1
|
Karakaya E, Abdul Y, Edwards J, Jamil S, Albayram O, Ergul A. Complex regulation of tau phosphorylation by the endothelin system in brain microvascular endothelial cells (BMVECs): link to barrier function. Clin Sci (Lond) 2024; 138:1329-1341. [PMID: 39356969 DOI: 10.1042/cs20240616] [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: 03/30/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/04/2024]
Abstract
Endothelin-1 (ET-1), the most potent vasoconstrictor identified to date, contributes to cerebrovascular dysfunction. ET-1 levels in postmortem brain specimens from individuals diagnosed with Alzheimer's disease (AD) and related dementias (ADRD) were shown to be related to cerebral hypoxia and disease severity. ET-1-mediated vascular dysfunction and ensuing cognitive deficits have also been reported in experimental models of AD and ADRD. Moreover, studies also showed that ET-1 secreted from brain microvascular endothelial cells (BMVECs) can affect neurovascular unit integrity in an autocrine and paracrine manner. Vascular contributions to cognitive impairment and dementia (VCID) is a leading ADRD cause known to be free of neuronal tau pathology, a hallmark of AD. However, a recent study reported cytotoxic hyperphosphorylated tau (p-tau) accumulation, which fails to bind or stabilize microtubules in BMVECs in VCID. Thus, the study aimed to determine the impact of ET-1 on tau pathology, microtubule organization, and barrier function in BMVECs. Cells were stimulated with 1 μM ET-1 for 24 h in the presence/absence of ETA (BQ123; 20 μM) or ETB (BQ788; 20 μM) receptor antagonists. Cell lysates were assayed for an array of phosphorylation site-specific antibodies and microtubule organization/stabilization markers. ET-1 stimulation increased p-tau Thr231 but decreased p-tau Ser199, Ser262, Ser396, and Ser214 levels only in the presence of ETA or ETB antagonism. ET-1 also impaired barrier function in the presence of ETA antagonism. These novel findings suggest that (1) dysregulation of endothelial tau phosphorylation may contribute to cerebral microvascular dysfunction and (2) the ET system may be an early intervention target to prevent hyperphosphorylated tau-mediated disruption of BMVEC barrier function.
Collapse
Affiliation(s)
- Eda Karakaya
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, U.S.A
- Ralph H. Johnson Veterans Affairs Health Care System, Charleston, SC, U.S.A
| | - Yasir Abdul
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, U.S.A
- Ralph H. Johnson Veterans Affairs Health Care System, Charleston, SC, U.S.A
| | - Jazlyn Edwards
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, U.S.A
| | - Sarah Jamil
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, U.S.A
- Ralph H. Johnson Veterans Affairs Health Care System, Charleston, SC, U.S.A
| | - Onder Albayram
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, U.S.A
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, U.S.A
| | - Adviye Ergul
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, U.S.A
- Ralph H. Johnson Veterans Affairs Health Care System, Charleston, SC, U.S.A
| |
Collapse
|
2
|
Tyagi SC. Lactobacillus Eats Amyloid Plaque and Post-Biotically Attenuates Senescence Due to Repeat Expansion Disorder and Alzheimer's Disease. Antioxidants (Basel) 2024; 13:1225. [PMID: 39456478 PMCID: PMC11506100 DOI: 10.3390/antiox13101225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Patients with Alzheimer's disease and related dementia (ADRD) are faced with a formidable challenge of focal amyloid deposits and cerebral amyloid angiopathy (CAA). The treatment of amyloid deposits in ADRD by targeting only oxidative stress, inflammation and hyperlipidemia has not yielded significant positive clinical outcomes. The chronic high-fat diet (HFD), or gut dysbiosis, is one of the major contributors of ADRD in part by disrupted transport, epigenetic DNMT1 and the folate 1-carbon metabolism (FOCM) cycle, i.e., rhythmic methylation/de-methylation on DNA, an active part of epigenetic memory during genes turning off and on by the gene writer (DNMT1) and eraser (TET2/FTO) and the transsulfuration pathway by mitochondrial 3-mercaptopyruvate sulfur transferase (3MST)-producing H2S. The repeat CAG expansion and m6A disorder causes senescence and AD. We aim to target the paradigm-shift pathway of the gut-brain microbiome axis that selectively inhibits amyloid deposits and increases mitochondrial transsulfuration and H2S. We have observed an increase in DNMT1 and decreased FTO levels in the cortex of the brain of AD mice. Interestingly, we also observed that probiotic lactobacillus-producing post-biotic folate and lactone/ketone effectively prevented FOCM-associated gut dysbiosis and amyloid deposits. The s-adenosine-methionine (SAM) transporter (SLC25A) was increased by hyperhomocysteinemia (HHcy). Thus, we hypothesize that chronic gut dysbiosis induces SLC25A, the gene writer, and HHcy, and decreases the gene eraser, leading to a decrease in SLC7A and mitochondrial transsulfuration H2S production and bioenergetics. Lactobacillus engulfs lipids/cholesterol and a tri-directional post-biotic, folic acid (an antioxidant and inhibitor of beta amyloid deposits; reduces Hcy levels), and the lactate ketone body (fuel for mitochondria) producer increases SLC7A and H2S (an antioxidant, potent vasodilator and neurotransmitter gas) production and inhibits amyloid deposits. Therefore, it is important to discuss whether lactobacillus downregulates SLC25A and DNMT1 and upregulates TET2/FTO, inhibiting β-amyloid deposits by lowering homocysteine. It is also important to discuss whether lactobacillus upregulates SLC7A and inhibits β-amyloid deposits by increasing the mitochondrial transsulfuration of H2S production.
Collapse
Affiliation(s)
- Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
3
|
Libecap TJ, Pappas CA, Bauer CE, Zachariou V, Raslau FD, Gold BT. Enlarged perivascular space burden predicts declines in cognitive and functional performance. J Neurol Sci 2024; 466:123232. [PMID: 39298972 DOI: 10.1016/j.jns.2024.123232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 09/07/2024] [Accepted: 09/08/2024] [Indexed: 09/22/2024]
Abstract
INTRODUCTION We evaluated the relationship between baseline enlarged perivascular space (ePVS) burden and later cognitive decline. METHODS 83 community-dwelling, older adults (aged 56-86) completed three annual cognitive assessments that included the Clinical Dementia Rating (CDR®) Dementia Staging Instrument Sum of Boxes (CDR-SB) and composite measures of executive function and episodic memory. An MRI scan at baseline was used to count ePVS in the basal ganglia and centrum semiovale. Mixed effects models were run with ePVS as the predictor variable and cognitive measures as the dependent variable. Covariates included age, sex, education, cerebral small vessel disease (cSVD) risk factors, and cSVD neuroimaging biomarkers. RESULTS At baseline, high basal ganglia ePVS counts were associated with lower executive function scores and episodic memory scores. Moreover, baseline basal ganglia ePVS predicted worse longitudinal CDR-SB scores over the study period. DISCUSSION Basal ganglia ePVS burden is a promising biomarker for cSVD-related cognitive and functional decline.
Collapse
Affiliation(s)
- T J Libecap
- MD/PhD Program, University of Kentucky College of Medicine, Lexington, KY, USA; Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Colleen A Pappas
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Christopher E Bauer
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Valentinos Zachariou
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Flavius D Raslau
- Department of Radiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Brian T Gold
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA; Department of Radiology, University of Kentucky College of Medicine, Lexington, KY, USA; Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
4
|
Vitali F, Torrandell-Haro G, Branigan G, Arias Aristizabal J, Reiman E, Bedrick EJ, Brinton RD, Weinkauf C. Asymptomatic carotid artery stenosis is associated with increased Alzheimer's disease and non-Alzheimer's disease dementia risk. Stroke Vasc Neurol 2024:svn-2024-003164. [PMID: 39266210 DOI: 10.1136/svn-2024-003164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 08/23/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND In the absence of a cerebrovascular accident, whether asymptomatic extracranial carotid atherosclerotic disease (aECAD) affects Alzheimer's disease (AD) and non-AD dementia risk is not clear. Understanding whether aECAD is associated with an increased risk for AD is important as it is present in roughly 10% of the population over 60 and could represent a modifiable risk factor for AD and non-AD dementia. METHODS This retrospective cohort study analysed Mariner insurance claims. Enrolment criteria included patients aged 55 years or older with at least 5 years of data and no initial dementia diagnosis. Subjects with and without aECAD were evaluated for subsequent AD and non-AD dementia diagnoses. Propensity score matching was performed using confounding factors identified by logistic regression. χ2 tests and Kaplan-Meier survival curves were used to evaluate the impact of aECAD diagnosis on AD and non-AD dementia risk over time. RESULTS 767 354 patients met enrolment criteria. After propensity score matching, 62 963 subjects with aECAD and 62 963 subjects without ECAD were followed through data records. The aECAD cohort exhibited an increased relative risk of 1.22 (95% CI 1.15 to 1.29, p<0.001) for AD and 1.48 (95% CI 1.38 to 1.59, p<0.001) for non-AD dementias compared with the propensity score-matched cohort without aECAD. The increased AD risk associated with aECAD was evident in patients younger than 75 years old and was less apparent in patients over 75 years of age. CONCLUSIONS aECAD is associated with an increased risk of developing AD and non-AD dementias. These findings underscore the need for further prospective evaluation of interactions between aECAD and dementia, with potential implications for change of clinical care in both of these large patient populations.
Collapse
Affiliation(s)
- Francesca Vitali
- Neurology, The University of Arizona College of Medicine, Tucson, Arizona, USA
- Center for Innovation In Brain Science, The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Georgina Torrandell-Haro
- Center for Innovation In Brain Science, The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Gregory Branigan
- The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Juan Arias Aristizabal
- Department of Surgery, The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Eric Reiman
- Banner Alzheimer's Institute, Phoenix, Arizona, USA
| | - Edward J Bedrick
- Center for Biomedical Informatics and Biostatistics, University of Arizona Medical Center - University Campus, Tucson, Arizona, USA
| | - Roberta Diaz Brinton
- Center for Innovation In Brain Science, The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Craig Weinkauf
- Department of Surgery, The University of Arizona College of Medicine, Tucson, Arizona, USA
| |
Collapse
|
5
|
Pang B, Dong G, Pang T, Sun X, Liu X, Nie Y, Chang X. Advances in pathogenesis and treatment of vascular endothelial injury-related diseases mediated by mitochondrial abnormality. Front Pharmacol 2024; 15:1422686. [PMID: 39281286 PMCID: PMC11394189 DOI: 10.3389/fphar.2024.1422686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/13/2024] [Indexed: 09/18/2024] Open
Abstract
Vascular endothelial cells, serving as a barrier between blood and the arterial wall, play a crucial role in the early stages of the development of atherosclerosis, cardiovascular diseases (CVDs), and Alzheimer's disease (AD). Mitochondria, known as the powerhouses of the cell, are not only involved in energy production but also regulate key biological processes in vascular endothelial cells, including redox signaling, cellular aging, calcium homeostasis, angiogenesis, apoptosis, and inflammatory responses. The mitochondrial quality control (MQC) system is essential for maintaining mitochondrial homeostasis. Current research indicates that mitochondrial dysfunction is a significant driver of endothelial injury and CVDs. This article provides a comprehensive overview of the causes of endothelial injury in CVDs, ischemic stroke in cerebrovascular diseases, and AD, elucidating the roles and mechanisms of mitochondria in these conditions, and aims to develop more effective therapeutic strategies. Additionally, the article offers treatment strategies for cardiovascular and cerebrovascular diseases, including the use of clinical drugs, antioxidants, stem cell therapy, and specific polyphenols, providing new insights and methods for the clinical diagnosis and treatment of related vascular injuries to improve patient prognosis and quality of life. Future research should delve deeper into the molecular and mechanistic links between mitochondrial abnormalities and endothelial injury, and explore how to regulate mitochondrial function to prevent and treat CVDs.
Collapse
Affiliation(s)
- Boxian Pang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Guangtong Dong
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Tieliang Pang
- Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Xinyao Sun
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xin Liu
- Bioscience Department, University of Nottingham, Nottingham, United Kingdom
| | - Yifeng Nie
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Fagerli E, Jackson CW, Escobar I, Ferrier FJ, Perez Lao EJ, Saul I, Gomez J, Dave KR, Bracko O, Perez-Pinzon MA. Resveratrol Mitigates Cognitive Impairments and Cholinergic Cell Loss in the Medial Septum in a Mouse Model of Gradual Cerebral Hypoperfusion. Antioxidants (Basel) 2024; 13:984. [PMID: 39199230 PMCID: PMC11351397 DOI: 10.3390/antiox13080984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
Vascular cognitive impairment and dementia (VCID) is the second leading cause of dementia. There is currently no effective treatment for VCID. Resveratrol (RSV) is considered an antioxidant; however, our group has observed pleiotropic effects in stroke paradigms, suggesting more effects may contribute to mechanistic changes beyond antioxidative properties. The main goal of this study was to investigate if administering RSV twice a week could alleviate cognitive declines following the induction of a VCID model. Additionally, our aim was to further describe whether this treatment regimen could decrease cell death in brain areas vulnerable to changes in cerebral blood flow, such as the hippocampus and medial septum. We hypothesized RSV treatments in a mouse model of gradual cerebral hypoperfusion protect against cognitive impairment. We utilized gradual bilateral common carotid artery stenosis (GBCCAS) via the surgical implantation of ameroid constrictor devices. RSV treatment was administered on the day of implantation and twice a week thereafter. Cerebral perfusion was measured by laser speckle contrast imaging, and cognitive functions, including the recognition memory, the spatial working memory, and associative learning, were assessed by novel object recognition (NOR), Y-maze testing, and contextual fear conditioning (CFC), respectively. RSV treatment did not alleviate cerebral perfusion deficits but mitigated cognitive deficits in CFC and NOR after GBCCAS. Despite these deficits, no hippocampal pathology was observed; however, cholinergic cell loss in the medial septum was significantly increased after GBCCAS. This cholinergic cell loss was mitigated by RSV. This study describes a novel mechanism by which chronic RSV treatments protect against a VCID-induced cognitive decline through the preservation of cholinergic cell viability to improve memory performance.
Collapse
Affiliation(s)
- Eric Fagerli
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Charles W. Jackson
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Iris Escobar
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Fernando J. Ferrier
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Efrain J. Perez Lao
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Isabel Saul
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
| | - Jorge Gomez
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
| | - Kunjan R. Dave
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Oliver Bracko
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
- Department of Biology, University of Miami, Coral Gables, FL 33146, USA
| | - Miguel A. Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA; (E.F.); (C.W.J.); (I.E.); (F.J.F.); (E.J.P.L.); (I.S.); (J.G.); (K.R.D.)
