1
|
Nyúl-Tóth Á, Patai R, Csiszar A, Ungvari A, Gulej R, Mukli P, Yabluchanskiy A, Benyo Z, Sotonyi P, Prodan CI, Liotta EM, Toth P, Elahi F, Barsi P, Maurovich-Horvat P, Sorond FA, Tarantini S, Ungvari Z. Linking peripheral atherosclerosis to blood-brain barrier disruption: elucidating its role as a manifestation of cerebral small vessel disease in vascular cognitive impairment. GeroScience 2024; 46:6511-6536. [PMID: 38831182 PMCID: PMC11494622 DOI: 10.1007/s11357-024-01194-0] [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: 04/10/2024] [Accepted: 05/06/2024] [Indexed: 06/05/2024] Open
Abstract
Aging plays a pivotal role in the pathogenesis of cerebral small vessel disease (CSVD), contributing to the onset and progression of vascular cognitive impairment and dementia (VCID). In older adults, CSVD often leads to significant pathological outcomes, including blood-brain barrier (BBB) disruption, which in turn triggers neuroinflammation and white matter damage. This damage is frequently observed as white matter hyperintensities (WMHs) in neuroimaging studies. There is mounting evidence that older adults with atherosclerotic vascular diseases, such as peripheral artery disease, ischemic heart disease, and carotid artery stenosis, face a heightened risk of developing CSVD and VCID. This review explores the complex relationship between peripheral atherosclerosis, the pathogenesis of CSVD, and BBB disruption. It explores the continuum of vascular aging, emphasizing the shared pathomechanisms that underlie atherosclerosis in large arteries and BBB disruption in the cerebral microcirculation, exacerbating both CSVD and VCID. By reviewing current evidence, this paper discusses the impact of endothelial dysfunction, cellular senescence, inflammation, and oxidative stress on vascular and neurovascular health. This review aims to enhance understanding of these complex interactions and advocate for integrated approaches to manage vascular health, thereby mitigating the risk and progression of CSVD and VCID.
Collapse
Affiliation(s)
- Ádám Nyúl-Tóth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Anna Ungvari
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary.
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - Zoltan Benyo
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN, Semmelweis University, 1094, Budapest, Hungary
| | - Peter Sotonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Calin I Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Eric M Liotta
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Peter Toth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- Neurotrauma Research Group, Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- ELKH-PTE Clinical Neuroscience MR Research Group, University of Pecs, Pecs, Hungary
| | - Fanny Elahi
- Departments of Neurology and Neuroscience Ronald M. Loeb Center for Alzheimer's Disease Friedman Brain Institute Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Péter Barsi
- ELKH-SE Cardiovascular Imaging Research Group, Department of Radiology, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Pál Maurovich-Horvat
- ELKH-SE Cardiovascular Imaging Research Group, Department of Radiology, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Farzaneh A Sorond
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| |
Collapse
|
2
|
Mogollón García HD, de Andrade Ferrazza R, Ochoa JC, de Athayde FF, Vidigal PMP, Wiltbank M, Kastelic JP, Sartori R, Ferreira JCP. Landscape transcriptomic analysis of bovine follicular cells during key phases of ovarian follicular development. Biol Res 2024; 57:76. [PMID: 39468655 PMCID: PMC11514973 DOI: 10.1186/s40659-024-00558-2] [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: 05/24/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND There are many gaps in our understanding of the mechanisms involved in ovarian follicular development in cattle, particularly regarding follicular deviation, acquisition of ovulatory capacity, and preovulatory changes. Molecular evaluations of ovarian follicular cells during follicular development in cattle, especially serial transcriptomic analyses across key growth phases, have not been reported. This study aims to address this gap by analyzing gene expression using RNA-seq in granulosa and antral cells recovered from ovarian follicular fluid during critical phases of ovarian follicular development in Holstein cows. RESULTS Integrated analysis of gene ontology (GO), gene set enrichment (GSEA), protein-protein interaction (PPI), and gene topology identified that differentially expressed genes (DEGs) in the largest ovarian follicles at deviation (Dev) were primarily involved in FSH-negative feedback, steroidogenesis, cell proliferation, apoptosis, and the prevention of early follicle rupture. In contrast, DEGs in the second largest follicles (DevF2) were mainly related to loss of cell viability, apoptosis, and immune cell invasion. In the dominant (PostDev) and preovulatory (PreOv) follicles, DEGs were associated with vascular changes and inflammatory responses. CONCLUSIONS The transcriptome of ovarian follicular fluid cells had a predominance of granulosa cells in the dominant follicle at deviation, with upregulation of genes involved in cell viability, steroidogenesis, and apoptosis prevention, whereas in the non-selected follicle there was upregulation of cell death-related transcripts. Immune cell transcripts increased significantly after deviation, particularly in preovulatory follicles, indicating strong intrafollicular chemotactic activity. We inferred that immune cell invasion occurred despite an intact basal lamina, contributing to follicular maturation.
