that demonstrate significant variability regarding volumetric quantification of white matter hyperintensities (WMH) in the aging population. The sources of such variability have not previously been fully explored. NEW METHOD 3D FLAIR sequences from a group of randomly selected aged subjects were analyzed to identify sources-of-variability in post-acquisition processing that can be problematic when comparing WMH volumetric data across disparate sites. The methods developed focused on standardizing post-acquisition protocol processing methods to develop a protocol with less than 0.5% inter-rater variance. RESULTS A series of experiments using standard MRI acquisition sequences explored post-acquisition sources-of-variability in the quantification of WMH volumetric data. Sources-of-variability included: the choice of image center, software suite and version, thresholding selection, and manual editing procedures (when used). Controlling for the identified sources-of-variability led to a protocol with less than 0.5% variability between independent raters in post-acquisition WMH volumetric quantification. COMPARISON WITH EXISTING METHOD(S) Post-acquisition processing techniques can introduce an average variance approaching 15% in WMH volume quantification despite identical scan acquisitions. Understanding and controlling for such sources-of-variability can reduce post-acquisition quantitative image processing variance to less than 0.5%. DISCUSSION Considerations of potential sources-of-variability in MRI volume quantification techniques and reduction in such variability is imperative to allow for reliable cross-site and cross-study comparisons.
Collapse
Affiliation(s)
- Ahmed A Bahrani
- Department of Biomedical Engineering, College of Engineering, University of Kentucky, Lexington, KY, 40506, United States; Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States; Biomedical Engineering Department, Al-Khwarizmi College of Engineering, University of Baghdad, Baghdad, Iraq
| | - Omar M Al-Janabi
- Department of Behavioral Science, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States; Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
| | - Erin L Abner
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States; Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY, 40506, United States; Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
| | - Shoshana H Bardach
- Department of Gerontology, College of Public Health, University of Kentucky, Lexington, KY, 40506, United States; Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
| | - Richard J Kryscio
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, 40506, United States; Department of Statistics, College of Arts and Science, University of Kentucky, Lexington, KY, 40506, United States; Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
| | - Donna M Wilcock
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States; Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
| | - Charles D Smith
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States; Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States; Magnetic Resonance Imaging and Spectroscopy Center (MRISC), College of Medicine, University of Kentucky, Lexington, KY, 40506, United States
| | - Gregory A Jicha
- Department of Behavioral Science, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States; Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States; Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, 40506, United States.
| |
Collapse
|
26
|
Montaner J, Ramiro L, Simats A, Hernández-Guillamon M, Delgado P, Bustamante A, Rosell A. Matrix metalloproteinases and ADAMs in stroke. Cell Mol Life Sci 2019; 76:3117-3140. [PMID: 31165904 PMCID: PMC11105215 DOI: 10.1007/s00018-019-03175-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 05/22/2019] [Accepted: 05/29/2019] [Indexed: 12/27/2022]
Abstract
Stroke is a leading cause of death and disability worldwide. However, after years of in-depth research, the pathophysiology of stroke is still not fully understood. Increasing evidence shows that matrix metalloproteinases (MMPs) and "a disintegrin and metalloproteinase" (ADAMs) participate in the neuro-inflammatory cascade that is triggered during stroke but also in recovery phases of the disease. This review covers the involvement of these proteins in brain injury following cerebral ischemia which has been widely studied in recent years, with efforts to modulate this group of proteins in neuroprotective therapies, together with their implication in neurorepair mechanisms. Moreover, the review also discusses the role of these proteins in specific forms of neurovascular disease, such as small vessel diseases and intracerebral hemorrhage. Finally, the potential use of MMPs and ADAMs as guiding biomarkers of brain injury and repair for decision-making in cases of stroke is also discussed.
Collapse
Affiliation(s)
- Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain.
| | - Laura Ramiro
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Alba Simats
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Pilar Delgado
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Alejandro Bustamante
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| |
Collapse
|
27
|
Cipollini V, Troili F, Giubilei F. Emerging Biomarkers in Vascular Cognitive Impairment and Dementia: From Pathophysiological Pathways to Clinical Application. Int J Mol Sci 2019; 20:ijms20112812. [PMID: 31181792 PMCID: PMC6600494 DOI: 10.3390/ijms20112812] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 12/13/2022] Open
Abstract
Vascular pathology is the second most common neuropathology of dementia after Alzheimer’s disease (AD), with small vessels disease (SVD) being considered the major cause of vascular cognitive impairment and dementia (VCID). This review aims to evaluate pathophysiological pathways underlying a diagnosis of VCID. Firstly, we will discuss the role of endothelial dysfunction, blood-brain barrier disruption and neuroinflammation in its pathogenesis. Then, we will analyse different biomarkers including the ones of inflammatory responses to central nervous system tissue injuries, of coagulation and thrombosis and of circulating microRNA. Evidences on peripheral biomarkers for VCID are still poor and large-scale, prospectively designed studies are needed to translate these findings into clinical practice, in order to set different combinations of biomarkers to use for differential diagnosis among types of dementia.
Collapse
Affiliation(s)
- Virginia Cipollini
- S. Andrea Hospital, NESMOS Department, Faculty of Medicine and Psychology, Sapienza University of Rome, Via di Grottarossa 1035, 00189 Roma, Italy.
| | - Fernanda Troili
- S. Andrea Hospital, NESMOS Department, Faculty of Medicine and Psychology, Sapienza University of Rome, Via di Grottarossa 1035, 00189 Roma, Italy.
| | - Franco Giubilei
- S. Andrea Hospital, NESMOS Department, Faculty of Medicine and Psychology, Sapienza University of Rome, Via di Grottarossa 1035, 00189 Roma, Italy.
| |
Collapse
|
28
|
Xu M, Wang MM, Gao Y, Keep RF, Shi Y. The effect of age-related risk factors and comorbidities on white matter injury and repair after ischemic stroke. Neurobiol Dis 2018; 126:13-22. [PMID: 30017454 DOI: 10.1016/j.nbd.2018.07.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/17/2018] [Accepted: 07/10/2018] [Indexed: 02/06/2023] Open
Abstract
White matter injury is a crucial component of human stroke, but it has often been neglected in preclinical studies. Most human stroke is associated with one or more comorbidities, including aging, hypertension, diabetes and metabolic syndrome including hyperlipidemia. The purpose of this review is to examine how age and hypertension impact stroke-induced white matter injury as well as white matter repair in both human stroke and preclinical models. It is essential that comorbidities be examined in preclinical trials as they may impact translatability to the clinic. In addition, understanding how comorbidities impact white matter injury and repair may provide new therapeutic opportunities for patients with those conditions.