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
7
|
Sarfo FS, Adu-Gyamfi R, Opare-Addo PA, Agyei B, Ampofo M, Nguah SB, Ovbiagele B. Effect of a Cardiovascular Polypill on Poststroke Cognition Among Ghanaians: Secondary Analysis of a Randomized Clinical Trial. J Am Heart Assoc 2024; 13:e034346. [PMID: 39082406 DOI: 10.1161/jaha.124.034346] [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: 01/07/2024] [Accepted: 07/09/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Poststroke cognitive impairment is prevalent worldwide, with no satisfactory preventative therapeutic strategies. We report on the effect of a cardiovascular polypill on cognitive performance among recent stroke survivors. METHODS AND RESULTS The SMAART (Stroke Minimization through Additive Anti-atherosclerotic Agents in Routine Treatment) trial was a phase II randomized trial primarily assessing the polypill versus usual care for secondary prevention after a recent ischemic stroke. Participants allocated to the experimental arm were provided 2 Polycaps taken orally once a day for 12 months. A capsule of Polycap contained aspirin 100 mg, simvastatin 20 mg, hydrochlorothiazide 12.5 mg, ramipril 5 mg, and atenolol 50 mg. Participants in the usual care arm received standard secondary prevention therapy. We compared slopes of the trajectory of raw scores in the executive, language, memory, and visuospatial cognitive domains and aggregated cognitive scores over 12 months via a linear mixed-effects model. We enrolled 148 eligible participants (n=74 in each arm) and 59 versus 64 participants in the polypill and usual care arms, respectively, at month 12. Compared with the usual care arm, the slopes of cognitive performance over 12 months in the polypill arm were steeper by 2.02 units (95% CI, 0.52-3.53), P=0.009 in executive domain, 1.88 units (95% CI, 0.42-3.34), P=0.012 in language domain, 2.60 (0.03-5.17), P=0.049 in memory domain, 0.55 (-0.80 to 1.91), P=0.42 in the visuospatial domain, and global cognitive performance 6.87 units (95% CI, 1.44-12.30), P=0.013. CONCLUSIONS The cardiovascular polypill is associated with a signal of better cognitive performance over 12 months among stroke survivors. Further definitive trials are warranted. REGISTRATION URL: https://www.clinicaltrials.gov; Unique Identifier: NCT03329599.
Collapse
Affiliation(s)
- Fred Stephen Sarfo
- Department of Medicine Kwame Nkrumah University of Science and Technology Kumasi Ghana
- Komfo Anokye Teaching Hospital Kumasi Ghana
| | | | | | | | | | | | | |
Collapse
|
8
|
Whinnery CD, Nie Y, Boskovic DS, Soriano S, Kirsch WM. CD59 Protects Primary Human Cerebrovascular Smooth Muscle Cells from Cytolytic Membrane Attack Complex. Brain Sci 2024; 14:601. [PMID: 38928601 PMCID: PMC11202098 DOI: 10.3390/brainsci14060601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Cerebral amyloid angiopathy is characterized by a weakening of the small- and medium-sized cerebral arteries, as their smooth muscle cells are progressively replaced with acellular amyloid β, increasing vessel fragility and vulnerability to microhemorrhage. In this context, an aberrant overactivation of the complement system would further aggravate this process. The surface protein CD59 protects most cells from complement-induced cytotoxicity, but expression levels can fluctuate due to disease and varying cell types. The degree to which CD59 protects human cerebral vascular smooth muscle (HCSM) cells from complement-induced cytotoxicity has not yet been determined. To address this shortcoming, we selectively blocked the activity of HCSM-expressed CD59 with an antibody, and challenged the cells with complement, then measured cellular viability. Unblocked HCSM cells proved resistant to all tested concentrations of complement, and this resistance decreased progressively with increasing concentrations of anti-CD59 antibody. Complete CD59 blockage, however, did not result in a total loss of cellular viability, suggesting that additional factors may have some protective functions. Taken together, this implies that CD59 plays a predominant role in HCSM cellular protection against complement-induced cytotoxicity. The overexpression of CD59 could be an effective means of protecting these cells from excessive complement system activity, with consequent reductions in the incidence of microhemorrhage. The precise extent to which cellular repair mechanisms and other complement repair proteins contribute to this resistance has yet to be fully elucidated.
Collapse
Affiliation(s)
- Carson D. Whinnery
- Division of Biochemistry, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (C.D.W.); (D.S.B.); (W.M.K.)
- Neurosurgery Center for Research, Training and Education, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA;
| | - Ying Nie
- Neurosurgery Center for Research, Training and Education, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA;
| | - Danilo S. Boskovic
- Division of Biochemistry, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (C.D.W.); (D.S.B.); (W.M.K.)
| | - Salvador Soriano
- Laboratory of Neurodegenerative Diseases, Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Wolff M. Kirsch
- Division of Biochemistry, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (C.D.W.); (D.S.B.); (W.M.K.)
- Neurosurgery Center for Research, Training and Education, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA;
| |
Collapse
|
9
|
Pappas C, Bauer CE, Zachariou V, Maillard P, Caprihan A, Shao X, Wang DJ, Gold BT. MRI free water mediates the association between water exchange rate across the blood brain barrier and executive function among older adults. IMAGING NEUROSCIENCE (CAMBRIDGE, MASS.) 2024; 2:1-15. [PMID: 38947942 PMCID: PMC11211995 DOI: 10.1162/imag_a_00183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 03/27/2024] [Accepted: 05/03/2024] [Indexed: 07/02/2024]
Abstract
Vascular risk factors contribute to cognitive aging, with one such risk factor being dysfunction of the blood brain barrier (BBB). Studies using non-invasive magnetic resonance imaging (MRI) techniques, such as diffusion prepared arterial spin labeling (DP-ASL), can estimate BBB function by measuring water exchange rate (kw). DP-ASL kw has been associated with cognition, but the directionality and strength of the relationship is still under investigation. An additional variable that measures water in extracellular space and impacts cognition, MRI free water (FW), may help explain prior findings. A total of 94 older adults without dementia (Mean age = 74.17 years, 59.6% female) underwent MRI (DP-ASL, diffusion weighted imaging (DWI)) and cognitive assessment. Mean kw was computed across the whole brain (WB), and mean white matter FW was computed across all white matter. The relationship between kw and three cognitive domains (executive function, processing speed, memory) was tested using multiple linear regression. FW was tested as a mediator of the kw-cognitive relationship using the PROCESS macro. A positive association was found between WB kw and executive function [F(4,85) = 7.81, p < .001, R2= 0.269; β = .245, p = .014]. Further, this effect was qualified by subsequent results showing that FW was a mediator of the WB kw-executive function relationship (indirect effect results: standardized effect = .060, bootstrap confidence interval = .0006 to .1411). Results suggest that lower water exchange rate (kw) may contribute to greater total white matter (WM) FW which, in turn, may disrupt executive function. Taken together, proper fluid clearance at the BBB contributes to higher-order cognitive abilities.
Collapse
Affiliation(s)
- Colleen Pappas
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Christopher E. Bauer
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Valentinos Zachariou
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
- Department of Behavioral Science, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Pauline Maillard
- Department of Neurology, University of California at Davis, Davis, CA, United States
- Center for Neurosciences, University of California at Davis, Davis, CA, United States
| | | | - Xingfeng Shao
- Laboratory of FMRI Technology (LOFT), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Danny J.J. Wang
- Laboratory of FMRI Technology (LOFT), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Brian T. Gold
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
- Department of Radiology, College of Medicine, University of Kentucky, Lexington, KY, United States
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
10
|
Liu YJ, Zhao JY, Han WW, Yang HH, Wu XB, Xie F, Wang HP, Wang J, Zhao X, Wan ZX, Chen GC, Qin LQ, Li FR. Microvascular burden and long-term risk of stroke and dementia in type 2 diabetes mellitus. J Affect Disord 2024; 354:68-74. [PMID: 38479499 DOI: 10.1016/j.jad.2024.03.053] [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: 11/26/2023] [Revised: 02/27/2024] [Accepted: 03/09/2024] [Indexed: 04/15/2024]
Abstract
OBJECTIVE To examine the associations between microvascular disease (MVD) and risk of stroke, dementia, and their major subtypes among individuals with type 2 diabetes mellitus (T2DM). METHODS We included 26,173 participants with T2DM from the UK Biobank who had no known stroke or dementia at baseline. MVD burden was reflected by the presence of retinopathy, peripheral neuropathy, and chronic kidney disease. Cox regression models were used to estimate hazard ratios (HRs) and 95 % confidential intervals (CIs) of stroke and dementia associated with overall MVD burden and individual MVD. RESULTS During a median follow-up of 11.5 years, 1103 incident stroke (964 ischemic and 269 hemorrhagic stroke) and 813 incident dementia (312 Alzheimer's disease and 304 vascular dementia) cases were identified. The risk of stroke, dementia, and their major subtypes all increased with an increasing number of MVD (all P-trend <0.001). The adjusted HRs (95 % CIs) comparing three with no MVD were 5.03 (3.16, 8.02) for all stroke, 4.57 (2.75, 7.59) for ischemic stroke, and 6.60 (2.65, 16.43) for hemorrhagic stroke. The corresponding estimates were 4.28 (2.33, 7.86) for all-cause dementia, 6.96 (3.02, 16.01) for Alzheimer's disease, and 3.81 (1.40, 10.42) for vascular dementia. Among the three MVD, chronic kidney disease showed the strongest associations with both stroke subtypes, while peripheral neuropathy was most strongly associated with both dementia subtypes. CONCLUSIONS Risk of stroke, dementia, and their major subtypes increased with an increasing number of MVD. The associations of individual MVD with stroke and dementia varied substantially by types of MVD.
Collapse
Affiliation(s)
- Yu-Jie Liu
- Department of Nutrition and Food Hygiene, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jun-Yu Zhao
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Wen-Wen Han
- Department of Nutrition and Food Hygiene, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Huan-Huan Yang
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Xian-Bo Wu
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Fei Xie
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hai-Peng Wang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Juan Wang
- Changzhou Geriatric Hospital affiliated to Soochow University, Changzhou, China
| | - Xin Zhao
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhong-Xiao Wan
- Department of Nutrition and Food Hygiene, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Guo-Chong Chen
- Department of Nutrition and Food Hygiene, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China.
| | - Li-Qiang Qin
- Department of Nutrition and Food Hygiene, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China; Changzhou Geriatric Hospital affiliated to Soochow University, Changzhou, China.
| | - Fu-Rong Li
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China; School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China; Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Shenzhen, China.
| |
Collapse
|
11
|
van Veluw SJ, Benveniste H, Bakker ENTP, Carare RO, Greenberg SM, Iliff JJ, Lorthois S, Van Nostrand WE, Petzold GC, Shih AY, van Osch MJP. Is CAA a perivascular brain clearance disease? A discussion of the evidence to date and outlook for future studies. Cell Mol Life Sci 2024; 81:239. [PMID: 38801464 PMCID: PMC11130115 DOI: 10.1007/s00018-024-05277-1] [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: 11/14/2023] [Revised: 04/20/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
The brain's network of perivascular channels for clearance of excess fluids and waste plays a critical role in the pathogenesis of several neurodegenerative diseases including cerebral amyloid angiopathy (CAA). CAA is the main cause of hemorrhagic stroke in the elderly, the most common vascular comorbidity in Alzheimer's disease and also implicated in adverse events related to anti-amyloid immunotherapy. Remarkably, the mechanisms governing perivascular clearance of soluble amyloid β-a key culprit in CAA-from the brain to draining lymphatics and systemic circulation remains poorly understood. This knowledge gap is critically important to bridge for understanding the pathophysiology of CAA and accelerate development of targeted therapeutics. The authors of this review recently converged their diverse expertise in the field of perivascular physiology to specifically address this problem within the framework of a Leducq Foundation Transatlantic Network of Excellence on Brain Clearance. This review discusses the overarching goal of the consortium and explores the evidence supporting or refuting the role of impaired perivascular clearance in the pathophysiology of CAA with a focus on translating observations from rodents to humans. We also discuss the anatomical features of perivascular channels as well as the biophysical characteristics of fluid and solute transport.
Collapse
Affiliation(s)
- Susanne J van Veluw
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Erik N T P Bakker
- Department of Biomedical Engineering, Amsterdam University Medical Center, Location AMC, Amsterdam Neuroscience Research Institute, Amsterdam, The Netherlands
| | - Roxana O Carare
- Clinical Neurosciences, University of Southampton, Southampton, UK
| | - Steven M Greenberg
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeffrey J Iliff
- VA Puget Sound Health Care System, University of Washington, Seattle, WA, USA
| | - Sylvie Lorthois
- Institut de Mécanique Des Fluides de Toulouse, IMFT, Université de Toulouse, CNRS, Toulouse, France
| | - William E Van Nostrand
- Department of Biomedical and Pharmaceutical Science, George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| | - Gabor C Petzold
- German Center for Neurodegenerative Disease, Bonn, Germany
- Division of Vascular Neurology, Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Andy Y Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
12
|
Moseholm KF, Horn JW, Fitzpatrick AL, Djoussé L, Longstreth WT, Lopez OL, Hoofnagle AN, Jensen MK, Lemaitre RN, Mukamal KJ. Circulating sphingolipids and subclinical brain pathology: the cardiovascular health study. Front Neurol 2024; 15:1385623. [PMID: 38765262 PMCID: PMC11099203 DOI: 10.3389/fneur.2024.1385623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/08/2024] [Indexed: 05/21/2024] Open
Abstract
Background Sphingolipids are implicated in neurodegeneration and neuroinflammation. We assessed the potential role of circulating ceramides and sphingomyelins in subclinical brain pathology by investigating their association with brain magnetic resonance imaging (MRI) measures and circulating biomarkers of brain injury, neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP) in the Cardiovascular Health Study (CHS), a large and intensively phenotyped cohort of older adults. Methods Brain MRI was offered twice to CHS participants with a mean of 5 years between scans, and results were available from both time points in 2,116 participants (mean age 76 years; 40% male; and 25% APOE ε4 allele carriers). We measured 8 ceramide and sphingomyelin species in plasma samples and examined the associations with several MRI, including worsening grades of white matter hyperintensities and ventricular size, number of brain infarcts, and measures of brain atrophy in a subset with quantitative measures. We also investigated the sphingolipid associations with serum NfL and GFAP. Results In the fully adjusted model, higher plasma levels of ceramides and sphingomyelins with a long (16-carbon) saturated fatty acid were associated with higher blood levels of NfL [β = 0.05, false-discovery rate corrected P (PFDR) = 0.004 and β = 0.06, PFDR = < 0.001, respectively]. In contrast, sphingomyelins with very long (20- and 22-carbon) saturated fatty acids tended to have an inverse association with levels of circulating NfL. In secondary analyses, we found an interaction between ceramide d18:1/20:0 and sex (P for interaction = <0.001), such that ceramide d18:1/20:0 associated with higher odds for infarcts in women [OR = 1.26 (95%CI: 1.07, 1.49), PFDR = 0.03]. We did not observe any associations with GFAP blood levels, white matter grade, ventricular grade, mean bilateral hippocampal volume, or total brain volume. Conclusion Overall, our comprehensive investigation supports the evidence that ceramides and sphingomyelins are associated with increased aging brain pathology and that the direction of association depends on the fatty acid attached to the sphingosine backbone.