Collapse
Affiliation(s)
- Henry David Mogollón García
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Rua Prof. Doutor Walter Mauricio Correa, s/n, Botucatu, São Paulo, 18618-681, Brazil
- Department of Genetic, Evolution, Microbiology and Immunology. Biology Institute, Campinas State University, Campinas, São Paulo, Brazil
- Computational Systems Biology Laboratory (CSBL), Institut Pasteur, University of São Paulo (USP), São Paulo, Brazil
| | | | - Julian Camilo Ochoa
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Rua Prof. Doutor Walter Mauricio Correa, s/n, Botucatu, São Paulo, 18618-681, Brazil
| | - Flávia Florencio de Athayde
- Department of Animal Production and Health, School of Veterinary Medicine, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | | | - Milo Wiltbank
- Department of Animal & Dairy Sciences, University of Wisconsin-Madison, Madison, USA
| | | | - Roberto Sartori
- Department of Animal Science, Luiz de Queiroz College of Agriculture (ESALQ), University of São Paulo, Piracicaba, São Paulo, Brazil
| | - João Carlos Pinheiro Ferreira
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Rua Prof. Doutor Walter Mauricio Correa, s/n, Botucatu, São Paulo, 18618-681, Brazil.
| |
Collapse
|
3
|
Nguyen S, McEvoy LK, Espeland MA, Whitsel EA, Lu A, Horvath S, Manson JE, Rapp SR, Shadyab AH. Associations of Epigenetic Age Estimators With Cognitive Function Trajectories in the Women's Health Initiative Memory Study. Neurology 2024; 103:e209534. [PMID: 38857479 PMCID: PMC11226313 DOI: 10.1212/wnl.0000000000209534] [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: 01/04/2024] [Accepted: 04/05/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Epigenetic age estimators indicating faster/slower biological aging vs chronological age independently associate with several age-related outcomes; however, longitudinal associations with cognitive function are understudied. We examined associations of epigenetic age estimators with cognitive function measured annually. METHODS This longitudinal study consisted of older women enrolled in the Women's Health Initiative Memory Study with DNA methylation (DNAm) collected at baseline (1995-1998) from 3 ancillary studies and were followed up to 13 years. Global cognitive function was measured annually by Modified Mini-Mental State Examination (3MS; baseline-2007) and by modified Telephone Interview for Cognitive Status (TICS-m, 2008-2021). We calculated 5 epigenetic age estimators: extrinsic AgeAccel, intrinsic AgeAccel, AgeAccelPheno, AgeAccelGrim2, Dunedin Pace of Aging Calculated From the Epigenome (DunedinPACE), and AgeAccelGrim2 components (DNA-based plasma protein surrogates). We estimated longitudinal epigenetic age estimator-cognitive function associations using linear mixed-effects models containing age, education, race or ethnicity, and subsequently alcohol, smoking, body mass index, and comorbidities. We examined effect modification by APOE ε4 carriage. RESULTS A total of 795 participants were enrolled. The mean baseline age was 70.8 ± 4 years (10.7% Black, 3.9% Hispanic or Latina, 85.4% White), A 1-SD (0.12) increment in DunedinPACE associated with faster annual declines in TICS-m scores in minimally adjusted (β = -0.118, 95% CI -0.202 to -0.034; p = 0.0006) and fully adjusted (β = -0.123, 95% CI -0.211 to -0.036; p = 0.006) models. AgeAccelPheno associated with faster annual declines in TICS-m with minimal adjustment (β = -0.091, 95% CI -0.176 to -0.006; p = 0.035) but not with full adjustment. No other epigenetic age estimators associated with changes in 3MS or TICS-m. Higher values of DNAm-based surrogates of growth differentiation factor 15, beta-2 microglobulin, Cystatin C, tissue inhibitor metalloproteinase 1, and adrenomedullin associated with faster annual declines in 3MS and TICS-m. Higher DNAm log A1c associated with faster annual declines in TICS-m only. DunedinPACE associated with faster annual declines in 3MS among APOE ε4 carriers but not among noncarriers (p-interaction = 0.020). DISCUSSION Higher DunedinPACE associated with faster declines in TICS-m and 3MS scores among APOE ε4 carriers. DunedinPACE may help identify older women at risk of future cognitive decline. Limitations include the ancillary studies that collected epigenetic data not designed to study epigenetics and cognitive function. We examined epigenetic age estimators with global cognitive function and not specific cognitive domains. Findings may not generalize to men and more diverse populations.