Collapse
Affiliation(s)
- Mingyue Xu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Michael M Wang
- Departments of Neurology and Physiology, University of Michigan, Ann Arbor, MI 48109, USA; VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Yejie Shi
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
29
|
Ma J, Yan H, Wang R, Bo S, Lu X, Zhang J, Xu A. Protective effect of carnosine on white matter damage in corpus striatum induced by chronic cerebral hypoperfusion. Neurosci Lett 2018; 683:54-60. [PMID: 29928953 DOI: 10.1016/j.neulet.2018.06.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/25/2018] [Accepted: 06/17/2018] [Indexed: 11/27/2022]
Abstract
Subcortical ischemic vascular dementia caused by chronic cerebral hypoperfusion due to small-artery disease is a common subtype of vascular dementia, which is recognized as the second most prevalent type of dementia. The aim of this study was to determine the effect of carnosine on white matter damage in corpus striatum. Adult male mice (C57BL/6 strain) were subjected to right unilateral common carotid arteries occlusion (rUCCAO), and treated with carnosine or saline. Klüver-Barrera staining, immunohistochemical analyses, Western blots and neurochemical analysis were performed after rUCCAO. The white matter in corpus striatum was damaged at day 37 after rUCCAO, which was largely rescued by carnosine (200, 500 mg/kg). Carnosine (200, 500 mg/kg) significantly recovered the expression of myelin basic protein, suppressed the activation of microglia and reversed the decrease of 5-hydroxytryptamine and dopamine levels in corpus striatum. Moreover, carnosine (200, 500 mg/kg) significantly inhibited the apoptosis in corpus striatum. These data suggest that carnosine has the neuroprotective effect in corpus striatum on rUCCAO in mice, may be due to its protection of neurotransmitters and inhibition of apoptosis.
Collapse
Affiliation(s)
- Jing Ma
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China.
| | - Haijing Yan
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Ranran Wang
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Shuhong Bo
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Xiaotong Lu
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Jian Zhang
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Ajing Xu
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
30
|
Li G, Morris-Blanco KC, Lopez MS, Yang T, Zhao H, Vemuganti R, Luo Y. Impact of microRNAs on ischemic stroke: From pre- to post-disease. Prog Neurobiol 2018; 163-164:59-78. [DOI: 10.1016/j.pneurobio.2017.08.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/12/2017] [Accepted: 08/16/2017] [Indexed: 12/21/2022]
|
31
|
Lang B, Kindy MS, Kozel FA, Schultz SK, Taheri S. Multi-Parametric Classification of Vascular Cognitive Impairment and Dementia: The Impact of Diverse Cerebrovascular Injury Biomarkers. J Alzheimers Dis 2018; 62:39-60. [DOI: 10.3233/jad-170733] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Brittany Lang
- Clinical Psychology Program, University of South Florida, Tampa, FL, USA
| | - Mark S. Kindy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida Tampa, FL, USA
- James A. Haley VA Medical Center, Tampa, FL, USA
| | - F. Andrew Kozel
- James A. Haley VA Medical Center, Tampa, FL, USA
- Psychiatry and Behavioral Sciences, University of South Florida, Tampa, FL, USA
| | - Susan K. Schultz
- James A. Haley VA Medical Center, Tampa, FL, USA
- Psychiatry and Behavioral Sciences, University of South Florida, Tampa, FL, USA
| | - Saeid Taheri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida Tampa, FL, USA
- Byrd Alzheimer’s Institute, Tampa, FL, USA
| |
Collapse
|
32
|
Wallin A, Román GC, Esiri M, Kettunen P, Svensson J, Paraskevas GP, Kapaki E. Update on Vascular Cognitive Impairment Associated with Subcortical Small-Vessel Disease. J Alzheimers Dis 2018; 62:1417-1441. [PMID: 29562536 PMCID: PMC5870030 DOI: 10.3233/jad-170803] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2017] [Indexed: 02/06/2023]
Abstract
Subcortical small-vessel disease (SSVD) is a disorder well characterized from the clinical, imaging, and neuropathological viewpoints. SSVD is considered the most prevalent ischemic brain disorder, increasing in frequency with age. Vascular risk factors include hypertension, diabetes, hyperlipidemia, elevated homocysteine, and obstructive sleep apnea. Ischemic white matter lesions are the hallmark of SSVD; other pathological lesions include arteriolosclerosis, dilatation of perivascular spaces, venous collagenosis, cerebral amyloid angiopathy, microbleeds, microinfarcts, lacunes, and large infarcts. The pathogenesis of SSVD is incompletely understood but includes endothelial changes and blood-brain barrier alterations involving metalloproteinases, vascular endothelial growth factors, angiotensin II, mindin/spondin, and the mammalian target of rapamycin pathway. Metabolic and genetic conditions may also play a role but hitherto there are few conclusive studies. Clinical diagnosis of SSVD includes early executive dysfunction manifested by impaired capacity to use complex information, to formulate strategies, and to exercise self-control. In comparison with Alzheimer's disease (AD), patients with SSVD show less pronounced episodic memory deficits. Brain imaging has advanced substantially the diagnostic tools for SSVD. With the exception of cortical microinfarcts, all other lesions are well visualized with MRI. Diagnostic biomarkers that separate AD from SSVD include reduction of cerebrospinal fluid amyloid-β (Aβ)42 and of the ratio Aβ42/Aβ40 often with increased total tau levels. However, better markers of small-vessel function of intracerebral blood vessels are needed. The treatment of SSVD remains unsatisfactory other than control of vascular risk factors. There is an urgent need of finding targets to slow down and potentially halt the progression of this prevalent, but often unrecognized, disorder.