Collapse
Affiliation(s)
- Kristine F. Moseholm
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Copenhagen, Denmark
| | - Jens W. Horn
- Department of Internal Medicine, Levanger Hospital, Health Trust Nord-Trøndelag, Levanger, Norway
| | - Annette L. Fitzpatrick
- Departments of Family Medicine and Epidemiology, School of Public Health, University of Washington, Seattle, WA, United States
| | - Luc Djoussé
- Division of Aging, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - W. T. Longstreth
- Departments of Family Medicine and Epidemiology, School of Public Health, University of Washington, Seattle, WA, United States
- Department of Neurology, School of Medicine, University of Washington, Seattle, WA, United States
| | - Oscar L. Lopez
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Andrew N. Hoofnagle
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, United States
| | - Majken K. Jensen
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Copenhagen, Denmark
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Rozenn N. Lemaitre
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Kenneth J. Mukamal
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
| |
Collapse
|
13
|
He Y, He T, Li H, Chen W, Zhong B, Wu Y, Chen R, Hu Y, Ma H, Wu B, Hu W, Han Z. Deciphering mitochondrial dysfunction: Pathophysiological mechanisms in vascular cognitive impairment. Biomed Pharmacother 2024; 174:116428. [PMID: 38599056 DOI: 10.1016/j.biopha.2024.116428] [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: 12/20/2023] [Revised: 02/26/2024] [Accepted: 03/08/2024] [Indexed: 04/12/2024] Open
Abstract
Vascular cognitive impairment (VCI) encompasses a range of cognitive deficits arising from vascular pathology. The pathophysiological mechanisms underlying VCI remain incompletely understood; however, chronic cerebral hypoperfusion (CCH) is widely acknowledged as a principal pathological contributor. Mitochondria, crucial for cellular energy production and intracellular signaling, can lead to numerous neurological impairments when dysfunctional. Recent evidence indicates that mitochondrial dysfunction-marked by oxidative stress, disturbed calcium homeostasis, compromised mitophagy, and anomalies in mitochondrial dynamics-plays a pivotal role in VCI pathogenesis. This review offers a detailed examination of the latest insights into mitochondrial dysfunction within the VCI context, focusing on both the origins and consequences of compromised mitochondrial health. It aims to lay a robust scientific groundwork for guiding the development and refinement of mitochondrial-targeted interventions for VCI.
Collapse
Affiliation(s)
- Yuyao He
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Tiantian He
- Sichuan Academy of Chinese Medicine Sciences, China
| | - Hongpei Li
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Wei Chen
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Biying Zhong
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yue Wu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Runming Chen
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yuli Hu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Huaping Ma
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Bin Wu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Wenyue Hu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China.
| | - Zhenyun Han
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China.
| |
Collapse
|
14
|
Rahman MS, Islam R, Bhuiyan MIH. Ion transporter cascade, reactive astrogliosis and cerebrovascular diseases. Front Pharmacol 2024; 15:1374408. [PMID: 38659577 PMCID: PMC11041382 DOI: 10.3389/fphar.2024.1374408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/21/2024] [Indexed: 04/26/2024] Open
Abstract
Cerebrovascular diseases and their sequalae, such as ischemic stroke, chronic cerebral hypoperfusion, and vascular dementia are significant contributors to adult disability and cognitive impairment in the modern world. Astrocytes are an integral part of the neurovascular unit in the CNS and play a pivotal role in CNS homeostasis, including ionic and pH balance, neurotransmission, cerebral blood flow, and metabolism. Astrocytes respond to cerebral insults, inflammation, and diseases through unique molecular, morphological, and functional changes, collectively known as reactive astrogliosis. The function of reactive astrocytes has been a subject of debate. Initially, astrocytes were thought to primarily play a supportive role in maintaining the structure and function of the nervous system. However, recent studies suggest that reactive astrocytes may have both beneficial and detrimental effects. For example, in chronic cerebral hypoperfusion, reactive astrocytes can cause oligodendrocyte death and demyelination. In this review, we will summarize the (1) roles of ion transporter cascade in reactive astrogliosis, (2) role of reactive astrocytes in vascular dementia and related dementias, and (3) potential therapeutic approaches for dementing disorders targeting reactive astrocytes. Understanding the relationship between ion transporter cascade, reactive astrogliosis, and cerebrovascular diseases may reveal mechanisms and targets for the development of therapies for brain diseases associated with reactive astrogliosis.
Collapse
Affiliation(s)
- Md Shamim Rahman
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX, United States
| | | | - Mohammad Iqbal H. Bhuiyan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX, United States
| |
Collapse
|
15
|
Smith EE. Introduction to Focused Update Vascular Cognitive Impairment and Dementia: Better Diagnosis, More Avenues for Prevention. Stroke 2024; 55:788-790. [PMID: 38527146 DOI: 10.1161/strokeaha.124.046673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Affiliation(s)
- Eric E Smith
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Alberta, Canada
| |
Collapse
|
16
|
Whinnery C, Nie Y, Boskovic DS, Soriano S, Kirsch WM. CD59 Protects Primary Human Cerebrovascular Smooth Muscle Cells from Cytolytic Membrane Attack Complex. RESEARCH SQUARE 2024:rs.3.rs-4165045. [PMID: 38645247 PMCID: PMC11030535 DOI: 10.21203/rs.3.rs-4165045/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Cerebral amyloid angiopathy is characterized by a weakening of the small and medium sized cerebral arteries, as their smooth muscle cells are progressively replaced with acellular amyloid β, increasing vessel fragility and vulnerability to microhemorrhage. In this context, an aberrant overactivation of the complement system would further aggravate this process. The surface protein CD59 protects most cells from complement-induced cytotoxicity, but expression levels can fluctuate due to disease and vary between cell types. The degree to which CD59 protects human cerebral vascular smooth muscle (HCSM) cells from complement-induced cytotoxicity has not yet been determined. To address this shortcoming, we selectively blocked the activity of HCSM-expressed CD59 with an antibody and challenged the cells with complement, then measured cellular viability. Unblocked HCSM cells proved resistant to all tested concentrations of complement, and this resistance decreased progressively with increasing concentrations of anti-CD59 antibody. Complete CD59 blockage, however, did not result in total loss of cellular viability, suggesting that additional factors may have some protective functions. Taken together, this implies that CD59 plays a predominant role in HCSM cellular protection against complement-induced cytotoxicity. Over-expression of CD59 could be an effective means of protecting these cells from excessive complement system activity, with consequent reduction in the incidence of microhemorrhage. The precise extent to which cellular repair mechanisms and other complement repair proteins contribute to this resistance has yet to be fully elucidated.
Collapse
|
17
|
Bhuiyan MIH, Habib K, Sultan MT, Chen F, Jahan I, Weng Z, Rahman MS, Islam R, Foley LM, Hitchens TK, Deng X, Canna SW, Sun D, Cao G. SPAK inhibitor ZT-1a attenuates reactive astrogliosis and oligodendrocyte degeneration in a mouse model of vascular dementia. CNS Neurosci Ther 2024; 30:e14654. [PMID: 38433018 PMCID: PMC10909630 DOI: 10.1111/cns.14654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/08/2024] [Accepted: 01/28/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Astrogliosis and white matter lesions (WML) are key characteristics of vascular contributions to cognitive impairment and dementia (VCID). However, the molecular mechanisms underlying VCID remain poorly understood. Stimulation of Na-K-Cl cotransport 1 (NKCC1) and its upstream kinases WNK (with no lysine) and SPAK (the STE20/SPS1-related proline/alanine-rich kinase) play a role in astrocytic intracellular Na+ overload, hypertrophy, and swelling. Therefore, in this study, we assessed the effect of SPAK inhibitor ZT-1a on pathogenesis and cognitive function in a mouse model of VCID induced by bilateral carotid artery stenosis (BCAS). METHODS Following sham or BCAS surgery, mice were randomly assigned to receive either vehicle (DMSO) or SPAK inhibitor ZT-1a treatment regimen (days 14-35 post-surgery). Mice were then evaluated for cognitive functions by Morris water maze, WML by ex vivo MRI-DTI analysis, and astrogliosis/demyelination by immunofluorescence and immunoblotting. RESULTS Compared to sham control mice, BCAS-Veh mice exhibited chronic cerebral hypoperfusion and memory impairments, accompanied by significant MRI DTI-detected WML and oligodendrocyte (OL) death. Increased activation of WNK-SPAK-NKCC1-signaling proteins was detected in white matter tissues and in C3d+ GFAP+ cytotoxic astrocytes but not in S100A10+ GFAP+ homeostatic astrocytes in BCAS-Veh mice. In contrast, ZT-1a-treated BCAS mice displayed reduced expression and phosphorylation of NKCC1, decreased astrogliosis, OL death, and WML, along with improved memory functions. CONCLUSION BCAS-induced upregulation of WNK-SPAK-NKCC1 signaling contributes to white matter-reactive astrogliosis, OL death, and memory impairment. Pharmacological inhibition of the SPAK activity has therapeutic potential for alleviating pathogenesis and memory impairment in VCID.
Collapse
Affiliation(s)
- Mohammad Iqbal H. Bhuiyan
- Department of Pharmaceutical Sciences, School of PharmacyUniversity of Texas at El PasoEl PasoTexasUSA
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Pittsburgh Institute for Neurodegenerative DisordersUniversity of PittsburghPittsburghPennsylvaniaUSA
- Veterans Affairs Pittsburgh Health Care System Pittsburgh Healthcare SystemGeriatric Research Education and Clinical CenterPittsburghPennsylvaniaUSA
| | - Khadija Habib
- Department of Pharmaceutical Sciences, School of PharmacyUniversity of Texas at El PasoEl PasoTexasUSA
| | - Md Tipu Sultan
- Department of Pharmaceutical Sciences, School of PharmacyUniversity of Texas at El PasoEl PasoTexasUSA
| | - Fenghua Chen
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Israt Jahan
- Department of Pharmaceutical Sciences, School of PharmacyUniversity of Texas at El PasoEl PasoTexasUSA
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Zhongfang Weng
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Md Shamim Rahman
- Department of Pharmaceutical Sciences, School of PharmacyUniversity of Texas at El PasoEl PasoTexasUSA
| | | | - Lesley M. Foley
- Animal Imaging CenterUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - T. Kevin Hitchens
- Animal Imaging CenterUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of NeurobiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Xianming Deng
- State Key Laboratory of Cellular Stress Biology, School of Life SciencesXiamen UniversityXiamenFujianChina
| | - Scott W. Canna
- Department of Pediatric RheumatologyThe Children's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Dandan Sun
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Pittsburgh Institute for Neurodegenerative DisordersUniversity of PittsburghPittsburghPennsylvaniaUSA
- Veterans Affairs Pittsburgh Health Care System Pittsburgh Healthcare SystemGeriatric Research Education and Clinical CenterPittsburghPennsylvaniaUSA
| | - Guodong Cao
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Veterans Affairs Pittsburgh Health Care System Pittsburgh Healthcare SystemGeriatric Research Education and Clinical CenterPittsburghPennsylvaniaUSA
| |
Collapse
|
18
|
Mukli P, Pinto CB, Owens CD, Csipo T, Lipecz A, Szarvas Z, Peterfi A, Langley ACDCP, Hoffmeister J, Racz FS, Perry JW, Tarantini S, Nyúl‐Tóth Á, Sorond FA, Yang Y, James JA, Kirkpatrick AC, Prodan CI, Toth P, Galindo J, Gardner AW, Sonntag WE, Csiszar A, Ungvari Z, Yabluchanskiy A. Impaired Neurovascular Coupling and Increased Functional Connectivity in the Frontal Cortex Predict Age-Related Cognitive Dysfunction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303516. [PMID: 38155460 PMCID: PMC10962492 DOI: 10.1002/advs.202303516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 11/19/2023] [Indexed: 12/30/2023]
Abstract
Impaired cerebrovascular function contributes to the genesis of age-related cognitive decline. In this study, the hypothesis is tested that impairments in neurovascular coupling (NVC) responses and brain network function predict cognitive dysfunction in older adults. Cerebromicrovascular and working memory function of healthy young (n = 21, 33.2±7.0 years) and aged (n = 30, 75.9±6.9 years) participants are assessed. To determine NVC responses and functional connectivity (FC) during a working memory (n-back) paradigm, oxy- and deoxyhemoglobin concentration changes from the frontal cortex using functional near-infrared spectroscopy are recorded. NVC responses are significantly impaired during the 2-back task in aged participants, while the frontal networks are characterized by higher local and global connection strength, and dynamic FC (p < 0.05). Both impaired NVC and increased FC correlate with age-related decline in accuracy during the 2-back task. These findings suggest that task-related brain states in older adults require stronger functional connections to compensate for the attenuated NVC responses associated with working memory load.
Collapse
|
19
|
Alshammari A, Pillai B, Kamat P, Jones TW, Bosomtwi A, Khan MB, Hess DC, Li W, Somanath PR, Sayed MA, Ergul A, Fagan SC. Angiotensin II Type 2 Receptor Agonism Alleviates Progressive Post-stroke Cognitive Impairment in Aged Spontaneously Hypertensive Rats. Transl Stroke Res 2024:10.1007/s12975-024-01232-1. [PMID: 38302738 DOI: 10.1007/s12975-024-01232-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/27/2023] [Accepted: 01/17/2024] [Indexed: 02/03/2024]
Abstract
Hypertension and aging are leading risk factors for stroke and vascular contributions to cognitive impairment and dementia (VCID). Most animal models fail to capture the complex interplay between these pathophysiological processes. In the current study, we examined the development of cognitive impairment in 18-month-old spontaneously hypertensive rats (SHR) before and following ischemic stroke. Sixty SHRs were housed for 18 months with cognitive assessments every 6 months and post-surgery. MRI scans were performed at baseline and throughout the study. On day 3 post-stroke, rats were randomized to receive either angiotensin II type 2 receptor (AT2R) agonist Compound 21 (C21) or plain water for 8 weeks. SHRs demonstrated a progressive cognitive decline and significant MRI abnormalities before stroke. Perioperative mortality within 72 h of stroke was low. Stroke resulted in significant acute brain swelling, chronic brain atrophy, and sustained sensorimotor and behavioral deficits. There was no evidence of anhedonia at week 8. C21 enhanced sensorimotor recovery and ischemic lesion resolution at week 8. SHRs represent a clinically relevant animal model to study aging and stroke-associated VCID. This study underscores the importance of translational disease modeling and provides evidence that modulation of the AT2R signaling via C21 may be a useful therapeutic option to improve sensorimotor and cognitive outcomes even in aged animals.
