1
|
Luo H, Zheng Z, Yuan Z, Hu H, Sun C. The effectiveness of multicomponent exercise in older adults with cognitive frailty: a systematic review and meta-analysis. Arch Public Health 2024; 82:229. [PMID: 39614334 DOI: 10.1186/s13690-024-01441-y] [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: 03/15/2024] [Accepted: 10/31/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND Cognitive frailty, intimately tied to adverse outcomes such as falls, early mortality, and hospitalization, represents a dynamic, reversible process. Multicomponent exercise has emerged as one of the most potent means of mitigating cognitive frailty. AIMS This research seeks to quantitively amalgamate the effects of multicomponent exercise on various domains: cognitive function, frailty status, and other health-related outcomes in cognitively frail older adults. METHODS Our methodology entailed a comprehensive review of literature in databases including PubMed, EMbase, CINAHL, Cochrane Library, Web of Science, Wanfang, Sinomed, VIP, and CNKI from the inception of these databases to December 10, 2023. For our statistical analysis, we utilized RevMan 5.3, Stata 17.0 and R 4.3.2 software. Adherence was maintained to the PRISMA checklist, with the study being registered with PROSPERO (CRD42024499808). RESULTS Our review encapsulated a total of 2,222 participants and 11 trials. The findings intimate that multicomponent exercise enhances cognitive function [MD = 2.52, p = 0.03]), grip strength[SMD = 0.39, p = 0.008] and lower limb muscle strength[MD = 4.30, p < 0.001], while alleviating frailty[MD = -2.21, p < 0.001] and depression [MD = -1.20, p = 0.001]. However, cogent evidence is still lacking to endorse the positive effects of multicomponent exercises on both ADL(p = 0.19) and quality of life(p = 0.16). Subgroup analyses revealed beneficial effects on cognitive frailty for multicomponent exercise whose type of exercise consisted of aerobic, the duration of which exceeded 120 min per week, and whose form of exercise was group exercise. CONCLUSION Multicomponent exercises offer significant improvements in cognitive function, muscle strength, and have the added benefit of reducing frailty and depression in older adults. However, these exercises do not appear to influence activities of daily living and quality of life positively.
Collapse
Affiliation(s)
- Huanhuan Luo
- Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of MedicalSciences and Peking Union Medical College, Beijing, People's Republic of China
- Department of Nursing, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Science, NO.1 Da Hua Road, DongDan, Beijing, 100730, People's Republic of China
| | - Zitian Zheng
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, People's Republic of China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, People's Republic of China
| | - Zhe Yuan
- Department of Orthopedics, the Fourth Medical Centre, Chineses PLA General Hospital, 51 Fucheng Road, Beijing, 100048, People's Republic of China.
| | - Huixiu Hu
- Department of Nursing, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Science, NO.1 Da Hua Road, DongDan, Beijing, 100730, People's Republic of China.
| | - Chao Sun
- Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of MedicalSciences and Peking Union Medical College, Beijing, People's Republic of China.
- Department of Nursing, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Science, NO.1 Da Hua Road, DongDan, Beijing, 100730, People's Republic of China.
| |
Collapse
|
2
|
Son HJ, Kim JS, Bateman RJ, Kim S, Llibre-Guerra JJ, Day GS, Chhatwal JP, Berman SB, Schofield PR, Jucker M, Levin J, Lee JH, Perrin RJ, Morris JC, Cruchaga C, Hassenstab J, Salloway SP, Lee JH, Daniels A. Association of Resilience-Related Life Experiences on Variability on Age of Onset in Dominantly Inherited Alzheimer Disease. Neurology 2024; 103:e209766. [PMID: 39270149 PMCID: PMC11399067 DOI: 10.1212/wnl.0000000000209766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/25/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND AND OBJECTIVES It remains unknown whether the associations between protective lifestyles and sporadic dementia risk reported in observational studies also affect age at symptom onset (AAO) in autosomal dominant Alzheimer disease (ADAD) with predominant genetic influences. We investigated the associations between resilience-related life experiences and interindividual AAO variability in ADAD. METHODS We performed a longitudinal and confirmatory analysis of the Dominantly Inherited Alzheimer Network prospective observational cohort (January 2009-June 2018, follow-up duration 2.13 ± 2.22 years), involving clinical, CSF, and lifestyle/behavioral assessments. We performed a 2-pronged comprehensive resilience assessment in each cohort. Cohort 1, incorporating the general resilience definition (cognitive maintenance [Clinical Dementia Rating = 0] despite high pathology), included carriers during the periods of significant CSFp-tau181 variability and grouped into resilience/resistance outcome bins according to