Collapse
Affiliation(s)
- Anders Wallin
- Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg Sweden and Memory Clinic at Department of Neuropsychiatry, Sahlgrenska University, Hospital, Gothenburg, Sweden
| | - Gustavo C. Román
- Department of Neurology, Methodist Neurological Institute, Houston, TX, USA
- Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Margaret Esiri
- Neuropathology Department, West Wing, John Radcliffe Hospital, Oxford, UK
| | - Petronella Kettunen
- Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg Sweden and Memory Clinic at Department of Neuropsychiatry, Sahlgrenska University, Hospital, Gothenburg, Sweden
- Nuffield Department of Clinical Neurosciences, University of Oxford, West Wing, John Radcliffe Hospital, Oxford, UK
| | - Johan Svensson
- Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - George P. Paraskevas
- 1st Department of Neurology, Neurochemistry Unit, National and Kapodistrian University of Athens, Athens, Greece
| | - Elisabeth Kapaki
- 1st Department of Neurology, Neurochemistry Unit, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
33
|
Gattringer T, Pinter D, Enzinger C, Seifert-Held T, Kneihsl M, Fandler S, Pichler A, Barro C, Gröbke S, Voortman M, Pirpamer L, Hofer E, Ropele S, Schmidt R, Kuhle J, Fazekas F, Khalil M. Serum neurofilament light is sensitive to active cerebral small vessel disease. Neurology 2017; 89:2108-2114. [PMID: 29046363 DOI: 10.1212/wnl.0000000000004645] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 08/31/2017] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To explore whether serum neurofilament light chain protein (NfL) levels are increased in patients with MRI-confirmed recent small subcortical infarcts (RSSI) compared to healthy controls and to determine the subsequent course and determinants of NfL levels in a longitudinal manner. METHODS In a prospectively collected group of symptomatic patients with an RSSI (n = 79, mean age 61 ± 11 years, 67% male), we analyzed brain MRI and serum NfL using a Single Molecule Array (Simoa) assay at baseline and at 3 and 15 months after stroke. Community-dwelling healthy age- and sex-matched individuals with comparable severity of MRI white matter hyperintensities (WMH) (n = 53) served as controls. RESULTS Patients with an RSSI had higher NfL baseline levels compared to controls (73.45 vs 34.59 pg/mL, p < 0.0001), and they were increasingly higher with the time from stroke symptom onset to blood sampling (median 4 days, range 1-11 days, rs = 0.51, p < 0.0001). NfL levels remained increased at the 3-month follow-up but returned to normal at 15 months after stroke. NfL levels were associated with RSSI size and baseline WMH severity and were especially high in patients with new, clinically silent cerebral small vessel disease (CSVD)-related lesions at follow-up. CONCLUSIONS Serum NfL is increased in patients with an RSSI and the occurrence of new CSVD-related MRI lesions, even when clinically silent. This suggests NfL as a blood biomarker for active CSVD.
Collapse
Affiliation(s)
- Thomas Gattringer
- From the Department of Neurology (T.G., D.P., C.E., T.S.-H., M.K., S.F., A.P., M.V., L.P., E.H., S.R., R.S., F.F., M.K.), Division of Neuroradiology, Vascular and Interventional Radiology (C.E.), and Institute for Medical Informatics, Statistics and Documentation (E.H.), Medical University of Graz, Austria; and Neurologic Clinic and Policlinic (C.B., S.G., J,K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland.
| | - Daniela Pinter
- From the Department of Neurology (T.G., D.P., C.E., T.S.-H., M.K., S.F., A.P., M.V., L.P., E.H., S.R., R.S., F.F., M.K.), Division of Neuroradiology, Vascular and Interventional Radiology (C.E.), and Institute for Medical Informatics, Statistics and Documentation (E.H.), Medical University of Graz, Austria; and Neurologic Clinic and Policlinic (C.B., S.G., J,K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Christian Enzinger
- From the Department of Neurology (T.G., D.P., C.E., T.S.-H., M.K., S.F., A.P., M.V., L.P., E.H., S.R., R.S., F.F., M.K.), Division of Neuroradiology, Vascular and Interventional Radiology (C.E.), and Institute for Medical Informatics, Statistics and Documentation (E.H.), Medical University of Graz, Austria; and Neurologic Clinic and Policlinic (C.B., S.G., J,K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Thomas Seifert-Held
- From the Department of Neurology (T.G., D.P., C.E., T.S.-H., M.K., S.F., A.P., M.V., L.P., E.H., S.R., R.S., F.F., M.K.), Division of Neuroradiology, Vascular and Interventional Radiology (C.E.), and Institute for Medical Informatics, Statistics and Documentation (E.H.), Medical University of Graz, Austria; and Neurologic Clinic and Policlinic (C.B., S.G., J,K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Markus Kneihsl
- From the Department of Neurology (T.G., D.P., C.E., T.S.-H., M.K., S.F., A.P., M.V., L.P., E.H., S.R., R.S., F.F., M.K.), Division of Neuroradiology, Vascular and Interventional Radiology (C.E.), and Institute for Medical Informatics, Statistics and Documentation (E.H.), Medical University of Graz, Austria; and Neurologic Clinic and Policlinic (C.B., S.G., J,K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Simon Fandler
- From the Department of Neurology (T.G., D.P., C.E., T.S.-H., M.K., S.F., A.P., M.V., L.P., E.H., S.R., R.S., F.F., M.K.), Division of Neuroradiology, Vascular and Interventional Radiology (C.E.), and Institute for Medical Informatics, Statistics and Documentation (E.H.), Medical University of Graz, Austria; and Neurologic Clinic and Policlinic (C.B., S.G., J,K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Alexander Pichler
- From the Department of Neurology (T.G., D.P., C.E., T.S.-H., M.K., S.F., A.P., M.V., L.P., E.H., S.R., R.S., F.F., M.K.), Division of Neuroradiology, Vascular and Interventional Radiology (C.E.), and Institute for Medical Informatics, Statistics and Documentation (E.H.), Medical University of Graz, Austria; and Neurologic Clinic and Policlinic (C.B., S.G., J,K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Christian Barro
- From the Department of Neurology (T.G., D.P., C.E., T.S.-H., M.K., S.F., A.P., M.V., L.P., E.H., S.R., R.S., F.F., M.K.), Division of Neuroradiology, Vascular and Interventional Radiology (C.E.), and Institute for Medical Informatics, Statistics and Documentation (E.H.), Medical University of Graz, Austria; and Neurologic Clinic and Policlinic (C.B., S.G., J,K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Svenya Gröbke