Collapse
Affiliation(s)
- Abdulkarim Alshammari
- Program in Clinical and Experimental Therapeutics, Charlie Norwood Veterans Affairs Health Care System and College of Pharmacy, University of Georgia, Augusta, GA, USA
- Department of Clinical Pharmacy, Faculty of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Bindu Pillai
- Program in Clinical and Experimental Therapeutics, Charlie Norwood Veterans Affairs Health Care System and College of Pharmacy, University of Georgia, Augusta, GA, USA
| | - Pradip Kamat
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Timothy W Jones
- Program in Clinical and Experimental Therapeutics, Charlie Norwood Veterans Affairs Health Care System and College of Pharmacy, University of Georgia, Augusta, GA, USA
| | - Asamoah Bosomtwi
- Georgia Cancer Center and Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | | | - David C Hess
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Weiguo Li
- Ralph H. Johnson Veterans Affairs Health Care System and Department of Pathology & Lab. Medicine, Medical University of South Carolina, 171 Ashley Ave. MSC 908, Charleston, SC, 29492, USA
| | - Payaningal R Somanath
- Program in Clinical and Experimental Therapeutics, Charlie Norwood Veterans Affairs Health Care System and College of Pharmacy, University of Georgia, Augusta, GA, USA
| | | | - Adviye Ergul
- Ralph H. Johnson Veterans Affairs Health Care System and Department of Pathology & Lab. Medicine, Medical University of South Carolina, 171 Ashley Ave. MSC 908, Charleston, SC, 29492, USA.
| | - Susan C Fagan
- Program in Clinical and Experimental Therapeutics, Charlie Norwood Veterans Affairs Health Care System and College of Pharmacy, University of Georgia, Augusta, GA, USA
| |
Collapse
|
20
|
Yan L, Wu L, Li H, Qian Y, Wang M, Wang Y, Dou B, Yu T. Effect of non-invasive neuromodulation techniques on vascular cognitive impairment: A Bayesian network meta-analysis protocol. PLoS One 2024; 19:e0284447. [PMID: 38175852 PMCID: PMC10766188 DOI: 10.1371/journal.pone.0284447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 03/30/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND VCI is a severe public health problem facing the world today. In addition to pharmacological treatment, non-invasive neuromodulation techniques have also been effective. At this stage, non-invasive neuromodulation techniques combined with pharmacological treatment are the mainstay of clinical treatment, and clinical trials are continuing to be conducted, which is becoming the direction of treatment for VCI. Therefore, we outline this systematic review and network meta-analysis protocol to evaluate and rank clinical data in future studies which can develop optimal protocols for the clinical treatment of VCI with non-invasive neuromodulation techniques in combination with drugs. METHODS The network meta-analysis will search eight databases, including PubMed, Embase, Cochrane Library, Web of Science, China Knowledge Infrastructure Library (CNKI), China Biology Medicine disc (CBM)), Wanfang Data Knowledge Service Platform and Vipshop Journal Service Platform (VIP), for a period of from the establishment of the library to January 30 2022. The quality of the studies will be evaluated using the Cochrane Review's Handbook 5.1 and the PEDro scale to assess the evidence and quality of the included randomised controlled trials. Risk of bias assessment and heterogeneity tests will be performed using the Review Manager 5.4 program, and Bayesian network meta-analysis will be performed using the Stata 16.0 and WinBUGS 1.4.3 program. RESULTS The results of the network meta-analysis will be published in a peer-reviewed journal. CONCLUSIONS Our study is expected to provide high quality evidence-based medical evidence for the treatment of VCI by clinicians. TRIAL REGISTRATION PROSPERO: CRD42022308580.
Collapse
Affiliation(s)
- Long Yan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P. R. China
| | - Linna Wu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P. R. China
| | - Hong Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P. R. China
| | - Yulin Qian
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Meng Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Yu Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Baomin Dou
- Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Tao Yu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| |
Collapse
|
21
|
Steiner K, Humpel C. Beta-Amyloid Enhances Vessel Formation in Organotypic Brain Slices Connected to Microcontact Prints. Biomolecules 2023; 14:3. [PMID: 38275744 PMCID: PMC10812928 DOI: 10.3390/biom14010003] [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: 11/03/2023] [Revised: 11/24/2023] [Accepted: 12/13/2023] [Indexed: 01/27/2024] Open
Abstract
In Alzheimer's disease, the blood-brain barrier breakdown, blood vessel damage and re-organization are early events. Deposits of the small toxic peptide beta-amyloid (Aβ) cause the formation of extracellular plaques and accumulate in vessels disrupting the blood flow but may also play a role in blood clotting. In the present study, we aim to explore the impact of Aβ on the migration of endothelial cells and subsequent vessel formation. We use organotypic brain slices of postnatal day 10 wildtype mice (C57BL/6) and connect them to small microcontact prints (µCPs) of collagen. Our data show that laminin-positive endothelial cells migrate onto collagen µCPs, but without any vessel formation after 4 weeks. When the µCPs are loaded with human Aβ40, (aggregated) human Aβ42 and mouse Aβ42 peptides, the number and migration distance of endothelial cells are significantly reduced, but with a more pronounced subsequent vessel formation. The vessel formation is verified by zonula occludens (ZO)-1 and -2 stainings and confocal microscopy. In addition, the vessel formation is accompanied by a stronger GFAP-positive astroglial formation. Finally, we show that vessels can grow towards convergence when two opposed slices are connected via microcontact-printed lanes. In conclusion, our data show that Aβ promotes vessel formation, and organotypic brain slices connected to collagen µCPs provide a potent tool to study vessel formation.
Collapse
Affiliation(s)
| | - Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| |
Collapse
|
22
|
Saks DG, Smith EE, Sachdev PS. National and international collaborations to advance research into vascular contributions to cognitive decline. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2023; 6:100195. [PMID: 38226362 PMCID: PMC10788430 DOI: 10.1016/j.cccb.2023.100195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 01/17/2024]
Abstract
Cerebrovascular disease is the second most common cause of cognitive disorders, usually referred to as vascular contributions to cognitive impairment and dementia (VCID) and makes some contribution to about 70 % of all dementias. Despite its importance, research into VCID has lagged as compared to cognitive impairment due to Alzheimer's disease. There is an increasing appreciation that closing this gap requires large national and international collaborations. This paper highlights 24 notable large-scale national and international efforts to advance research into VCID (MarkVCID, DiverseVCID, DISCOVERY, COMPASS-ND, HBC, RHU SHIVA, UK DRI Vascular Theme, STROKOG, Meta VCI Map, ISGC, ENIGMA-Stroke Recovery, CHARGE, SVDs@target, BRIDGET, CADASIL Consortium, CADREA, AusCADASIL, DPUK, DPAU, STRIVE, HARNESS, FINESSE, VICCCS, VCD-CRE Delphi). These collaborations aim to investigate the effects on cognition from cerebrovascular disease or impaired cerebral blood flow, the mechanisms of action, means of prevention and avenues for treatment. Consensus groups have been developed to harmonise global approaches to VCID, standardise terminology and inform management and treatment, and data sharing is becoming the norm. VCID research is increasingly a global collaborative enterprise which bodes well for rapid advances in this field.
Collapse
Affiliation(s)
- Danit G Saks
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Eric E Smith
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
23
|
Adeniyi PA, Gong X, MacGregor E, Degener-O’Brien K, McClendon E, Garcia M, Romero O, Russell J, Srivastava T, Miller J, Keene CD, Back SA. Ferroptosis of Microglia in Aging Human White Matter Injury. Ann Neurol 2023; 94:1048-1066. [PMID: 37605362 PMCID: PMC10840747 DOI: 10.1002/ana.26770] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/07/2023] [Accepted: 08/14/2023] [Indexed: 08/23/2023]
Abstract
OBJECTIVE Because the role of white matter (WM) degenerating microglia (DM) in remyelination failure is unclear, we sought to define the core features of this novel population of aging human microglia. METHODS We analyzed postmortem human brain tissue to define a population of DM in aging WM lesions. We used immunofluorescence staining and gene expression analysis to investigate molecular mechanisms related to the degeneration of DM. RESULTS We found that DM, which accumulated myelin debris were selectively enriched in the iron-binding protein light chain ferritin, and accumulated PLIN2-labeled lipid droplets. DM displayed lipid peroxidation injury and enhanced expression for TOM20, a mitochondrial translocase, and a sensor of oxidative stress. DM also displayed enhanced expression of the DNA fragmentation marker phospho-histone H2A.X. We identified a unique set of ferroptosis-related genes involving iron-mediated lipid dysmetabolism and oxidative stress that were preferentially expressed in WM injury relative to gray matter neurodegeneration. INTERPRETATION Ferroptosis appears to be a major mechanism of WM injury in Alzheimer's disease and vascular dementia. WM DM are a novel therapeutic target to potentially reduce the impact of WM injury and myelin loss on the progression of cognitive impairment. ANN NEUROL 2023;94:1048-1066.
Collapse
Affiliation(s)
- Philip A. Adeniyi
- Departments of Pediatrics and, Oregon Health & Science University, Portland, Oregon, USA
| | - Xi Gong
- Departments of Pediatrics and, Oregon Health & Science University, Portland, Oregon, USA
| | - Ellie MacGregor
- Departments of Pediatrics and, Oregon Health & Science University, Portland, Oregon, USA
| | - Kiera Degener-O’Brien
- Departments of Pediatrics and, Oregon Health & Science University, Portland, Oregon, USA
| | - Evelyn McClendon
- Departments of Pediatrics and, Oregon Health & Science University, Portland, Oregon, USA
| | - Mariel Garcia
- Departments of Pediatrics and, Oregon Health & Science University, Portland, Oregon, USA
| | - Oscar Romero
- Departments of Pediatrics and, Oregon Health & Science University, Portland, Oregon, USA
| | - Joshua Russell
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Taasin Srivastava
- Departments of Pediatrics and, Oregon Health & Science University, Portland, Oregon, USA
| | - Jeremy Miller
- Allen Institute for Brain Science, Seattle, Washington, USA
| | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Stephen A. Back
- Departments of Pediatrics and, Oregon Health & Science University, Portland, Oregon, USA
- Neurology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
24
|
Zhang Q, Liu X, Gao S, Yan S, Li A, Wei Z, Han S, Hou Y, Li X, Cao D, Yue J. Multimodal magnetic resonance imaging on brain structure and function changes in vascular cognitive impairment without dementia. Front Aging Neurosci 2023; 15:1278390. [PMID: 38035274 PMCID: PMC10687453 DOI: 10.3389/fnagi.2023.1278390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
Vascular cognitive impairment not dementia (VCIND) is one of the three subtypes of vascular cognitive impairment (VCI), with cognitive dysfunction and symptoms ranging between normal cognitive function and vascular dementia. The specific mechanisms underlying VCIND are still not fully understood, and there is a lack of specific diagnostic markers in clinical practice. With the rapid development of magnetic resonance imaging (MRI) technology, structural MRI (sMRI) and functional MRI (fMRI) have become effective methods for exploring the neurobiological mechanisms of VCIND and have made continuous progress. This article provides a comprehensive overview of the research progress in VCIND using multimodal MRI, including sMRI, diffusion tensor imaging, resting-state fMRI, and magnetic resonance spectroscopy. By integrating findings from these multiple modalities, this study presents a novel perspective on the neuropathological mechanisms underlying VCIND. It not only highlights the importance of multimodal MRI in unraveling the complex nature of VCIND but also lays the foundation for future research examining the relationship between brain structure, function, and cognitive impairment in VCIND. These new perspectives and strategies ultimately hold the potential to contribute to the development of more effective diagnostic tools and therapeutic interventions for VCIND.
Collapse
Affiliation(s)
- Qinhong Zhang
- Shenzhen Frontiers in Chinese Medicine Research Co., Ltd., Shenzhen, China
- Department of Acupuncture and Moxibustion, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiao Liu
- Department of Pediatrics, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shenglan Gao
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shiyan Yan
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ang Li
- Servier (Beijing) Pharmaceutical Research and Development Co., Ltd., Beijing, China
| | - Zeyi Wei
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shengwang Han
- Third Ward of Rehabilitation Department, Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yu Hou
- Department of Gynecology, Harbin Traditional Chinese Medicine Hospital, Harbin, China
| | - Xiaoling Li
- Division of CT and MRI, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Danna Cao
- Division of CT and MRI, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jinhuan Yue
- Shenzhen Frontiers in Chinese Medicine Research Co., Ltd., Shenzhen, China
- Department of Acupuncture and Moxibustion, Vitality University, Hayward, CA, United States
| |
Collapse
|
25
|
Canepa E, Parodi-Rullan R, Vazquez-Torres R, Gamallo-Lana B, Guzman-Hernandez R, Lemon NL, Angiulli F, Debure L, Ilies MA, Østergaard L, Wisniewski T, Gutiérrez-Jiménez E, Mar AC, Fossati S. FDA-approved carbonic anhydrase inhibitors reduce amyloid β pathology and improve cognition, by ameliorating cerebrovascular health and glial fitness. Alzheimers Dement 2023; 19:5048-5073. [PMID: 37186121 PMCID: PMC10600328 DOI: 10.1002/alz.13063] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 05/17/2023]
Abstract
INTRODUCTION Cerebrovascular pathology is an early and causal hallmark of Alzheimer's disease (AD), in need of effective therapies. METHODS Based on the success of our previous in vitro studies, we tested for the first time in a model of AD and cerebral amyloid angiopathy (CAA), the carbonic anhydrase inhibitors (CAIs) methazolamide and acetazolamide, Food and Drug Administration-approved against glaucoma and high-altitude sickness. RESULTS Both CAIs reduced cerebral, vascular, and glial amyloid beta (Aβ) accumulation and caspase activation, diminished gliosis, and ameliorated cognition in TgSwDI mice. The CAIs also improved microvascular fitness and induced protective glial pro-clearance pathways, resulting in the reduction of Aβ deposition. Notably, we unveiled that the mitochondrial carbonic anhydrase-VB (CA-VB) is upregulated in TgSwDI brains, CAA and AD+CAA human subjects, and in endothelial cells upon Aβ treatment. Strikingly, CA-VB silencing specifically reduces Aβ-mediated endothelial apoptosis. DISCUSSION This work substantiates the potential application of CAIs in clinical trials for AD and CAA.