Collapse
Affiliation(s)
- Steve Nguyen
- From the Division of Epidemiology (S.N., L.K.M., A.H.S.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla; Kaiser Permanente Washington Health Research Institute (L.K.M.), Seattle, WA; Departments of Internal Medicine and Biostatistics and Data Science (M.A.E.), Wake Forest University School of Medicine, Winston-Salem, NC; Department of Epidemiology (E.A.W.), Gillings School of Global Public Health; Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill; Altos Labs (A.L., S.H.), San Diego, CA; Department of Epidemiology (S.H.), UCLA Fielding School of Public Health, Los Angeles, CA; Division of Preventive Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Psychiatry & Behavioral Medicine (S.R.R.), Wake Forest School of Medicine, Winston-Salem, NC; and Division of Geriatrics, Gerontology, and Palliative Care (A.H.S.), Department of Medicine, University of California, San Diego, La Jolla
| | - Linda K McEvoy
- From the Division of Epidemiology (S.N., L.K.M., A.H.S.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla; Kaiser Permanente Washington Health Research Institute (L.K.M.), Seattle, WA; Departments of Internal Medicine and Biostatistics and Data Science (M.A.E.), Wake Forest University School of Medicine, Winston-Salem, NC; Department of Epidemiology (E.A.W.), Gillings School of Global Public Health; Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill; Altos Labs (A.L., S.H.), San Diego, CA; Department of Epidemiology (S.H.), UCLA Fielding School of Public Health, Los Angeles, CA; Division of Preventive Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Psychiatry & Behavioral Medicine (S.R.R.), Wake Forest School of Medicine, Winston-Salem, NC; and Division of Geriatrics, Gerontology, and Palliative Care (A.H.S.), Department of Medicine, University of California, San Diego, La Jolla
| | - Mark A Espeland
- From the Division of Epidemiology (S.N., L.K.M., A.H.S.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla; Kaiser Permanente Washington Health Research Institute (L.K.M.), Seattle, WA; Departments of Internal Medicine and Biostatistics and Data Science (M.A.E.), Wake Forest University School of Medicine, Winston-Salem, NC; Department of Epidemiology (E.A.W.), Gillings School of Global Public Health; Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill; Altos Labs (A.L., S.H.), San Diego, CA; Department of Epidemiology (S.H.), UCLA Fielding School of Public Health, Los Angeles, CA; Division of Preventive Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Psychiatry & Behavioral Medicine (S.R.R.), Wake Forest School of Medicine, Winston-Salem, NC; and Division of Geriatrics, Gerontology, and Palliative Care (A.H.S.), Department of Medicine, University of California, San Diego, La Jolla
| | - Eric A Whitsel
- From the Division of Epidemiology (S.N., L.K.M., A.H.S.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla; Kaiser Permanente Washington Health Research Institute (L.K.M.), Seattle, WA; Departments of Internal Medicine and Biostatistics and Data Science (M.A.E.), Wake Forest University School of Medicine, Winston-Salem, NC; Department of Epidemiology (E.A.W.), Gillings School of Global Public Health; Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill; Altos Labs (A.L., S.H.), San Diego, CA; Department of Epidemiology (S.H.), UCLA Fielding School of Public Health, Los Angeles, CA; Division of Preventive Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Psychiatry & Behavioral Medicine (S.R.R.), Wake Forest School of Medicine, Winston-Salem, NC; and Division of Geriatrics, Gerontology, and Palliative Care (A.H.S.), Department of Medicine, University of California, San Diego, La Jolla
| | - Ake Lu
- From the Division of Epidemiology (S.N., L.K.M., A.H.S.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla; Kaiser Permanente Washington Health Research Institute (L.K.M.), Seattle, WA; Departments of Internal Medicine and Biostatistics and Data Science (M.A.E.), Wake Forest University School of Medicine, Winston-Salem, NC; Department of Epidemiology (E.A.W.), Gillings School of Global Public Health; Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill; Altos Labs (A.L., S.H.), San Diego, CA; Department of Epidemiology (S.H.), UCLA Fielding School of Public Health, Los Angeles, CA; Division of Preventive Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Psychiatry & Behavioral Medicine (S.R.R.), Wake Forest School of Medicine, Winston-Salem, NC; and Division of Geriatrics, Gerontology, and Palliative Care (A.H.S.), Department of Medicine, University of California, San Diego, La Jolla