- From the Department of Neurology (T.G., D.P., C.E., T.S.-H., M.K., S.F., A.P., M.V., L.P., E.H., S.R., R.S., F.F., M.K.), Division of Neuroradiology, Vascular and Interventional Radiology (C.E.), and Institute for Medical Informatics, Statistics and Documentation (E.H.), Medical University of Graz, Austria; and Neurologic Clinic and Policlinic (C.B., S.G., J,K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Margarete Voortman
- From the Department of Neurology (T.G., D.P., C.E., T.S.-H., M.K., S.F., A.P., M.V., L.P., E.H., S.R., R.S., F.F., M.K.), Division of Neuroradiology, Vascular and Interventional Radiology (C.E.), and Institute for Medical Informatics, Statistics and Documentation (E.H.), Medical University of Graz, Austria; and Neurologic Clinic and Policlinic (C.B., S.G., J,K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Lukas Pirpamer
- From the Department of Neurology (T.G., D.P., C.E., T.S.-H., M.K., S.F., A.P., M.V., L.P., E.H., S.R., R.S., F.F., M.K.), Division of Neuroradiology, Vascular and Interventional Radiology (C.E.), and Institute for Medical Informatics, Statistics and Documentation (E.H.), Medical University of Graz, Austria; and Neurologic Clinic and Policlinic (C.B., S.G., J,K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Edith Hofer
- From the Department of Neurology (T.G., D.P., C.E., T.S.-H., M.K., S.F., A.P., M.V., L.P., E.H., S.R., R.S., F.F., M.K.), Division of Neuroradiology, Vascular and Interventional Radiology (C.E.), and Institute for Medical Informatics, Statistics and Documentation (E.H.), Medical University of Graz, Austria; and Neurologic Clinic and Policlinic (C.B., S.G., J,K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Stefan Ropele
- From the Department of Neurology (T.G., D.P., C.E., T.S.-H., M.K., S.F., A.P., M.V., L.P., E.H., S.R., R.S., F.F., M.K.), Division of Neuroradiology, Vascular and Interventional Radiology (C.E.), and Institute for Medical Informatics, Statistics and Documentation (E.H.), Medical University of Graz, Austria; and Neurologic Clinic and Policlinic (C.B., S.G., J,K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Reinhold Schmidt
- From the Department of Neurology (T.G., D.P., C.E., T.S.-H., M.K., S.F., A.P., M.V., L.P., E.H., S.R., R.S., F.F., M.K.), Division of Neuroradiology, Vascular and Interventional Radiology (C.E.), and Institute for Medical Informatics, Statistics and Documentation (E.H.), Medical University of Graz, Austria; and Neurologic Clinic and Policlinic (C.B., S.G., J,K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Jens Kuhle
- From the Department of Neurology (T.G., D.P., C.E., T.S.-H., M.K., S.F., A.P., M.V., L.P., E.H., S.R., R.S., F.F., M.K.), Division of Neuroradiology, Vascular and Interventional Radiology (C.E.), and Institute for Medical Informatics, Statistics and Documentation (E.H.), Medical University of Graz, Austria; and Neurologic Clinic and Policlinic (C.B., S.G., J,K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Franz Fazekas
- From the Department of Neurology (T.G., D.P., C.E., T.S.-H., M.K., S.F., A.P., M.V., L.P., E.H., S.R., R.S., F.F., M.K.), Division of Neuroradiology, Vascular and Interventional Radiology (C.E.), and Institute for Medical Informatics, Statistics and Documentation (E.H.), Medical University of Graz, Austria; and Neurologic Clinic and Policlinic (C.B., S.G., J,K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Michael Khalil
- From the Department of Neurology (T.G., D.P., C.E., T.S.-H., M.K., S.F., A.P., M.V., L.P., E.H., S.R., R.S., F.F., M.K.), Division of Neuroradiology, Vascular and Interventional Radiology (C.E.), and Institute for Medical Informatics, Statistics and Documentation (E.H.), Medical University of Graz, Austria; and Neurologic Clinic and Policlinic (C.B., S.G., J,K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| |
Collapse
|
34
|
Wang Y, Meng R, Song H, Liu G, Hua Y, Cui D, Zheng L, Feng W, Liebeskind DS, Fisher M, Ji X. Remote Ischemic Conditioning May Improve Outcomes of Patients With Cerebral Small-Vessel Disease. Stroke 2017; 48:3064-3072. [PMID: 29042490 DOI: 10.1161/strokeaha.117.017691] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/29/2017] [Accepted: 09/08/2017] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND PURPOSE We aimed to evaluate the efficacy of remote ischemic conditioning (RIC) in patients with cerebral small-vessel disease. METHODS Thirty patients with cerebral small-vessel disease-related mild cognitive impairment were enrolled in this prospective, randomized controlled study for 1 year. Besides routine medical treatment, participants were randomized into the experimental group (n=14) undergoing 5 cycles consisting of ischemia followed by reperfusion for 5 minutes on both upper limbs twice daily for 1 year or the control group (n=16) who were treated with sham ischemia-reperfusion cycles. The primary outcome was the change of brain lesions, and secondary outcomes were changes of cognitive function, plasma biomarkers, and cerebral hemodynamic parameters both at baseline and at the end of 1-year follow-up. RESULTS Compared with pretreatment, the post-treatment white matter hyperintensities volume in the RIC group was significantly reduced (9.10±7.42 versus 6.46±6.05 cm3; P=0.020), whereas no significant difference was observed in the sham-RIC group (8.99±6.81 versus 8.07±6.56 cm3; P=0.085). The reduction of white matter hyperintensities volume in the RIC group was more substantial than that in sham group (-2.632 versus -0.935 cm3; P=0.049). No significant difference was found in the change of the number of lacunes between 2 groups (0 versus 0; P=0.694). A significant treatment difference at 1 year on visuospatial and executive ability was found between the 2 groups (0.639 versus 0.191; P=0.048). RIC showed greater effects compared with sham-RIC on plasma triglyceride (-0.433 versus 0.236 mmol/L; P=0.005), total cholesterol (-0.975 versus 0.134 mmol/L; P<0.001), low-density lipoprotein (-0.645 versus -0.029 mmol/L; P=0.034), and homocysteine (-4.737 versus -1.679 µmol/L; P=0.044). Changes of the pulsation indices of middle cerebral arteries from the baseline to 1 year were different between the 2 groups (right: -0.075 versus 0.043; P=0.030; left: -0.085 versus 0.043; P=0.010). CONCLUSIONS RIC seems to be potentially effective in patients with cerebral small-vessel disease in slowing cognition decline and reducing white matter hyperintensities. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT01658306.