Collapse
Affiliation(s)
- Elisa Canepa
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Rebecca Parodi-Rullan
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Rafael Vazquez-Torres
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Begona Gamallo-Lana
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Roberto Guzman-Hernandez
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Nicole L. Lemon
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Federica Angiulli
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Ludovic Debure
- Department on Neurology, Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Marc A. Ilies
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Temple University, Philadelphia, PA, 19140, USA
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience (CFIN), Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Thomas Wisniewski
- Department on Neurology, Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Eugenio Gutiérrez-Jiménez
- Center of Functionally Integrative Neuroscience (CFIN), Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Adam C. Mar
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Silvia Fossati
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| |
Collapse
|
26
|
Larsen RA, Constantopoulos E, Kodishala C, Lovering E, Kumar R, Hulshizer CA, Lennon RJ, Crowson CS, Nguyen AT, Myasoedova E. Neuropathologic evaluation of cerebrovascular disease in patients with rheumatoid arthritis. Rheumatology (Oxford) 2023; 62:SI296-SI303. [PMID: 37871918 PMCID: PMC10593511 DOI: 10.1093/rheumatology/kead396] [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: 03/15/2023] [Accepted: 07/19/2023] [Indexed: 10/25/2023] Open
Abstract
OBJECTIVES Active RA has been associated with an increased risk of both cardiovascular and peripheral vascular disease. We aimed to compare cerebrovascular changes in patients with and without RA, both with and without a neuropathologic diagnosis of neurodegenerative disease. METHODS Patients with RA (n = 32) who died and underwent autopsy between 1994 and 2021 were matched to non-RA controls (n = 32) on age, sex and level of neurodegenerative proteinopathy. Routine neuropathologic examination was performed at the time of autopsy. Cerebrovascular disease severity was evaluated using modified Kalaria and Strozyk scales. Clinical dementia diagnoses were manually collected from patients' medical records. RESULTS Prior to death, 15 (47%) RA patients and 14 (44%) controls were diagnosed with dementia; 9 patients in each group (60% and 64%, respectively) had Alzheimer's disease. The prevalence of cerebral amyloid angiopathy, microinfarcts, infarcts or strokes was found to be similar between groups. Patients with RA were more likely to have more severe vascular changes in the basal ganglia by Kalaria scale (P = 0.04), but not in other brain areas. There were no significant differences in the presence of large infarcts, lacunar infarcts or leukoencephalopathy by Strozyk scale. Among patients with RA and no clinical diagnosis of dementia, the majority had mild-moderate cerebrovascular abnormalities, and a subset of patients had Alzheimer's disease neuropathologic changes. CONCLUSION In this small series of autopsies, patients with and without RA had largely similar cerebrovascular pathology when controlling for neurodegenerative proteinopathies, although patients with RA exhibited more pronounced cerebrovascular disease in the basal ganglia.
Collapse
Affiliation(s)
- Rachel A Larsen
- Department of Laboratory Medicine and Pathology, Neuropathology Mayo Clinic, Rochester, MN, USA
| | - Eleni Constantopoulos
- Department of Laboratory Medicine and Pathology, Neuropathology Mayo Clinic, Rochester, MN, USA
| | - Chanakya Kodishala
- Division of Rheumatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Edward Lovering
- Division of Rheumatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Rakesh Kumar
- Division of Rheumatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Ryan J Lennon
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Cynthia S Crowson
- Division of Rheumatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Aivi T Nguyen
- Department of Laboratory Medicine and Pathology, Neuropathology Mayo Clinic, Rochester, MN, USA
| | - Elena Myasoedova
- Division of Rheumatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
27
|
Wang Y, Liu Z. Research progress on the correlation between MRI and impairment caused by cerebral small vessel disease: A review. Medicine (Baltimore) 2023; 102:e35389. [PMID: 37800770 PMCID: PMC10553107 DOI: 10.1097/md.0000000000035389] [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: 04/13/2023] [Accepted: 09/05/2023] [Indexed: 10/07/2023] Open
Abstract
Cerebral small vessel disease (CSVD) is a chronic global brain disease mainly involving small blood vessels in the brain. The disease can be gradually aggravated with the increase of age, so it is the primary cause of brain dysfunction in the elderly. With the increasing aging of the world population and the high incidence of cerebrovascular risk factors, the incidence of CSVD is increasing day by day. CSVD is characterized by insidious onset, slow progression, diverse clinical manifestations, and difficult early diagnosis. CSVD can lead to cognitive impairment, gait impairment, affective impairment, and so on. however, it has not received enough attention from researchers in the past. In recent years, some studies have shown that CSVD patients have a high proportion of related impairment, which seriously affect patients daily life and social functions. Currently, no clear preventive measures or treatments exist to improve the condition. With the development of magnetic resonance imaging, CSVD has become more and more recognized and the detection rate has gradually improved. This paper reviews the research progress of magnetic resonance imaging and cognitive impairment, gait impairment, affective impairment, urination disorder, swallowing disorder, and other disorders to provide a useful reference for the early diagnosis and treatment of CSVD and expand new ideas.
Collapse
Affiliation(s)
- Yang Wang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- Department of Neurology, 980th Hospital of PLA Joint Logistical Support Force (Bethune International Peace Hospital), Shijiazhuang, China
| | - Zhirong Liu
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
28
|
Doumatey AP, Bentley AR, Akinyemi R, Olanrewaju TO, Adeyemo A, Rotimi C. Genes, environment, and African ancestry in cardiometabolic disorders. Trends Endocrinol Metab 2023; 34:601-621. [PMID: 37598069 PMCID: PMC10548552 DOI: 10.1016/j.tem.2023.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/21/2023]
Abstract
The past two decades have been characterized by a substantial global increase in cardiometabolic diseases, but the prevalence and incidence of these diseases and related traits differ across populations. African ancestry populations are among the most affected yet least included in research. Populations of African descent manifest significant genetic and environmental diversity and this under-representation is a missed opportunity for discovery and could exacerbate existing health disparities and curtail equitable implementation of precision medicine. Here, we discuss cardiometabolic diseases and traits in the context of African descent populations, including both genetic and environmental contributors and emphasizing novel discoveries. We also review new initiatives to include more individuals of African descent in genomics to address current gaps in the field.
Collapse
Affiliation(s)
- Ayo P Doumatey
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rufus Akinyemi
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training and Centre for Genomic and Precision Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria; Department of Neurology, University College Hospital, Ibadan, Nigeria
| | - Timothy O Olanrewaju
- Division of Nephrology, Department of Medicine, University of Ilorin & University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Adebowale Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Charles Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
29
|
Wang L, Chaudhari K, Winters A, Sun Y, Berry R, Tang C, Yang SH, Liu R. Recurrent Transient Ischemic Attack Induces Neural Cytoskeleton Modification and Gliosis in an Experimental Model. Transl Stroke Res 2023; 14:740-751. [PMID: 35867329 DOI: 10.1007/s12975-022-01068-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 01/28/2023]
Abstract
Transient ischemic attack (TIA) presents a high risk for subsequent stroke, Alzheimer's disease (AD), and related dementia (ADRD). However, the neuropathophysiology of TIA has been rarely studied. By evaluating recurrent TIA-induced neuropathological changes, our study aimed to explore the potential mechanisms underlying the contribution of TIA to ADRD. In the current study, we established a recurrent TIA model by three times 10-min middle cerebral artery occlusion within a week in rat. Neither permanent neurological deficit nor apoptosis was observed following recurrent TIA. No increase of AD-related biomarkers was indicated after TIA, including increase of tau hyperphosphorylation and β-site APP cleaving enzyme 1 (BACE1). Neuronal cytoskeleton modification and neuroinflammation was found at 1, 3, and 7 days after recurrent TIA, evidenced by the reduction of microtubule-associated protein 2 (MAP2), elevation of neurofilament-light chain (NFL), and increase of glial fibrillary acidic protein (GFAP)-positive astrocytes and ionized calcium binding adaptor molecule 1 (Iba1)-positive microglia at the TIA-affected cerebral cortex and basal ganglion. Similar NFL, GFAP and Iba1 alteration was found in the white matter of corpus callosum. In summary, the current study demonstrated that recurrent TIA may trigger neuronal cytoskeleton change, astrogliosis, and microgliosis without induction of cell death at the acute and subacute stage. Our study indicates that TIA-induced neuronal cytoskeleton modification and neuroinflammation may be involved in the vascular contribution to cognitive impairment and dementia.
Collapse
Affiliation(s)
- Linshu Wang
- Departments of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107-2699, USA
| | - Kiran Chaudhari
- Departments of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107-2699, USA
| | - Ali Winters
- Departments of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107-2699, USA
| | - Yuanhong Sun
- Departments of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107-2699, USA
| | - Raymond Berry
- Departments of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107-2699, USA
| | - Christina Tang
- Departments of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107-2699, USA
| | - Shao-Hua Yang
- Departments of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107-2699, USA.
| | - Ran Liu
- Departments of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107-2699, USA.
| |
Collapse
|
30
|
DiLucia SG, Kendrick BJ, Sims-Robinson C. Hyperinsulinemia Impairs Clathrin-Mediated Endocytosis of the Insulin Receptor and Activation of Endothelial Nitric Oxide Synthase in Brain Endothelial Cells. Int J Mol Sci 2023; 24:14670. [PMID: 37834116 PMCID: PMC10572607 DOI: 10.3390/ijms241914670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/15/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Adequate perfusion of cerebral tissues, which is necessary for the preservation of optimal brain health, depends on insulin signaling within brain endothelial cells. Proper insulin signaling relies on the regulated internalization of insulin bound to the insulin receptor, a process which is disrupted by hyperinsulinemia via an unknown mechanism. Thus, the goal of this study was to characterize the impact of hyperinsulinemia on the regulation of molecular targets involved in cerebral blood flow and insulin receptor internalization into brain endothelial cells. The phosphorylation of molecular targets associated with cerebral blood flow and insulin receptor internalization was assessed in hyperinsulinemic brain endothelial cells. Insulin receptor uptake into cells was also examined in the setting of endocytosis blockade. Our data demonstrate that hyperinsulinemia impairs the activation of endothelial nitric oxide synthase. These data correspond with an impairment in clathrin-mediated endocytosis of the insulin receptor and dysregulated phosphorylation of key internalization effectors. We conclude that hyperinsulinemia alters the phosphorylation of molecular targets involved in clathrin-mediated endocytosis, disrupts signaling through the insulin receptor, and hinders the capacity for blood flow regulation by brain endothelial cells.
Collapse
Affiliation(s)
- Stephanie G. DiLucia
- Department of Neurology, Medical University of South Carolina, Charleston, SC 29425, USA;
- Ralph H. Johnson VA Medical Center, Charleston, SC 29401, USA
| | - B. Jacob Kendrick
- Flow Cytometry and Cell Sorting Shared Resource, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Catrina Sims-Robinson
- Department of Neurology, Medical University of South Carolina, Charleston, SC 29425, USA;
- Ralph H. Johnson VA Medical Center, Charleston, SC 29401, USA
| |
Collapse
|
31
|
Palmer JM, Huentelman M, Ryan L. More than just risk for Alzheimer's disease: APOE ε4's impact on the aging brain. Trends Neurosci 2023; 46:750-763. [PMID: 37460334 DOI: 10.1016/j.tins.2023.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/16/2023] [Accepted: 06/12/2023] [Indexed: 08/18/2023]
Abstract
The apolipoprotein ε4 (APOE ε4) allele is most commonly associated with increased risk for late-onset Alzheimer's disease (AD). However, recent longitudinal studies suggest that these risks are overestimated; most ε4 carriers will not develop dementia in their lifetime. In this article, we review new evidence regarding the impact of APOE ε4 on cognition among healthy older adults. We discuss emerging work from animal models suggesting that ε4 impacts brain structure and function in multiple ways that may lead to age-related cognitive impairment, independent from AD pathology. We discuss the importance of taking an individualized approach in future studies by incorporating biomarkers and neuroimaging methods that may better disentangle the phenotypic influences of APOE ε4 on the aging brain from prodromal AD pathology.
Collapse
Affiliation(s)
- Justin M Palmer
- The University of Arizona, Tucson, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA.
| | - Matthew Huentelman
- Translational Genomics Research Institute, Phoenix, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Lee Ryan
- The University of Arizona, Tucson, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA.
| |
Collapse
|
32
|
Thakore P, Yamasaki E, Ali S, Sanchez Solano A, Labelle-Dumais C, Gao X, Chaumeil MM, Gould DB, Earley S. PI3K block restores age-dependent neurovascular coupling defects associated with cerebral small vessel disease. Proc Natl Acad Sci U S A 2023; 120:e2306479120. [PMID: 37607233 PMCID: PMC10467353 DOI: 10.1073/pnas.2306479120] [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: 04/20/2023] [Accepted: 07/17/2023] [Indexed: 08/24/2023] Open
Abstract
Neurovascular coupling (NVC), a vital physiological process that rapidly and precisely directs localized blood flow to the most active regions of the brain, is accomplished in part by the vast network of cerebral capillaries acting as a sensory web capable of detecting increases in neuronal activity and orchestrating the dilation of upstream parenchymal arterioles. Here, we report a Col4a1 mutant mouse model of cerebral small vessel disease (cSVD) with age-dependent defects in capillary-to-arteriole dilation, functional hyperemia in the brain, and memory. The fundamental defect in aged mutant animals was the depletion of the minor membrane phospholipid phosphatidylinositol 4,5 bisphosphate (PIP2) in brain capillary endothelial cells, leading to the loss of inwardly rectifying K+ (Kir2.1) channel activity. Blocking phosphatidylinositol-3-kinase (PI3K), an enzyme that diminishes the bioavailability of PIP2 by converting it to phosphatidylinositol (3, 4, 5)-trisphosphate (PIP3), restored Kir2.1 channel activity, capillary-to-arteriole dilation, and functional hyperemia. In longitudinal studies, chronic PI3K inhibition also improved the memory function of aged Col4a1 mutant mice. Our data suggest that PI3K inhibition is a viable therapeutic strategy for treating defective NVC and cognitive impairment associated with cSVD.