| | - Steve Horvath
- From the Division of Epidemiology (S.N., L.K.M., A.H.S.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla; Kaiser Permanente Washington Health Research Institute (L.K.M.), Seattle, WA; Departments of Internal Medicine and Biostatistics and Data Science (M.A.E.), Wake Forest University School of Medicine, Winston-Salem, NC; Department of Epidemiology (E.A.W.), Gillings School of Global Public Health; Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill; Altos Labs (A.L., S.H.), San Diego, CA; Department of Epidemiology (S.H.), UCLA Fielding School of Public Health, Los Angeles, CA; Division of Preventive Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Psychiatry & Behavioral Medicine (S.R.R.), Wake Forest School of Medicine, Winston-Salem, NC; and Division of Geriatrics, Gerontology, and Palliative Care (A.H.S.), Department of Medicine, University of California, San Diego, La Jolla
| | - Joann E Manson
- From the Division of Epidemiology (S.N., L.K.M., A.H.S.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla; Kaiser Permanente Washington Health Research Institute (L.K.M.), Seattle, WA; Departments of Internal Medicine and Biostatistics and Data Science (M.A.E.), Wake Forest University School of Medicine, Winston-Salem, NC; Department of Epidemiology (E.A.W.), Gillings School of Global Public Health; Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill; Altos Labs (A.L., S.H.), San Diego, CA; Department of Epidemiology (S.H.), UCLA Fielding School of Public Health, Los Angeles, CA; Division of Preventive Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Psychiatry & Behavioral Medicine (S.R.R.), Wake Forest School of Medicine, Winston-Salem, NC; and Division of Geriatrics, Gerontology, and Palliative Care (A.H.S.), Department of Medicine, University of California, San Diego, La Jolla
| | - Stephen R Rapp
- From the Division of Epidemiology (S.N., L.K.M., A.H.S.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla; Kaiser Permanente Washington Health Research Institute (L.K.M.), Seattle, WA; Departments of Internal Medicine and Biostatistics and Data Science (M.A.E.), Wake Forest University School of Medicine, Winston-Salem, NC; Department of Epidemiology (E.A.W.), Gillings School of Global Public Health; Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill; Altos Labs (A.L., S.H.), San Diego, CA; Department of Epidemiology (S.H.), UCLA Fielding School of Public Health, Los Angeles, CA; Division of Preventive Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Psychiatry & Behavioral Medicine (S.R.R.), Wake Forest School of Medicine, Winston-Salem, NC; and Division of Geriatrics, Gerontology, and Palliative Care (A.H.S.), Department of Medicine, University of California, San Diego, La Jolla
| | - Aladdin H Shadyab
- From the Division of Epidemiology (S.N., L.K.M., A.H.S.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla; Kaiser Permanente Washington Health Research Institute (L.K.M.), Seattle, WA; Departments of Internal Medicine and Biostatistics and Data Science (M.A.E.), Wake Forest University School of Medicine, Winston-Salem, NC; Department of Epidemiology (E.A.W.), Gillings School of Global Public Health; Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill; Altos Labs (A.L., S.H.), San Diego, CA; Department of Epidemiology (S.H.), UCLA Fielding School of Public Health, Los Angeles, CA; Division of Preventive Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Psychiatry & Behavioral Medicine (S.R.R.), Wake Forest School of Medicine, Winston-Salem, NC; and Division of Geriatrics, Gerontology, and Palliative Care (A.H.S.), Department of Medicine, University of California, San Diego, La Jolla
| |
Collapse
|
4
|
Washida K, Saito S, Tanaka T, Nakaoku Y, Ishiyama H, Abe S, Kuroda T, Nakazawa S, Kakuta C, Omae K, Tanaka K, Minami M, Morita Y, Fukuda T, Shindo A, Maki T, Kitamura K, Tomimoto H, Aso T, Ihara M. A multicenter, single-arm, phase II clinical trial of adrenomedullin in patients with cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2024; 6:100211. [PMID: 38375188 PMCID: PMC10875187 DOI: 10.1016/j.cccb.2024.100211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/21/2024] [Accepted: 01/31/2024] [Indexed: 02/21/2024]