Collapse
Affiliation(s)
- Yuan Wang
- From the Department of Neurology (Y.W., R.M., H.S., G.L.), Department of Neurosurgery (X.J.), Department of Vascular Ultrasound (Y.H.), Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine (D.C., L.Z.), Xuanwu Hospital, Capital Medicine University, Beijing, China; Peking University Health Science Center, Beijing, China (D.C., L.Z.); Department of Neurology, Medical University of South Carolina, Charleston (W.F.); Neurovascular Imaging Research Core and Department of Neurology, University of California in Los Angeles (D.S.L.); and Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.)
| | - Ran Meng
- From the Department of Neurology (Y.W., R.M., H.S., G.L.), Department of Neurosurgery (X.J.), Department of Vascular Ultrasound (Y.H.), Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine (D.C., L.Z.), Xuanwu Hospital, Capital Medicine University, Beijing, China; Peking University Health Science Center, Beijing, China (D.C., L.Z.); Department of Neurology, Medical University of South Carolina, Charleston (W.F.); Neurovascular Imaging Research Core and Department of Neurology, University of California in Los Angeles (D.S.L.); and Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.)
| | - Haiqing Song
- From the Department of Neurology (Y.W., R.M., H.S., G.L.), Department of Neurosurgery (X.J.), Department of Vascular Ultrasound (Y.H.), Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine (D.C., L.Z.), Xuanwu Hospital, Capital Medicine University, Beijing, China; Peking University Health Science Center, Beijing, China (D.C., L.Z.); Department of Neurology, Medical University of South Carolina, Charleston (W.F.); Neurovascular Imaging Research Core and Department of Neurology, University of California in Los Angeles (D.S.L.); and Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.)
| | - Gang Liu
- From the Department of Neurology (Y.W., R.M., H.S., G.L.), Department of Neurosurgery (X.J.), Department of Vascular Ultrasound (Y.H.), Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine (D.C., L.Z.), Xuanwu Hospital, Capital Medicine University, Beijing, China; Peking University Health Science Center, Beijing, China (D.C., L.Z.); Department of Neurology, Medical University of South Carolina, Charleston (W.F.); Neurovascular Imaging Research Core and Department of Neurology, University of California in Los Angeles (D.S.L.); and Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.)
| | - Yang Hua
- From the Department of Neurology (Y.W., R.M., H.S., G.L.), Department of Neurosurgery (X.J.), Department of Vascular Ultrasound (Y.H.), Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine (D.C., L.Z.), Xuanwu Hospital, Capital Medicine University, Beijing, China; Peking University Health Science Center, Beijing, China (D.C., L.Z.); Department of Neurology, Medical University of South Carolina, Charleston (W.F.); Neurovascular Imaging Research Core and Department of Neurology, University of California in Los Angeles (D.S.L.); and Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.)
| | - Dehua Cui
- From the Department of Neurology (Y.W., R.M., H.S., G.L.), Department of Neurosurgery (X.J.), Department of Vascular Ultrasound (Y.H.), Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine (D.C., L.Z.), Xuanwu Hospital, Capital Medicine University, Beijing, China; Peking University Health Science Center, Beijing, China (D.C., L.Z.); Department of Neurology, Medical University of South Carolina, Charleston (W.F.); Neurovascular Imaging Research Core and Department of Neurology, University of California in Los Angeles (D.S.L.); and Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.)
| | - Lemin Zheng
- From the Department of Neurology (Y.W., R.M., H.S., G.L.), Department of Neurosurgery (X.J.), Department of Vascular Ultrasound (Y.H.), Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine (D.C., L.Z.), Xuanwu Hospital, Capital Medicine University, Beijing, China; Peking University Health Science Center, Beijing, China (D.C., L.Z.); Department of Neurology, Medical University of South Carolina, Charleston (W.F.); Neurovascular Imaging Research Core and Department of Neurology, University of California in Los Angeles (D.S.L.); and Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.)
| | - Wuwei Feng
- From the Department of Neurology (Y.W., R.M., H.S., G.L.), Department of Neurosurgery (X.J.), Department of Vascular Ultrasound (Y.H.), Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine (D.C., L.Z.), Xuanwu Hospital, Capital Medicine University, Beijing, China; Peking University Health Science Center, Beijing, China (D.C., L.Z.); Department of Neurology, Medical University of South Carolina, Charleston (W.F.); Neurovascular Imaging Research Core and Department of Neurology, University of California in Los Angeles (D.S.L.); and Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.)
| | - David S Liebeskind
- From the Department of Neurology (Y.W., R.M., H.S., G.L.), Department of Neurosurgery (X.J.), Department of Vascular Ultrasound (Y.H.), Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine (D.C., L.Z.), Xuanwu Hospital, Capital Medicine University, Beijing, China; Peking University Health Science Center, Beijing, China (D.C., L.Z.); Department of Neurology, Medical University of South Carolina, Charleston (W.F.); Neurovascular Imaging Research Core and Department of Neurology, University of California in Los Angeles (D.S.L.); and Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.)
| | - Marc Fisher
- From the Department of Neurology (Y.W., R.M., H.S., G.L.), Department of Neurosurgery (X.J.), Department of Vascular Ultrasound (Y.H.), Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine (D.C., L.Z.), Xuanwu Hospital, Capital Medicine University, Beijing, China; Peking University Health Science Center, Beijing, China (D.C., L.Z.); Department of Neurology, Medical University of South Carolina, Charleston (W.F.); Neurovascular Imaging Research Core and Department of Neurology, University of California in Los Angeles (D.S.L.); and Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.)
| | - Xunming Ji
- From the Department of Neurology (Y.W., R.M., H.S., G.L.), Department of Neurosurgery (X.J.), Department of Vascular Ultrasound (Y.H.), Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine (D.C., L.Z.), Xuanwu Hospital, Capital Medicine University, Beijing, China; Peking University Health Science Center, Beijing, China (D.C., L.Z.); Department of Neurology, Medical University of South Carolina, Charleston (W.F.); Neurovascular Imaging Research Core and Department of Neurology, University of California in Los Angeles (D.S.L.); and Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (M.F.).