Collapse
Affiliation(s)
- Pratish Thakore
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System University of Nevada, Reno School of Medicine, Reno, NV89557-0318
| | - Evan Yamasaki
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System University of Nevada, Reno School of Medicine, Reno, NV89557-0318
| | - Sher Ali
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System University of Nevada, Reno School of Medicine, Reno, NV89557-0318
| | - Alfredo Sanchez Solano
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System University of Nevada, Reno School of Medicine, Reno, NV89557-0318
| | - Cassandre Labelle-Dumais
- Department of Ophthalmology and Anatomy, Institute for Human Genetics, University of California San Francisco School of Medicine, San Francisco, CA94143
| | - Xiao Gao
- Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA94158
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA94143-0628
| | - Myriam M. Chaumeil
- Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA94158
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA94143-0628
| | - Douglas B. Gould
- Department of Ophthalmology and Anatomy, Institute for Human Genetics, University of California San Francisco School of Medicine, San Francisco, CA94143
| | - Scott Earley
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System University of Nevada, Reno School of Medicine, Reno, NV89557-0318
| |
Collapse
|
33
|
Hinman JD, Elahi F, Chong D, Radabaugh H, Ferguson A, Maillard P, Thompson JF, Rosenberg GA, Sagare A, Moghekar A, Lu H, Lee T, Wilcock D, Satizabal CL, Tracy R, Seshadri S, Schwab K, Helmer K, Singh H, Kivisäkk P, Greenberg S, DeCarli C, Kramer J. Placental growth factor as a sensitive biomarker for vascular cognitive impairment. Alzheimers Dement 2023; 19:3519-3527. [PMID: 36815663 PMCID: PMC10440207 DOI: 10.1002/alz.12974] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/14/2022] [Accepted: 12/19/2022] [Indexed: 02/24/2023]
Abstract
INTRODUCTION High-performing biomarkers measuring the vascular contributions to cognitive impairment and dementia are lacking. METHODS Using a multi-site observational cohort study design, we examined the diagnostic accuracy of plasma placental growth factor (PlGF) within the MarkVCID Consortium (n = 335; CDR 0-1). Subjects underwent clinical evaluation, cognitive testing, MRI, and blood sampling as defined by Consortium protocols. RESULTS In the prospective population of 335 subjects (72.2 ± 7.8 years of age, 49.3% female), plasma PlGF (pg/mL) shows an ordinal odds ratio (OR) of 1.16 (1.07-1.25; P = .0003) for increasing Fazekas score and ordinal OR of 1.22 (1.14-1.32; P < .0001) for functional cognitive impairment measured by the Clinical Dementia Rating scale. We achieved the primary study outcome of a site-independent association of plasma PlGF (pg/mL) with white matter injury and cognitive impairment in two of three study cohorts. Secondary outcomes using the full MarkVCID cohort demonstrated that plasma PlGF can significantly discriminate individuals with Fazekas ≥ 2 and CDR = 0.5 (area under the curve [AUC] = 0.74) and CDR = 1 (AUC = 0.89) from individuals with CDR = 0. DISCUSSION Plasma PlGF measured by standardized immunoassay functions as a stable, reliable, diagnostic biomarker for cognitive impairment associated with substantial white matter burden.
Collapse
Affiliation(s)
- Jason D. Hinman
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles
- Department of Neurology, West Los Angeles Veterans Association Medical Center, Department of Veterans Affairs
| | - Fanny Elahi
- Memory and Aging Center, Weill Institute for Neuroscience, University of California San Francisco
- Department of Neurology, San Francisco Veterans Association Medical Center, Department of Veterans Affairs
| | - Davis Chong
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles
| | - Hannah Radabaugh
- Department of Neurological Surgery, Weill Institute for Neuroscience, University of California San Francisco
| | - Adam Ferguson
- Department of Neurology, San Francisco Veterans Association Medical Center, Department of Veterans Affairs
- Department of Neurological Surgery, Weill Institute for Neuroscience, University of California San Francisco
| | | | | | | | - Abhay Sagare
- Zilkha Neurogenetic Institute, University of Southern California
| | | | - Hanzhang Lu
- Department of Radiology, Johns Hopkins University
| | - Tiffany Lee
- Sanders-Brown Center on Aging, Department of Physiology, University of Kentucky
| | - Donna Wilcock
- Sanders-Brown Center on Aging, Department of Physiology, University of Kentucky
| | - Claudia L. Satizabal
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, UT Health San Antonio
| | - Russell Tracy
- Department of Pathology & Laboratory Medicine, Larner College of Medicine, University of Vermont
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, UT Health San Antonio
| | - Kristin Schwab
- Department of Neurology, Massachusetts General Hospital, Harvard University
| | - Karl Helmer
- Department of Neurology, Massachusetts General Hospital, Harvard University
| | - Herpreet Singh
- Department of Neurology, Massachusetts General Hospital, Harvard University
| | - Pia Kivisäkk
- Department of Neurology, Massachusetts General Hospital, Harvard University
| | - Steve Greenberg
- Department of Neurology, Massachusetts General Hospital, Harvard University
| | | | - Joel Kramer
- Memory and Aging Center, Weill Institute for Neuroscience, University of California San Francisco
| |
Collapse
|
34
|
Torres-Espin A, Rabadaugh H, Fitzsimons S, Harvey D, Chou A, Lindberg C, Casaletto KB, Goldberger L, Staffaroni AM, Maillard P, Miller BL, DeCarli C, Hinman JD, Ferguson AR, Kramer JH, Elahi FM. Sexually dimorphic differences in angiogenesis markers predict brain aging trajectories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.16.549192. [PMID: 37503183 PMCID: PMC10370093 DOI: 10.1101/2023.07.16.549192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Aberrant angiogenesis could contribute to cognitive impairment, representing a therapeutic target for preventing dementia. However, most angiogenesis studies focus on model organisms. To test the relevance of angiogenesis to human cognitive aging, we evaluated associations of circulating blood markers of angiogenesis with brain aging trajectories in two deeply phenotyped human cohorts (n=435, age 74 + 9) with longitudinal cognitive assessments, biospecimens, structural brain imaging, and clinical data. Machine learning and traditional statistics revealed sex dimorphic associations of plasma angiogenic growth factors with brain aging outcomes. Specifically, angiogenesis is associated with higher executive function and less brain atrophy in younger women (not men), a directionality of association that reverses around age 75. Higher levels of basic fibroblast growth factor, known for pleiotropic effects on multiple cell types, predicted favorable cognitive trajectories. This work demonstrates the relevance of angiogenesis to brain aging with important therapeutic implications for vascular cognitive impairment and dementia.
Collapse
|
35
|
Hainsworth AH, Arancio O, Elahi FM, Isaacs JD, Cheng F. PDE5 inhibitor drugs for use in dementia. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2023; 9:e12412. [PMID: 37766832 PMCID: PMC10520293 DOI: 10.1002/trc2.12412] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 09/29/2023]
Abstract
Alzheimer's disease and related dementias (ADRD) remain a major health-care challenge with few licensed medications. Repurposing existing drugs may afford prevention and treatment. Phosphodiesterase-5 (PDE5) is widely expressed in vascular myocytes, neurons, and glia. Potent, selective, Food and Drug Administration-approved PDE5 inhibitors are already in clinical use (sildenafil, vardenafil, tadalafil) as vasodilators in erectile dysfunction and pulmonary arterial hypertension. Animal data indicate cognitive benefits of PDE5 inhibitors. In humans, real-world patient data suggest that sildenafil and vardenafil are associated with reduced dementia risk. While a recent clinical trial of acute tadalafil on cerebral blood flow was neutral, there may be chronic actions of PDE5 inhibition on cerebrovascular or synaptic function. We provide a perspective on the potential utility of PDE5 inhibitors for ADRD. We conclude that further prospective clinical trials with PDE5 inhibitors are warranted. The choice of drug will depend on brain penetration, tolerability in older people, half-life, and off-target effects. HIGHLIGHTS Potent phosphodiesterase-5 (PDE5) inhibitors are in clinical use as vasodilators.In animals PDE5 inhibitors enhance synaptic function and cognitive ability.In humans the PDE5 inhibitor sildenafil is associated with reduced risk of Alzheimer's disease.Licensed PDE5 inhibitors have potential for repurposing in dementia.Prospective clinical trials of PDE5 inhibitors are warranted.
Collapse
Affiliation(s)
- Atticus H. Hainsworth
- Molecular & Clinical Sciences Research InstituteSt George's University of LondonLondonUK
- Department of NeurologySt George's University Hospitals NHS Foundation TrustLondonUK
| | - Ottavio Arancio
- Department of Pathology and Cell BiologyTaub Institute for Research on Alzheimer's Disease and the Aging BrainDepartment of MedicineColumbia UniversityNew YorkNew YorkUSA
| | - Fanny M. Elahi
- Departments of Neurology and NeuroscienceRonald M. Loeb Center for Alzheimer's DiseaseFriedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jeremy D. Isaacs
- Molecular & Clinical Sciences Research InstituteSt George's University of LondonLondonUK
- Department of NeurologySt George's University Hospitals NHS Foundation TrustLondonUK
| | - Feixiong Cheng
- Genomic Medicine InstituteLerner Research InstituteCleveland ClinicClevelandOhioUSA
- Department of Molecular MedicineCleveland Clinic Lerner College of MedicineCase Western Reserve UniversityClevelandOhioUSA
| |
Collapse
|
36
|
Sulimai N, Brown J, Lominadze D. Vascular Effects on Cerebrovascular Permeability and Neurodegeneration. Biomolecules 2023; 13:biom13040648. [PMID: 37189395 DOI: 10.3390/biom13040648] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/29/2023] [Accepted: 04/02/2023] [Indexed: 05/17/2023] Open
Abstract
Neurons and glial cells in the brain are protected by the blood brain barrier (BBB). The local regulation of blood flow is determined by neurons and signal conducting cells called astrocytes. Although alterations in neurons and glial cells affect the function of neurons, the majority of effects are coming from other cells and organs of the body. Although it seems obvious that effects beginning in brain vasculature would play an important role in the development of various neuroinflammatory and neurodegenerative pathologies, significant interest has only been directed to the possible mechanisms involved in the development of vascular cognitive impairment and dementia (VCID) for the last decade. Presently, the National Institute of Neurological Disorders and Stroke applies considerable attention toward research related to VCID and vascular impairments during Alzheimer's disease. Thus, any changes in cerebral vessels, such as in blood flow, thrombogenesis, permeability, or others, which affect the proper vasculo-neuronal connection and interaction and result in neuronal degeneration that leads to memory decline should be considered as a subject of investigation under the VCID category. Out of several vascular effects that can trigger neurodegeneration, changes in cerebrovascular permeability seem to result in the most devastating effects. The present review emphasizes the importance of changes in the BBB and possible mechanisms primarily involving fibrinogen in the development and/or progression of neuroinflammatory and neurodegenerative diseases resulting in memory decline.
Collapse
Affiliation(s)
- Nurul Sulimai
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA
| | - Jason Brown
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA
| | - David Lominadze
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA
- Department of Molecular Pharmacology and Physiology, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA
| |
Collapse
|
37
|
Eisenmenger LB, Peret A, Famakin BM, Spahic A, Roberts GS, Bockholt JH, Johnson KM, Paulsen JS. Vascular contributions to Alzheimer's disease. Transl Res 2023; 254:41-53. [PMID: 36529160 PMCID: PMC10481451 DOI: 10.1016/j.trsl.2022.12.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and is characterized by progressive neurodegeneration and cognitive decline. Understanding the pathophysiology underlying AD is paramount for the management of individuals at risk of and suffering from AD. The vascular hypothesis stipulates a relationship between cardiovascular disease and AD-related changes although the nature of this relationship remains unknown. In this review, we discuss several potential pathological pathways of vascular involvement in AD that have been described including dysregulation of neurovascular coupling, disruption of the blood brain barrier, and reduced clearance of metabolite waste such as beta-amyloid, a toxic peptide considered the hallmark of AD. We will also discuss the two-hit hypothesis which proposes a 2-step positive feedback loop in which microvascular insults precede the accumulation of Aß and are thought to be at the origin of the disease development. At neuroimaging, signs of vascular dysfunction such as chronic cerebral hypoperfusion have been demonstrated, appearing early in AD, even before cognitive decline and alteration of traditional biomarkers. Cerebral small vessel disease such as cerebral amyloid angiopathy, characterized by the aggregation of Aß in the vessel wall, is highly prevalent in vascular dementia and AD patients. Current data is unclear whether cardiovascular disease causes, precipitates, amplifies, precedes, or simply coincides with AD. Targeted imaging tools to quantitatively evaluate the intracranial vasculature and longitudinal studies in individuals at risk for or in the early stages of the AD continuum could be critical in disentangling this complex relationship between vascular disease and AD.
Collapse
Affiliation(s)
- Laura B Eisenmenger
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Anthony Peret
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Bolanle M Famakin
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Alma Spahic
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Grant S Roberts
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jeremy H Bockholt
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, and Emory University, Atlanta, Georgia
| | - Kevin M Johnson
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jane S Paulsen
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin.
| |
Collapse
|
38
|
Thakore P, Yamasaki E, Ali S, Solano AS, Labelle-Dumais C, Gao X, Chaumeil MM, Gould DB, Earley S. PI3K block restores age-dependent neurovascular coupling defects associated with cerebral small vessel disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.03.531032. [PMID: 36945616 PMCID: PMC10028793 DOI: 10.1101/2023.03.03.531032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
Neurovascular coupling (NVC), a vital physiological process that rapidly and precisely directs localized blood flow to the most active regions of the brain, is accomplished in part by the vast network of cerebral capillaries acting as a sensory web capable of detecting increases in neuronal activity and orchestrating the dilation of upstream parenchymal arterioles. Here, we report a Col4a1 mutant mouse model of cerebral small vessel disease (cSVD) with age-dependent defects in capillary-to-arteriole dilation, functional hyperemia in the brain, and memory. The fundamental defect in aged mutant animals was the depletion of the minor membrane phospholipid phosphatidylinositol 4,5 bisphosphate (PIP 2 ) in brain capillary endothelial cells, leading to the loss of inwardly rectifier K + (Kir2.1) channel activity. Blocking phosphatidylinositol-3-kinase (PI3K), an enzyme that diminishes the bioavailability of PIP 2 by converting it to phosphatidylinositol (3,4,5)-trisphosphate (PIP 3 ), restored Kir2.1 channel activity, capillary-to-arteriole dilation, and functional hyperemia. In longitudinal studies, chronic PI3K inhibition also improved the memory function of aged Col4a1 mutant mice. Our data suggest that PI3K inhibition is a viable therapeutic strategy for treating defective NVC and cognitive impairment associated with cSVD. One-sentence summary PI3K inhibition rescues neurovascular coupling defects in cerebral small vessel disease.