Abstract
Background Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), the most common form of hereditary cerebral small vessel disease (SVD), currently lacks disease-modifying treatments. Adrenomedullin (AM), a vasoactive peptide with angiogenic, vasodilatory, anti-inflammatory, and anti-oxidative properties, shows potential effects on the neuro-glial-vascular unit. Objective The AdrenoMedullin for CADASIL (AMCAD) study aims to assess the efficacy and safety of AM in patients with CADASIL. Sample size Overall, 60 patients will be recruited. Methods The AMCAD is a multicenter, investigator-initiated, single-arm phase II trial. Patients with a confirmed CADASIL diagnosis, based on NOTCH3 genetic testing, will receive an 8-h AM treatment (15 ng/kg/min) for 14 days following a baseline assessment (from day 1 to day 14). Follow-up evaluations will be performed on days 15, 28, 90, and 180. Study outcomes The primary endpoint is the cerebral blood flow change rate in the frontal cortex, evaluated using arterial spin labeling magnetic resonance imaging, from baseline to day 28. Summary statistics, 95% confidence intervals, and a one-sample t-test will be used for analysis. Conclusion The AMCAD study aims to represent the therapeutic potential of AM in patients with CADASIL, addressing an unmet medical need in this challenging condition. Clinical Trial Registration jRCT 2,051,210,117 (https://jrct.niph.go.jp/en-latest-detail/jRCT2051210117).
Collapse
Affiliation(s)
- Kazuo Washida
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Satoshi Saito
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Tomotaka Tanaka
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Yuriko Nakaoku
- Department of Preventive Medicine and Epidemiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Hiroyuki Ishiyama
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Soichiro Abe
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Takehito Kuroda
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Shinsaku Nakazawa
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Chikage Kakuta
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Katsuhiro Omae
- Department of Data Science, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Kenta Tanaka
- Department of Data Science, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Manabu Minami
- Department of Data Science, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Yoshiaki Morita
- Department of Radiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Tetsuya Fukuda
- Department of Radiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate school of Medicine, Tsu, Japan
| | - Takakuni Maki
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kazuo Kitamura
- Department of Projects Research, Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate school of Medicine, Tsu, Japan
| | - Toshihiko Aso
- Laboratory for Brain Connectomics Imaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| |
Collapse
|
5
|
Hosoki S, Hansra GK, Jayasena T, Poljak A, Mather KA, Catts VS, Rust R, Sagare A, Kovacic JC, Brodtmann A, Wallin A, Zlokovic BV, Ihara M, Sachdev PS. Molecular biomarkers for vascular cognitive impairment and dementia. Nat Rev Neurol 2023; 19:737-753. [PMID: 37957261 DOI: 10.1038/s41582-023-00884-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 11/15/2023]
Abstract
As disease-specific interventions for dementia are being developed, the ability to identify the underlying pathology and dementia subtypes is increasingly important. Vascular cognitive impairment and dementia (VCID) is the second most common cause of dementia after Alzheimer disease, but progress in identifying molecular biomarkers for accurate diagnosis of VCID has been relatively limited. In this Review, we examine the roles of large and small vessel disease in VCID, considering the underlying pathophysiological processes that lead to vascular brain injury, including atherosclerosis, arteriolosclerosis, ischaemic injury, haemorrhage, hypoperfusion, endothelial dysfunction, blood-brain barrier breakdown, inflammation, oxidative stress, hypoxia, and neuronal and glial degeneration. We consider the key molecules in these processes, including proteins and peptides, metabolites, lipids and circulating RNA, and consider their potential as molecular biomarkers alone and in combination. We also discuss the challenges in translating the promise of these biomarkers into clinical application.