| |
Collapse
|
35
|
Sun BL, Wang LH, Yang T, Sun JY, Mao LL, Yang MF, Yuan H, Colvin RA, Yang XY. Lymphatic drainage system of the brain: A novel target for intervention of neurological diseases. Prog Neurobiol 2017; 163-164:118-143. [PMID: 28903061 DOI: 10.1016/j.pneurobio.2017.08.007] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 08/11/2017] [Accepted: 08/31/2017] [Indexed: 12/20/2022]
Abstract
The belief that the vertebrate brain functions normally without classical lymphatic drainage vessels has been held for many decades. On the contrary, new findings show that functional lymphatic drainage does exist in the brain. The brain lymphatic drainage system is composed of basement membrane-based perivascular pathway, a brain-wide glymphatic pathway, and cerebrospinal fluid (CSF) drainage routes including sinus-associated meningeal lymphatic vessels and olfactory/cervical lymphatic routes. The brain lymphatic systems function physiological as a route of drainage for interstitial fluid (ISF) from brain parenchyma to nearby lymph nodes. Brain lymphatic drainage helps maintain water and ion balance of the ISF, waste clearance, and reabsorption of macromolecular solutes. A second physiological function includes communication with the immune system modulating immune surveillance and responses of the brain. These physiological functions are influenced by aging, genetic phenotypes, sleep-wake cycle, and body posture. The impairment and dysfunction of the brain lymphatic system has crucial roles in age-related changes of brain function and the pathogenesis of neurovascular, neurodegenerative, and neuroinflammatory diseases, as well as brain injury and tumors. In this review, we summarize the key component elements (regions, cells, and water transporters) of the brain lymphatic system and their regulators as potential therapeutic targets in the treatment of neurologic diseases and their resulting complications. Finally, we highlight the clinical importance of ependymal route-based targeted gene therapy and intranasal drug administration in the brain by taking advantage of the unique role played by brain lymphatic pathways in the regulation of CSF flow and ISF/CSF exchange.
Collapse
Affiliation(s)
- Bao-Liang Sun
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China.
| | - Li-Hua Wang
- Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, China
| | - Tuo Yang
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jing-Yi Sun
- Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Gangwon 220-701, Republic of Korea
| | - Lei-Lei Mao
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China
| | - Ming-Feng Yang
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China
| | - Hui Yuan
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China
| | - Robert A Colvin
- Department of Biological Sciences, Interdisciplinary Graduate Program in Molecular and Cellular Biology, Neuroscience Program, Ohio University, Athens, OH 45701, USA
| | - Xiao-Yi Yang
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China.
| |
Collapse
|
36
|
Wallin A, Kapaki E, Boban M, Engelborghs S, Hermann DM, Huisa B, Jonsson M, Kramberger MG, Lossi L, Malojcic B, Mehrabian S, Merighi A, Mukaetova-Ladinska EB, Paraskevas GP, Popescu BO, Ravid R, Traykov L, Tsivgoulis G, Weinstein G, Korczyn A, Bjerke M, Rosenberg G. Biochemical markers in vascular cognitive impairment associated with subcortical small vessel disease - A consensus report. BMC Neurol 2017; 17:102. [PMID: 28535786 PMCID: PMC5442599 DOI: 10.1186/s12883-017-0877-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 05/09/2017] [Indexed: 12/17/2022] Open
Abstract
Background Vascular cognitive impairment (VCI) is a heterogeneous entity with multiple aetiologies, all linked to underlying vascular disease. Among these, VCI related to subcortical small vessel disease (SSVD) is emerging as a major homogeneous subtype. Its progressive course raises the need for biomarker identification and/or development for adequate therapeutic interventions to be tested. In order to shed light in the current status on biochemical markers for VCI-SSVD, experts in field reviewed the recent evidence and literature data. Method The group conducted a comprehensive search on Medline, PubMed and Embase databases for studies published until 15.01.2017. The proposal on current status of biochemical markers in VCI-SSVD was reviewed by all co-authors and the draft was repeatedly circulated and discussed before it was finalized. Results This review identifies a large number of biochemical markers derived from CSF and blood. There is a considerable overlap of VCI-SSVD clinical symptoms with those of Alzheimer’s disease (AD). Although most of the published studies are small and their findings remain to be replicated in larger cohorts, several biomarkers have shown promise in separating VCI-SSVD from AD. These promising biomarkers are closely linked to underlying SSVD pathophysiology, namely disruption of blood-CSF and blood–brain barriers (BCB-BBB) and breakdown of white matter myelinated fibres and extracellular matrix, as well as blood and brain inflammation. The leading biomarker candidates are: elevated CSF/blood albumin ratio, which reflects BCB/BBB disruption; altered CSF matrix metalloproteinases, reflecting extracellular matrix breakdown; CSF neurofilment as a marker of axonal damage, and possibly blood inflammatory cytokines and adhesion molecules. The suggested SSVD biomarker deviations contrasts the characteristic CSF profile in AD, i.e. depletion of amyloid beta peptide and increased phosphorylated and total tau. Conclusions Combining SSVD and AD biomarkers may provide a powerful tool to identify with greater precision appropriate patients for clinical trials of more homogeneous dementia populations. Thereby, biomarkers might promote therapeutic progress not only in VCI-SSVD, but also in AD.
Collapse
Affiliation(s)
- A Wallin
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden. .,Memory Clinic at Department of Neuropsychiatry, Sahlgrenska University Hospital, Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Wallinsgatan 6, SE-431 41, Mölndal, Sweden.