Collapse
|
39
|
Tian Y, Fopiano KA, Buncha V, Lang L, Suggs HA, Wang R, Rudic RD, Filosa JA, Bagi Z. The role of ADAM17 in cerebrovascular and cognitive function in the APP/PS1 mouse model of Alzheimer's disease. Front Mol Neurosci 2023; 16:1125932. [PMID: 36937050 PMCID: PMC10018024 DOI: 10.3389/fnmol.2023.1125932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/27/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction The disintegrin and metalloproteinase 17 (ADAM17) exhibits α-secretase activity, whereby it can prevent the production of neurotoxic amyloid precursor protein-α (APP). ADAM17 is abundantly expressed in vascular endothelial cells and may act to regulate vascular homeostatic responses, including vasomotor function, vascular wall morphology, and formation of new blood vessels. The role of vascular ADAM17 in neurodegenerative diseases remains poorly understood. Here, we hypothesized that cerebrovascular ADAM17 plays a role in the pathogenesis of Alzheimer's disease (AD). Methods and results We found that 9-10 months old APP/PS1 mice with b-amyloid accumulation and short-term memory and cognitive deficits display a markedly reduced expression of ADAM17 in cerebral microvessels. Systemic delivery and adeno-associated virus (AAV)-mediated re-expression of ADAM17 in APP/PS1 mice improved cognitive functioning, without affecting b-amyloid plaque density. In isolated and pressurized cerebral arteries of APP/PS1 mice the endothelium-dependent dilation to acetylcholine was significantly reduced, whereas the vascular smooth muscle-dependent dilation to the nitric oxide donor, sodium nitroprusside was maintained when compared to WT mice. The impaired endothelium-dependent vasodilation of cerebral arteries in APP/PS1 mice was restored to normal level by ADAM17 re-expression. The cerebral artery biomechanical properties (wall stress and elasticity) and microvascular network density was not affected by ADAM17 re-expression in the APP/PS1 mice. Additionally, proteomic analysis identified several differentially expressed molecules involved in AD neurodegeneration and neuronal repair mechanisms that were reversed by ADAM17 re-expression. Discussion Thus, we propose that a reduced ADAM17 expression in cerebral microvessels impairs vasodilator function, which may contribute to the development of cognitive dysfunction in APP/PS1 mice, and that ADAM17 can potentially be targeted for therapeutic intervention in AD.
Collapse
Affiliation(s)
- Yanna Tian
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Katie Anne Fopiano
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Vadym Buncha
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Liwei Lang
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Hayden A. Suggs
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Rongrong Wang
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - R. Daniel Rudic
- Department of Pharmacology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Jessica A. Filosa
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
40
|
Andjelkovic AV, Situ M, Citalan-Madrid AF, Stamatovic SM, Xiang J, Keep RF. Blood-Brain Barrier Dysfunction in Normal Aging and Neurodegeneration: Mechanisms, Impact, and Treatments. Stroke 2023; 54:661-672. [PMID: 36848419 PMCID: PMC9993074 DOI: 10.1161/strokeaha.122.040578] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Cerebral endothelial cells and their linking tight junctions form a unique, dynamic and multi-functional interface, the blood-brain barrier (BBB). The endothelium is regulated by perivascular cells and components forming the neurovascular unit. This review examines BBB and neurovascular unit changes in normal aging and in neurodegenerative disorders, particularly focusing on Alzheimer disease, cerebral amyloid angiopathy and vascular dementia. Increasing evidence indicates BBB dysfunction contributes to neurodegeneration. Mechanisms underlying BBB dysfunction are outlined (endothelium and neurovascular unit mediated) as is the BBB as a therapeutic target including increasing the uptake of systemically delivered therapeutics across the BBB, enhancing clearance of potential neurotoxic compounds via the BBB, and preventing BBB dysfunction. Finally, a need for novel biomarkers of BBB dysfunction is addressed.
Collapse
Affiliation(s)
- Anuska V. Andjelkovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor MI, USA
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor MI, USA
| | - Muyu Situ
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor MI, USA
| | | | | | - Jianming Xiang
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor MI, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor MI, USA
| |
Collapse
|
41
|
Branyan K, Labelle-Dumais C, Wang X, Hayashi G, Lee B, Peltz Z, Gorman S, Li BQ, Mao M, Gould DB. Elevated TGFβ signaling contributes to cerebral small vessel disease in mouse models of Gould syndrome. Matrix Biol 2023; 115:48-70. [PMID: 36435425 PMCID: PMC10393528 DOI: 10.1016/j.matbio.2022.11.007] [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: 06/26/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
Cerebral small vessel disease (CSVD) is a leading cause of stroke and vascular cognitive impairment and dementia. Studying monogenic CSVD can reveal pathways that are dysregulated in common sporadic forms of the disease and may represent therapeutic targets. Mutations in collagen type IV alpha 1 (COL4A1) and alpha 2 (COL4A2) cause highly penetrant CSVD as part of a multisystem disorder referred to as Gould syndrome. COL4A1 and COL4A2 form heterotrimers [a1α1α2(IV)] that are fundamental constituents of basement membranes. However, their functions are poorly understood and the mechanism(s) by which COL4A1 and COL4A2 mutations cause CSVD are unknown. We used histological, molecular, genetic, pharmacological, and in vivo imaging approaches to characterize central nervous system (CNS) vascular pathologies in Col4a1 mutant mouse models of monogenic CSVD to provide insight into underlying pathogenic mechanisms. We describe developmental CNS angiogenesis abnormalities characterized by impaired retinal vascular outgrowth and patterning, increased numbers of mural cells with abnormal morphologies, altered contractile protein expression in vascular smooth muscle cells (VSMCs) and age-related loss of arteriolar VSMCs in Col4a1 mutant mice. Importantly, we identified elevated TGFβ signaling as a pathogenic consequence of Col4a1 mutations and show that genetically suppressing TGFβ signaling ameliorated CNS vascular pathologies, including partial rescue of retinal vascular patterning defects, prevention of VSMC loss, and significant reduction of intracerebral hemorrhages in Col4a1 mutant mice aged up to 8 months. This study identifies a novel biological role for collagen α1α1α2(IV) as a regulator of TGFβ signaling and demonstrates that elevated TGFβ signaling contributes to CNS vascular pathologies caused by Col4a1 mutations. Our findings suggest that pharmacologically suppressing TGFβ signaling could reduce the severity of CSVD, and potentially other manifestations associated with Gould syndrome and have important translational implications that could extend to idiopathic forms of CSVD.
Collapse
Affiliation(s)
- Kayla Branyan
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Cassandre Labelle-Dumais
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Xiaowei Wang
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Genki Hayashi
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Bryson Lee
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Zoe Peltz
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Seán Gorman
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Bo Qiao Li
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Mao Mao
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Douglas B Gould
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States; Department of Anatomy, Cardiovascular Research Institute, Bakar Aging Research Institute, and Institute for Human Genetics, University of California, San Francisco, United States.
| |
Collapse
|
42
|
Cerebral Superficial Siderosis. Clin Neuroradiol 2022; 33:293-306. [DOI: 10.1007/s00062-022-01231-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 10/11/2022] [Indexed: 11/29/2022]
Abstract
AbstractSuperficial siderosis (SS) of the central nervous system constitutes linear hemosiderin deposits in the leptomeninges and the superficial layers of the cerebrum and the spinal cord. Infratentorial (i) SS is likely due to recurrent or continuous slight bleeding into the subarachnoid space. It is assumed that spinal dural pathologies often resulting in cerebrospinal fluid (CSF) leakage is the most important etiological group which causes iSS and detailed neuroradiological assessment of the spinal compartment is necessary. Further etiologies are neurosurgical interventions, trauma and arteriovenous malformations. Typical neurological manifestations of this classical type of iSS are slowly progressive sensorineural hearing impairment and cerebellar symptoms, such as ataxia, kinetic tremor, nystagmus and dysarthria. Beside iSS, a different type of SS restricted to the supratentorial compartment can be differentiated, i.e. cortical (c) SS, especially in older people often due to cerebral amyloid angiopathy (CAA). Clinical presentation of cSS includes transient focal neurological episodes or “amyloid spells”. In addition, spontaneous and amyloid beta immunotherapy-associated CAA-related inflammation may cause cSS, which is included in the hemorrhagic subgroup of amyloid-related imaging abnormalities (ARIA). Because a definitive diagnosis requires a brain biopsy, knowledge of neuroimaging features and clinical findings in CAA-related inflammation is essential. This review provides neuroradiological hallmarks of the two groups of SS and give an overview of neurological symptoms and differential diagnostic considerations.
Collapse
|
43
|
Mukli P, Detwiler S, Owens CD, Csipo T, Lipecz A, Pinto CB, Tarantini S, Nyul-Toth A, Balasubramanian P, Hoffmeister JR, Csiszar A, Ungvari Z, Kirkpatrick AC, Prodan CI, Yabluchanskiy A. Gait variability predicts cognitive impairment in older adults with subclinical cerebral small vessel disease. Front Aging Neurosci 2022; 14:1052451. [PMID: 36466602 PMCID: PMC9716182 DOI: 10.3389/fnagi.2022.1052451] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 11/02/2022] [Indexed: 11/19/2022] Open
Abstract
Introduction Advanced methods of gait research, including approaches to quantify variability, and orderliness/regularity/predictability, are increasingly used to identify patients at risk for the development of cognitive impairment. Cerebral small vessel disease (CSVD) is highly prevalent in older adults and is known to contribute to the development of vascular cognitive impairment and dementia (VCID). Studies in preclinical models demonstrate that subclinical alterations precede CSVD-related cognitive impairment in gait coordination. In humans, CSVD also associates with gait abnormalities. The present study was designed to test the hypothesis that increased gait variability and gait asymmetry predict a decline in cognitive performance in older adults with CSVD. Methods To test this hypothesis, we compared cognitive performance and gait function in patients with CSVD (age: 69.8 ± 5.3 years; n = 11) and age- and sex-matched control participants (age: 70.7 ± 5.8 years; n = 11). Based on imaging findings, patients with CSVD were identified [presence of white matter hyperintensities plus silent brain infarcts and/or microhemorrhages on magnetic resonance imaging (MRI) assessment]. Cognitive performance was assessed using the Cambridge Neuropsychological Test Automated Battery (CANTAB). Gait parameters were measured during the single and dual tasks, during which participants, in addition to the motor task, completed a series of mental arithmetic calculations. Spatial and temporal parameters of gait variability, symmetry, and permutation entropy were determined using a pressure-sensitive gait mat during single and dual cognitive task conditions. Results Patients with CSVD exhibited lower performance in a visual learning test (p = 0.030) and in a sustained attention test (p = 0.007). CSVD also affected step time variability (p = 0.009) and step length variability (p = 0.017). Step lengths of CSVD participants were more asymmetric (p = 0.043) than that of controls, while the two groups were statistically similar regarding step time symmetry and entropy of step time and length. Gait variability was inversely associated with sustained attention, especially among CSVD patients, and this relationship was significantly different between the two groups. The association of sustained attention with gait symmetry was also significantly different between the two groups. Discussion Our findings provide additional evidence in support of the concept that increased gait variability and asymmetry may predict cognitive impairment in older adults with CSVD.
Collapse
Affiliation(s)
- Peter Mukli
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Sam Detwiler
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Cameron D. Owens
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Tamas Csipo
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Agnes Lipecz
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Camila Bonin Pinto
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Stefano Tarantini
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary,Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Adam Nyul-Toth
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Priya Balasubramanian
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jordan R. Hoffmeister
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Anna Csiszar
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary,Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Zoltan Ungvari
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary,Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Angelia C. Kirkpatrick
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Veterans Affairs Medical Center, Oklahoma City, OK, United States
| | - Calin I. Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK, United States,Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Andriy Yabluchanskiy
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States,*Correspondence: Andriy Yabluchanskiy,
| |
Collapse
|
44
|
Chandran R, He L, Nie X, Voltin J, Jamil S, Doueiry C, Falangola MF, Ergul A, Li W. Magnetic resonance imaging reveals microemboli-mediated pathological changes in brain microstructure in diabetic rats: relevance to vascular cognitive impairment/dementia. Clin Sci (Lond) 2022; 136:1555-1570. [PMID: 36314470 PMCID: PMC10066787 DOI: 10.1042/cs20220465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/19/2022] [Accepted: 10/28/2022] [Indexed: 11/17/2022]
Abstract
Diabetes doubles the risk of vascular cognitive impairment, but the underlying reasons remain unclear. In the present study, we determined the temporal and spatial changes in the brain structure after microemboli (ME) injection using diffusion MRI (dMRI). Control and diabetic rats received cholesterol crystal ME (40-70 µm) injections. Cognitive tests were followed up to 16 weeks, while dMRI scans were performed at baseline and 12 weeks post-ME. The novel object recognition test had a lower d2 recognition index along with a decrease in spontaneous alternations in the Y maze test in diabetic rats with ME. dMRI showed that ME injection caused infarction in two diabetic animals (n=5) but none in controls (n=6). In diabetes, radial diffusivity (DR) was increased while fractional anisotropy (FA) was decreased in the cortex, indicating loss of tissue integrity and edema. In the dorsal hippocampus, mean diffusivity (MD), axial diffusivity (DA), and DR were significantly increased, indicating loss of axons and myelin damage. Histological analyses confirmed more tissue damage and microglial activation in diabetic rats with ME. These results suggest that ME injury and associated cerebrovascular dysfunction are greater in diabetes, which may cause cognitive deficits. Strategies to improve vascular function can be a preventive and therapeutic approach for vascular cognitive impairment.
Collapse
Affiliation(s)
- Raghavendar Chandran
- Ralph H. Johnson VA Medical Center, Charleston, SC
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| | - Lianying He
- Ralph H. Johnson VA Medical Center, Charleston, SC
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| | - Xingju Nie
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC
| | - Joshua Voltin
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC
| | - Sarah Jamil
- Ralph H. Johnson VA Medical Center, Charleston, SC
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| | - Caren Doueiry
- Ralph H. Johnson VA Medical Center, Charleston, SC
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| | - Maria Fatima Falangola
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC
| | - Adviye Ergul
- Ralph H. Johnson VA Medical Center, Charleston, SC
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| | - Weiguo Li
- Ralph H. Johnson VA Medical Center, Charleston, SC
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
45
|
Rudnicka-Drożak E, Drożak P, Mizerski G, Drożak M. Endothelial Progenitor Cells in Neurovascular Disorders—A Comprehensive Overview of the Current State of Knowledge. Biomedicines 2022; 10:biomedicines10102616. [PMID: 36289878 PMCID: PMC9599182 DOI: 10.3390/biomedicines10102616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/11/2022] [Accepted: 10/15/2022] [Indexed: 11/16/2022] Open
Abstract
Endothelial progenitor cells (EPCs) are a population of cells that circulate in the blood looking for areas of endothelial or vascular injury in order to repair them. Endothelial dysfunction is an important component of disorders with neurovascular involvement. Thus, the subject of involvement of EPCs in such conditions has been gaining increasing scientific interest in recent years. Overall, decreased levels of EPCs are associated with worse disease outcome. Moreover, their functionalities appear to decline with severity of disease. These findings inspired the application of EPCs as therapeutic targets and agents. So far, EPCs appear safe and promising based on the results of pre-clinical studies conducted on their use in the treatment of Alzheimer’s disease and ischemic stroke. In the case of the latter, human clinical trials have recently started to be performed in this subject and provided optimistic results thus far. Whereas in the case of migraine, existing findings pave the way for testing EPCs in in vitro studies. This review aims to thoroughly summarize current knowledge on the role EPCs in four disorders with neurovascular involvement, which are Alzheimer’s disease, cerebral small vessel disease, ischemic stroke and migraine, with a particular focus on the potential practical use of these cells as a treatment remedy.