Collapse
Affiliation(s)
- Satoshi Hosoki
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Gurpreet K Hansra
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Tharusha Jayasena
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Anne Poljak
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, Australia
| | - Karen A Mather
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Vibeke S Catts
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Ruslan Rust
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Abhay Sagare
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jason C Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, NY, USA
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Amy Brodtmann
- Department of Neurology, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Anders Wallin
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
6
|
Ishiyama H, Tanaka T, Saito S, Koyama T, Kitamura A, Inoue M, Fukushima N, Morita Y, Koga M, Toyoda K, Kuriyama N, Urushitani M, Ihara M. Plasma mid-regional pro-adrenomedullin: A biomarker of the ischemic penumbra in hyperacute stroke. Brain Pathol 2022; 33:e13110. [PMID: 35916272 PMCID: PMC10041162 DOI: 10.1111/bpa.13110] [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: 03/17/2022] [Accepted: 07/15/2022] [Indexed: 12/01/2022] Open
Abstract
Reperfusion therapy has improved the outcomes of ischemic stroke but also emphasized the importance of ischemic penumbra. However, blood biomarkers are currently unavailable for this region. Adrenomedullin (ADM) is a neuroprotective peptide, secreted in a compensatory response to brain ischemia. We thus investigated whether an increase in mid-regional pro-ADM (MR-proADM), a stable peptide fragment of the ADM precursor, could act as a biomarker by predicting the ischemic penumbra in hyperacute ischemic stroke (HAIS). We prospectively enrolled consecutive HAIS patients (n = 119; median age, 77 years; male, 59.7%) admitted to our institutes from July 2017 to March 2019 and evaluated plasma MR-proADM levels within 4.5 h of onset. MR-proADM levels in HAIS were compared to healthy controls (n = 1298; median age, 58 years; male, 33.2%) in the Japan Multi-Institutional Collaborative Cohort Study from 2013 to 2017. Furthermore, we evaluated whether MR-proADM levels were associated with the penumbra estimated by clinical-diffusion mismatch (CDM) (National Institute of Health Stroke Scale [NIHSS] ≥8, diffusion ischemic core volume ≤25 ml), or magnetic resonance angiography-diffusion-weighted imaging mismatch (MDM) (NIHSS ≥5, a proximal vessel occlusion with core volume ≤25 ml, or a proximal vessel stenosis/distal vessel occlusion with core volume ≤15 ml). In a case-control study, multivariate logistic analysis showed a significant association between HAIS and MR-proADM ≥0.54 nmol/L (adjusted odds ratio, 7.92 [95% CI, 4.17-15.02], p < 0.001). Though MR-proADM levels in HAIS did not correlate with the ischemic core volume (rs = 0.09, p = 0.348), they were higher in HAIS with CDM (n = 34; 0.81 vs. 0.61 nmol/L, p < 0.001) or MDM (n = 26; 0.83 vs. 0.62 nmol/L, p = 0.002). These differences remained significant after adjusting baseline factors (adjusted odds ratio, 4.06 [95% CI, 1.31-12.55], p = 0.015 and 4.65 [1.35-16.11], p = 0.015, respectively). Plasma MR-proADM is elevated in HAIS, especially in those with a substantial penumbra, suggesting potential as a blood biomarker in this region.
Collapse
Affiliation(s)
- Hiroyuki Ishiyama
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Tomotaka Tanaka
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Satoshi Saito
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Teruhide Koyama
- Department of Epidemiology for Community Health and MedicineKyoto Prefectural University of MedicineKyotoJapan
| | - Akihiro Kitamura
- Department of NeurologyShiga University of Medical ScienceOtsuJapan
| | - Manabu Inoue
- Department of Cerebrovascular MedicineNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Naoya Fukushima
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Yoshiaki Morita
- Department of RadiologyNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Masatoshi Koga
- Department of Cerebrovascular MedicineNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Kazunori Toyoda
- Department of Cerebrovascular MedicineNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Nagato Kuriyama
- Department of Epidemiology for Community Health and MedicineKyoto Prefectural University of MedicineKyotoJapan
- Shizuoka Graduate University of Public HealthShizuokaJapan
| | | | - Masafumi Ihara
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaJapan
| |
Collapse
|