| | - E Kapaki
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - M Boban
- Department of Neurology, University Hospital Centre Zagreb, Medical School, University of Zagreb, Zagreb, Croatia
| | - S Engelborghs
- Memory Clinic and Department of Neurology, Hospital Network Antwerp (ZNA) Middelheim and HogeBeuken, Antwerp, Belgium.,Reference Center for Biological Markers of Dementia, Department of Biomedical Sciences, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - D M Hermann
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - B Huisa
- Department of Neurology, University of California, Irvine, California, USA
| | - M Jonsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - M G Kramberger
- Department of Neurology, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - L Lossi
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - B Malojcic
- Department of Neurology, University Hospital Centre Zagreb, Medical School, University of Zagreb, Zagreb, Croatia
| | - S Mehrabian
- Department of Neurology, University Hospital "Alexandrovska", Medical University, Sofia, Bulgaria
| | - A Merighi
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - E B Mukaetova-Ladinska
- Institute of Neuroscience, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - G P Paraskevas
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - B O Popescu
- Department of Neurology, Colentina Clinical Hospital, School of Medicine, 'Carol Davila' University of Medicine and Pharmacy, Bucharest, Romania
| | - R Ravid
- Brain Bank Consultants, Amsterdam, The Netherlands
| | - L Traykov
- Department of Neurology, University Hospital "Alexandrovska", Medical University, Sofia, Bulgaria
| | - G Tsivgoulis
- 2nd Department of Neurology, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - G Weinstein
- School of Public Health, University of Haifa, Haifa, Israel
| | - A Korczyn
- Department of Neurology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - M Bjerke
- Reference Center for Biological Markers of Dementia, Department of Biomedical Sciences, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - G Rosenberg
- University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| |
Collapse
|
37
|
Hay M, Vanderah TW, Samareh-Jahani F, Constantopoulos E, Uprety AR, Barnes CA, Konhilas J. Cognitive impairment in heart failure: A protective role for angiotensin-(1-7). Behav Neurosci 2017; 131:99-114. [PMID: 28054808 DOI: 10.1037/bne0000182] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Patients with congestive heart failure (CHF) have increased hospital readmission rates and mortality if they are concomitantly diagnosed with cognitive decline and memory loss. Accordingly, we developed a preclinical model of CHF-induced cognitive impairment with the goal of developing novel protective therapies against CHF related cognitive decline. CHF was induced by ligation of the left coronary artery to instigate a myocardial infarction (MI). By 4- and 8-weeks post-MI, CHF mice had approximately a 50% and 70% decline in ejection fraction as measured by echocardiography. At both 4- and 8-weeks post-MI, spatial memory performance in CHF mice as tested using the Morris water task was significantly impaired as compared with sham. In addition, CHF mice had significantly worse performance on object recognition when compared with shams as measured by discrimination ratios during the novel object recognition NOR task. At 8-weeks post-MI, a subgroup of CHF mice were given Angiotensin (Ang)-(1-7) (50mcg/kg/hr) subcutaneously for 4 weeks. Following 3 weeks treatment with systemic Ang-(1-7), the CHF mice NOR discrimination ratios were similar to shams and significantly better than the performance of CHF mice treated with saline. Ang-(1-7) also improved spatial memory in CHF mice as compared with shams. Ang-(1-7) had no effect on cardiac function. Inflammatory biomarker studies from plasma revealed a pattern of neuroprotection that may underlie the observed improvements in cognition. These results demonstrate a preclinical mouse model of CHF that exhibits both spatial memory and object recognition dysfunction. Furthermore, this CHF-induced cognitive impairment is attenuated by treatment with systemic Ang-(1-7). (PsycINFO Database Record
Collapse
Affiliation(s)
| | | | | | | | - Ajay R Uprety
- Evelyn F. McKnight Brain Institute, University of Arizona
| | - Carol A Barnes
- Evelyn F. McKnight Brain Institute, University of Arizona
| | - John Konhilas
- Department of Physiology and Sarver Heart Center, University of Arizona
| |
Collapse
|
38
|
Diniz BS, Lin CW, Sibille E, Tseng G, Lotrich F, Aizenstein HJ, Reynolds CF, Butters MA. Circulating biosignatures of late-life depression (LLD): Towards a comprehensive, data-driven approach to understanding LLD pathophysiology. J Psychiatr Res 2016; 82:1-7. [PMID: 27447786 PMCID: PMC9344393 DOI: 10.1016/j.jpsychires.2016.07.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 06/28/2016] [Accepted: 07/05/2016] [Indexed: 11/30/2022]
Abstract
There is scarce information about the pathophysiological processes underlying Late-Life Depression (LLD). We aimed to determine the neurobiological abnormalities related to LLD through a multi-modal biomarker approach combining a large, unbiased peripheral proteomic panel and structural brain imaging. We examined data from 44 LLD and 31 control participants. Plasma proteomic analysis was performed using a multiplex immunoassay. We evaluated the differential protein expression between groups with random intercept models. We carried out enrichment pathway analyses (EPA) to uncover biological pathways and processes related to LLD. Machine learning analysis was applied to the combined dataset to determine the accuracy with which specific proteins could correctly discriminate LLD versus control participants. Sixty-one proteins were differentially expressed in LLD (p < 0.05 and FDR < 0.01). EPA showed that these proteins were related to abnormal immune-inflammatory control, cell survival and proliferation, proteostasis control, lipid metabolism, intracellular signaling. Machine learning analysis showed that a panel of three proteins (C-peptide, FABP-liver, ApoA-IV) discriminated LLD and control participants with 100% accuracy. The plasma proteomic profile in LLD revealed dysregulation in biological processes essential to the maintenance of homeostasis at cellular and systemic levels. These abnormalities increase brain and systemic allostatic load leading to the downstream negative outcomes of LLD, including increased risk of medical comorbidities and dementia. The peripheral biosignature of LLD has predictive power and may suggest novel putative therapeutic targets for prevention, treatment, and neuroprotection in LLD.
Collapse
Affiliation(s)
- Breno Satler Diniz
- Department of Psychiatry & Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chien-Wei Lin
- Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute of CAMH, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - George Tseng
- Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Francis Lotrich
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Howard J. Aizenstein
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Charles F. Reynolds
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Meryl A. Butters
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Corresponding author. 3811 O’Hara Street, Pittsburgh, PA, 15213, USA. (M.A. Butters)
| |
Collapse
|
39
|
Puy V, Zmudka-Attier J, Capel C, Bouzerar R, Serot JM, Bourgeois AM, Ausseil J, Balédent O. Interactions between Flow Oscillations and Biochemical Parameters in the Cerebrospinal Fluid. Front Aging Neurosci 2016; 8:154. [PMID: 27445797 PMCID: PMC4925673 DOI: 10.3389/fnagi.2016.00154] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 06/14/2016] [Indexed: 01/02/2023] Open
Abstract
The equilibrium between the ventricular and lumbar cerebrospinal fluid (CSF) compartments may be disturbed (in terms of flow and biochemistry) in patients with chronic hydrocephalus (CH). Using flow magnetic resonance imaging (MRI) and CSF assays, we sought to determine whether changes in CSF were associated with biochemical alterations. Nine elderly patients with CH underwent phase-contrast MRI. An index of CSF dynamics (Idyn) was defined as the product of the lumbar and ventricular CSF flows. During surgery, samples of CSF were collected from the lumbar and ventricular compartments and assayed for chloride, glucose and total protein. The lumbar/ventricular (L/V) ratio was calculated for each analyte. The ratio between measured and expected levels (Ibioch) was calculated for each analyte and compared with Idyn. Idyn varied from 0 to 100.10(3)μl(2).s(2). In contrast to the L/V ratios for chloride and glucose, the L/V ratio for total protein varied markedly from one patient to another (mean ± standard deviation (SD): 2.63 ± 1.24). The Ibioch for total protein was strongly correlated with the corresponding Idyn (Spearman's R: 0.98; p < 5 × 10(-5)).We observed correlated alterations in CSF flow and biochemical parameters in patients with CH. Our findings also highlight the value of dynamic flow analysis in the interpretation of data on CSF biochemistry.