Collapse
Affiliation(s)
- Ewa Rudnicka-Drożak
- Department of Family Medicine, Medical University of Lublin, Langiewicza 6a, 20-035 Lublin, Poland
| | - Paulina Drożak
- Student Scientific Society, Department of Family Medicine, Medical University of Lublin, Langiewicza 6a, 20-035 Lublin, Poland
- Correspondence: ; Tel.: +48-669-084-455
| | - Grzegorz Mizerski
- Department of Family Medicine, Medical University of Lublin, Langiewicza 6a, 20-035 Lublin, Poland
| | - Martyna Drożak
- Student Scientific Society, Department of Family Medicine, Medical University of Lublin, Langiewicza 6a, 20-035 Lublin, Poland
| |
Collapse
|
46
|
Reynolds CF, Jeste DV, Sachdev PS, Blazer DG. Mental health care for older adults: recent advances and new directions in clinical practice and research. World Psychiatry 2022; 21:336-363. [PMID: 36073714 PMCID: PMC9453913 DOI: 10.1002/wps.20996] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The world's population is aging, bringing about an ever-greater burden of mental disorders in older adults. Given multimorbidities, the mental health care of these people and their family caregivers is labor-intensive. At the same time, ageism is a big problem for older people, with and without mental disorders. Positive elements of aging, such as resilience, wisdom and prosocial behaviors, need to be highlighted and promoted, both to combat stigma and to help protect and improve mental health in older adults. The positive psychiatry of aging is not an oxymoron, but a scientific construct strongly informed by research evidence. We champion a broader concept of geriatric psychiatry - one that encompasses health as well as illness. In the present paper, we address these issues in the context of four disorders that are the greatest source of years lived with disability: neurocognitive disorders, major depression, schizophrenia, and substance use disorders. We emphasize the need for implementation of multidisciplinary team care, with comprehensive assessment, clinical management, intensive outreach, and coordination of mental, physical and social health services. We also underscore the need for further research into moderators and mediators of treatment response variability. Because optimal care of older adults with mental disorders is both patient-focused and family-centered, we call for further research into enhancing the well-being of family caregivers. To optimize both the safety and efficacy of pharmacotherapy, further attention to metabolic, cardiovascular and neurological tolerability is much needed, together with further development and testing of medications that reduce the risk for suicide. At the same time, we also address positive aging and normal cognitive aging, both as an antidote to ageism and as a catalyst for change in the way we think about aging per se and late-life mental disorders more specifically. It is in this context that we provide directions for future clinical care and research.
Collapse
Affiliation(s)
| | - Dilip V. Jeste
- Department of PsychiatryUniversity of California San DiegoLa JollaCAUSA
| | | | - Dan G. Blazer
- Department of Psychiatry and Behavioral SciencesDuke UniversityDurhamNCUSA
| |
Collapse
|
47
|
Bhatia K, Kindelin A, Nadeem M, Khan MB, Yin J, Fuentes A, Miller K, Turner GH, Preul MC, Ahmad AS, Mufson EJ, Waters MF, Ahmad S, Ducruet AF. Complement C3a Receptor (C3aR) Mediates Vascular Dysfunction, Hippocampal Pathology, and Cognitive Impairment in a Mouse Model of VCID. Transl Stroke Res 2022; 13:816-829. [PMID: 35258803 DOI: 10.1007/s12975-022-00993-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 01/12/2023]
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) secondary to chronic mild-moderate cerebral ischemia underlie a significant percentage of cases of dementia. We previously reported that either genetic deficiency of the complement C3a receptor (C3aR) or its pharmacological inhibition protects against cerebral ischemia in rodents, while others have implicated C3aR in the pathogenesis seen in rodent transgenic models of Alzheimer's disease. In the present study, we evaluated the role of complement C3a-C3aR signaling in the onset and progression of VCID. We utilized the bilateral common carotid artery stenosis (BCAS) model to induce VCID in male C57BL/6 wild-type and C3aR-knockout (C3aR-/-) mice. Cerebral blood flow (CBF) changes, hippocampal atrophy (HA), white matter degeneration (WMD), and ventricular size were assessed at 4 months post-BCAS using laser speckle contrast analysis (LSCI) and magnetic resonance imaging (MRI). Cognitive function was evaluated using the Morris water maze (MWM), and novel object recognition (NOR), immunostaining, and western blot were performed to assess the effect of genetic C3aR deletion on post-VCID outcomes. BCAS resulted in decreased CBF and increased HA, WMD, and neurovascular inflammation in WT (C57BL/6) compared to C3aR-/- (C3aR-KO) mice. Moreover, C3aR-/- mice exhibited improved cognitive function on NOR and MWM relative to WT controls. We conclude that over-activation of the C3a/C3aR axis exacerbates neurovascular inflammation leading to poor VCID outcomes which are mitigated by C3aR deletion. Future studies are warranted to dissect the role of cell-specific C3aR in VCID.
Collapse
Affiliation(s)
- Kanchan Bhatia
- Department of Neurosurgery, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, AZ, USA
| | - Adam Kindelin
- Department of Neurosurgery, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Muhammad Nadeem
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | | | - Junxiang Yin
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
- Department of Neurology, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Alberto Fuentes
- Barrow Neurological Institute/Arizona State University Center for Preclinical Imaging, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Karis Miller
- Department of Neurosurgery, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Gregory H Turner
- Barrow Neurological Institute/Arizona State University Center for Preclinical Imaging, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Mark C Preul
- Department of Neurosurgery, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Abdullah S Ahmad
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
- Department of Neurology, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Elliott J Mufson
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Michael F Waters
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
- Department of Neurology, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Saif Ahmad
- Department of Neurosurgery, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA.
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA.
| | - Andrew F Ducruet
- Departments of Neurosurgery & Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85086, USA.
| |
Collapse
|
48
|
Shaaban CE, Tudorascu DL, Glymour MM, Cohen AD, Thurston RC, Snyder HM, Hohman TJ, Mukherjee S, Yu L, Snitz BE. A guide for researchers seeking training in retrospective data harmonization for population neuroscience studies of Alzheimer's disease and related dementias. FRONTIERS IN NEUROIMAGING 2022; 1:978350. [PMID: 37464990 PMCID: PMC10353763 DOI: 10.3389/fnimg.2022.978350] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Due to needs surrounding rigor and reproducibility, subgroup specific disease knowledge, and questions of external validity, data harmonization is an essential tool in population neuroscience of Alzheimer's disease and related dementias (ADRD). Systematic harmonization of data elements is necessary to pool information from heterogeneous samples, and such pooling allows more expansive evaluations of health disparities, more precise effect estimates, and more opportunities to discover effective prevention or treatment strategies. The key goal of this Tutorial in Population Neuroimaging Curriculum, Instruction, and Pedagogy article is to guide researchers in creating a customized population neuroscience of ADRD harmonization training plan to fit their needs or those of their mentees. We provide brief guidance for retrospective data harmonization of multiple data types in this area, including: (1) clinical and demographic, (2) neuropsychological, and (3) neuroimaging data. Core competencies and skills are reviewed, and resources are provided to fill gaps in training as well as data needs. We close with an example study in which harmonization is a critical tool. While several aspects of this tutorial focus specifically on ADRD, the concepts and resources are likely to benefit population neuroscientists working in a range of research areas.
Collapse
Affiliation(s)
- C. Elizabeth Shaaban
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Dana L. Tudorascu
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - M. Maria Glymour
- Department of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Ann D. Cohen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rebecca C. Thurston
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Heather M. Snyder
- Medical and Scientific Relations, Alzheimer’s Association, Chicago, IL, United States
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | | | - Lan Yu
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Beth E. Snitz
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
49
|
Cogswell PM, Barakos JA, Barkhof F, Benzinger TS, Jack CR, Poussaint TY, Raji CA, Ramanan VK, Whitlow CT. Amyloid-Related Imaging Abnormalities with Emerging Alzheimer Disease Therapeutics: Detection and Reporting Recommendations for Clinical Practice. AJNR Am J Neuroradiol 2022; 43:E19-E35. [PMID: 35953274 PMCID: PMC9451628 DOI: 10.3174/ajnr.a7586] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Monoclonal antibodies are emerging disease-modifying therapies for Alzheimer disease that require brain MR imaging for eligibility assessment as well as for monitoring for amyloid-related imaging abnormalities. Amyloid-related imaging abnormalities result from treatment-related loss of vascular integrity and may occur in 2 forms. Amyloid-related imaging abnormalities with edema or effusion are transient, treatment-induced edema or sulcal effusion, identified on T2-FLAIR. Amyloid-related imaging abnormalities with hemorrhage are treatment-induced microhemorrhages or superficial siderosis identified on T2* gradient recalled-echo. As monoclonal antibodies become more widely available, treatment screening and monitoring brain MR imaging examinations may greatly increase neuroradiology practice volumes. Radiologists must become familiar with the imaging appearance of amyloid-related imaging abnormalities, how to select an appropriate imaging protocol, and report findings in clinical practice. On the basis of clinical trial literature and expert experience from clinical trial imaging, we summarize imaging findings of amyloid-related imaging abnormalities, describe potential interpretation pitfalls, and provide recommendations for a standardized imaging protocol and an amyloid-related imaging abnormalities reporting template. Standardized imaging and reporting of these findings are important because an amyloid-related imaging abnormalities severity score, derived from the imaging findings, is used along with clinical status to determine patient management and eligibility for continued monoclonal antibody dosing.
Collapse
Affiliation(s)
- P M Cogswell
- From the Departments of Radiology (P.M.C., C.R.J.)
| | - J A Barakos
- Department of Radiology (J.A.B.), California Pacific Medical Center, San Francisco, California
| | - F Barkhof
- Departments of Radiology (F.B.)
- Nuclear Medicine (F.B.), VU University Medical Center, Amsterdam, the Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, UK
| | - T S Benzinger
- Departments of Radiology (T.S.B., C.A.R.)
- Neurosurgery (T.S.B.)
| | - C R Jack
- From the Departments of Radiology (P.M.C., C.R.J.)
| | - T Y Poussaint
- Department of Radiology (T.Y.P.), Boston Children's Hospital, Boston, Massachusetts
| | - C A Raji
- Departments of Radiology (T.S.B., C.A.R.)
- Neurology (C.A.R.),Washington University School of Medicine, St. Louis, Missouri
| | - V K Ramanan
- Neurology (V.K.R.), Mayo Clinic, Rochester, Minnesota
| | - C T Whitlow
- Departments of Radiology (C.T.W.)
- Biomedical Engineering (C.T.W.), Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
50
|
Macaulay TR, Hegarty A, Yan L, Duncan D, Pa J, Kutch JJ, La Rocca M, Lane CJ, Schroeder ET. Effects of a 12-Week Periodized Resistance Training Program on Resting Brain Activity and Cerebrovascular Function: A Nonrandomized Pilot Trial. Neurosci Insights 2022; 17:26331055221119441. [PMID: 35983377 PMCID: PMC9379950 DOI: 10.1177/26331055221119441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 07/27/2022] [Indexed: 01/26/2023] Open
Abstract
Resistance training is a promising strategy to promote healthy cognitive aging; however, the brain mechanisms by which resistance training benefits cognition have yet to be determined. Here, we examined the effects of a 12-week resistance training program on resting brain activity and cerebrovascular function in 20 healthy older adults (14 females, mean age 69.1 years). In this single group clinical trial, multimodal 3 T magnetic resonance imaging was performed at 3 time points: baseline (preceding a 12-week control period), pre-intervention, and post-intervention. Along with significant improvements in fluid cognition (d = 1.27), 4 significant voxelwise clusters were identified for decreases in resting brain activity after the intervention (Cerebellum, Right Middle Temporal Gyrus, Left Inferior Parietal Lobule, and Right Inferior Parietal Lobule), but none were identified for changes in resting cerebral blood flow. Using a separate region of interest approach, we provide estimates for improved cerebral blood flow, compared with declines over the initial control period, in regions associated with cognitive impairment, such as hippocampal blood flow (d = 0.40), and posterior cingulate blood flow (d = 0.61). Finally, resistance training had a small countermeasure effect on the age-related progression of white matter lesion volume (rank-biserial = -0.22), a biomarker of cerebrovascular disease. These proof-of-concept data support larger trials to determine whether resistance training can attenuate or even reverse salient neurodegenerative processes.
Collapse
Affiliation(s)
- Timothy R Macaulay
- Division of Biokinesiology and Physical
Therapy, Ostrow School of Dentistry, University of Southern California, Los Angeles,
CA, USA,Timothy R Macaulay, Division of
Biokinesiology and Physical Therapy, Ostrow School of Dentistry, University of
Southern California, 1540 E. Alcazar Street, CHP149, Los Angeles, CA 90089, USA.
| | - Amy Hegarty
- Division of Biokinesiology and Physical
Therapy, Ostrow School of Dentistry, University of Southern California, Los Angeles,
CA, USA
| | - Lirong Yan
- Mark and Mary Stevens Neuroimaging and
Informatics Institute, Department of Neurology, Keck School of Medicine, University
of Southern California, Los Angeles, CA, USA
| | - Dominique Duncan
- Mark and Mary Stevens Neuroimaging and
Informatics Institute, Department of Neurology, Keck School of Medicine, University
of Southern California, Los Angeles, CA, USA
| | - Judy Pa
- Mark and Mary Stevens Neuroimaging and
Informatics Institute, Department of Neurology, Keck School of Medicine, University
of Southern California, Los Angeles, CA, USA
| | - Jason J Kutch
- Division of Biokinesiology and Physical
Therapy, Ostrow School of Dentistry, University of Southern California, Los Angeles,
CA, USA
| | - Marianna La Rocca
- Mark and Mary Stevens Neuroimaging and
Informatics Institute, Department of Neurology, Keck School of Medicine, University
of Southern California, Los Angeles, CA, USA,Department of Preventive Medicine, Keck
School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Christianne J Lane
- Dipartimento Interateneo di Fisica,
Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - E Todd Schroeder
- Division of Biokinesiology and Physical
Therapy, Ostrow School of Dentistry, University of Southern California, Los Angeles,
CA, USA
| |
Collapse
|