Collapse
Affiliation(s)
- Vincent Puy
- Biochemistry Unit, CBH, Amiens University Medical CenterAmiens, France; INSERM U1088, Research GroupAmiens, France
| | - Jadwiga Zmudka-Attier
- BioFlowImage Research Group, Jules Verne University of PicardyAmiens, France; Geriatric Unit, General HospitalSaint Quentin, France
| | - Cyrille Capel
- BioFlowImage Research Group, Jules Verne University of PicardyAmiens, France; Neurosurgery Unit, Amiens University Medical CenterAmiens, France
| | - Roger Bouzerar
- BioFlowImage Research Group, Jules Verne University of PicardyAmiens, France; Medical Imaging Unit, Amiens University Medical CenterAmiens, France
| | - Jean-Marie Serot
- BioFlowImage Research Group, Jules Verne University of PicardyAmiens, France; Geriatric Unit, General HospitalSaint Quentin, France
| | | | - Jérome Ausseil
- Biochemistry Unit, CBH, Amiens University Medical CenterAmiens, France; INSERM U1088, Research GroupAmiens, France
| | - Olivier Balédent
- BioFlowImage Research Group, Jules Verne University of PicardyAmiens, France; Medical Imaging Unit, Amiens University Medical CenterAmiens, France
| |
Collapse
|
40
|
Wang Y, Liu G, Hong D, Chen F, Ji X, Cao G. White matter injury in ischemic stroke. Prog Neurobiol 2016; 141:45-60. [PMID: 27090751 PMCID: PMC5677601 DOI: 10.1016/j.pneurobio.2016.04.005] [Citation(s) in RCA: 192] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/01/2016] [Accepted: 04/10/2016] [Indexed: 02/06/2023]
Abstract
Stroke is one of the major causes of disability and mortality worldwide. It is well known that ischemic stroke can cause gray matter injury. However, stroke also elicits profound white matter injury, a risk factor for higher stroke incidence and poor neurological outcomes. The majority of damage caused by stroke is located in subcortical regions and, remarkably, white matter occupies nearly half of the average infarct volume. Indeed, white matter is exquisitely vulnerable to ischemia and is often injured more severely than gray matter. Clinical symptoms related to white matter injury include cognitive dysfunction, emotional disorders, sensorimotor impairments, as well as urinary incontinence and pain, all of which are closely associated with destruction and remodeling of white matter connectivity. White matter injury can be noninvasively detected by MRI, which provides a three-dimensional assessment of its morphology, metabolism, and function. There is an urgent need for novel white matter therapies, as currently available strategies are limited to preclinical animal studies. Optimal protection against ischemic stroke will need to encompass the fortification of both gray and white matter. In this review, we discuss white matter injury after ischemic stroke, focusing on clinical features and tools, such as imaging, manifestation, and potential treatments. We also briefly discuss the pathophysiology of WMI and future research directions.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Neurology, Xuanwu Hospital, Capital University of Medicine, Beijing 100053, China
| | - Gang Liu
- Department of Neurology, Xuanwu Hospital, Capital University of Medicine, Beijing 100053, China
| | - Dandan Hong
- Department of Bioengineering, University of Pittsburgh School of Engineering, United States
| | - Fenghua Chen
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, United States
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital University of Medicine, Beijing 100053, China.
| | - Guodong Cao
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, United States; Geriatric Research Education and Clinical Centers, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, United States.
| |
Collapse
|
41
|
Vilar‐Bergua A, Riba‐Llena I, Vanhooren V, Dewaele S, Libert C, Penalba A, Montaner J, Delgado P. N-glycome Profile Levels Relate to Silent Brain Infarcts in a Cohort of Hypertensives. J Am Heart Assoc 2015; 4:e002669. [PMID: 26597154 PMCID: PMC4845233 DOI: 10.1161/jaha.115.002669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/01/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND Silent brain infarcts (SBIs) are highly prevalent in the aged population and relate to the occurrence of further stroke and dementia. Serum N-glycome levels have been previously associated with aging and they might be related as well to the presence of SBIs and age-related white matter hyperintensities. METHODS AND RESULTS We determined the serum N-glycome profile in a cohort study comprising 972 subjects and evaluated the relationship between N-glycome levels and the presence and number of SBIs and with age-related white matter hyperintensities grades, assessed by brain magnetic resonance imaging. Decreasing concentrations of bigalacto core-α-1,6-fucosylated biantennary glycan and increasing concentrations of branching α-1,3-fucosylated triantennary glycan remained as independent predictors of SBIs (odds ratio 0.4, 95% CI 0.3-0.7 and odds ratio 1.8, 95% CI 1-3.2, respectively), after controlling for the presence of age and classic vascular risk factors. A similar pattern was found to be related to an increasing number of SBIs and white matter hyperintensities grade. CONCLUSIONS N-glycome levels might be potentially useful as biomarkers for the presence of silent cerebrovascular disease.
Collapse
Affiliation(s)
- Andrea Vilar‐Bergua
- Neurovascular Research LaboratoryVall d'Hebron Research InstituteDepartament de MedicinaUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Iolanda Riba‐Llena
- Neurovascular Research LaboratoryVall d'Hebron Research InstituteDepartament de MedicinaUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Valerie Vanhooren
- Inflammation Research CenterUGent‐VIB Research Building FSVMGhentBelgium
- Biomedical Molecular Biology DepartmentGhent UniversityGhentBelgium
| | - Sylviane Dewaele
- Inflammation Research CenterUGent‐VIB Research Building FSVMGhentBelgium
- Biomedical Molecular Biology DepartmentGhent UniversityGhentBelgium
| | - Claude Libert
- Inflammation Research CenterUGent‐VIB Research Building FSVMGhentBelgium
- Biomedical Molecular Biology DepartmentGhent UniversityGhentBelgium
| | - Anna Penalba
- Neurovascular Research LaboratoryVall d'Hebron Research InstituteDepartament de MedicinaUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Joan Montaner
- Neurovascular Research LaboratoryVall d'Hebron Research InstituteDepartament de MedicinaUniversitat Autònoma de BarcelonaBarcelonaSpain
- Stroke UnitNeurology DepartmentVall d'Hebron HospitalBarcelonaSpain
| | - Pilar Delgado
- Neurovascular Research LaboratoryVall d'Hebron Research InstituteDepartament de MedicinaUniversitat Autònoma de BarcelonaBarcelonaSpain
| |
Collapse
|