the dichotomous pathologic and cognitive statuses, subcategorized by the estimated years from expected symptom onset (EYO). Cohort 2, focused on ADAD-specific genetically determined time frame characterizing the onset predictability, included asymptomatic participants with available preclinical lifestyle data and AAO outcomes and grouped into delayed or earlier AAO relative to the parental AAO. Associations of cognitive, CSFp-tau181, and lifestyle/behavioral predictors with binary outcomes were investigated using logistic regression. RESULTS Of 320 carriers (age 38.19 ± 10.94 years, female 56.25%), cohort 1 included 218 participants (39.00 ± 9.37 years, 57.34%) and cohort 2 included 28 participants (43.34 ± 7.40 years, 71.43%). In cohort 1, 218 carriers after -20 EYO, when the interindividual variability (SD) of CSFp-tau181 first became more than twice greater in carriers than in noncarriers, were grouped into low-risk control (asymptomatic, low pathology, n = 103), high-resilience (asymptomatic despite high pathology, n = 60), low-resilience (symptomatic despite low pathology, n = 15), and susceptible control (symptomatic, high pathology, n = 40) groups. Multivariable predictors of high resilience, controlling for age and depression, included higher conscientiousness (odds ratio 1.051 [95% CI 1.016-1.086], p = 0.004), openness to experience (1.068 [1.005-1.135], p = 0.03) (vs. susceptible controls), and agreeableness (1.082 [1.015-1.153], p = 0.02) (vs. low resilience). From 1 to 3 years before parental AAO (cohort 2), the multivariable predictor of delayed AAO, controlling for CSFp-tau181, was higher conscientiousness (0.916 [0.845-0.994], p = 0.036). DISCUSSION Among the cognitively and socially integrated life experiences associated with resilience, measures of conscientiousness were useful indicators for evaluating resilience and predicting future dementia onset in late preclinical ADAD.
Collapse
Affiliation(s)
- Hye Joo Son
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - Jae Seung Kim
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - Randall J Bateman
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - Seonok Kim
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - Jorge J Llibre-Guerra
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - Gregory S Day
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - Jasmeer P Chhatwal
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - Sarah B Berman
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - Peter R Schofield
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - Mathias Jucker
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - Johannes Levin
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - Jae-Hong Lee
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - Richard J Perrin
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - John C Morris
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - Carlos Cruchaga
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - Jason Hassenstab
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - Stephen P Salloway
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - Jai-Hyuen Lee
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| | - Alisha Daniels
- From the Department of Nuclear Medicine (H.J.S., Jai-Hyuen Lee), Dankook University College of Medicine, Cheonan, Chung Nam; Department of Nuclear Medicine (J.S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (R.J.B., J.J.L.-G., J.C.M., A.D.), Washington University School of Medicine, St. Louis, MO; Department of Clinical Epidemiology and Biostatistics (S.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Neurology (G.S.D.), Mayo Clinic College of Medicine and Science, Jacksonville, FL; Department of Neurology (J.P.C.), Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (S.B.B.), University of Pittsburgh School of Medicine, PA; Neuroscience Research Australia (P.R.S.); School of Medical Sciences (P.R.S.), University of New South Wales, Sydney, Australia; Department of Cellular Neurology (M.J.), Hertie Institute for Clinical Brain Research, University of Tübingen; German Center for Neurodegenerative Diseases (M.J.), Tübingen; Department of Neurology (J.L.), Ludwig-Maximilians-Universität München; German Center for Neurodegenerative Diseases (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.), Germany; Department of Neurology (Jae-Hong Lee), University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology and Immunology (R.J.P.), Knight Alzheimer's Disease Research Center (R.J.P., J.H.), and Department of Neurology (R.J.P., J.H.), Washington University in St. Louis; Department of Psychiatry (C.C.), Washington University School of Medicine; Department of Psychological and Brain Sciences (J.H.), Washington University, St. Louis, MO; and Department of Neurology (S.P.S.), The Warren Alpert Medical School of Brown University, Butler Hospital, Providence, RI
| |
Collapse
|
3
|
Luzum G, Gunnes M, Lydersen S, Saltvedt I, Tan X, Thingstad P, Thrane G, Askim T. Physical Activity Behavior and Its Association With Global Cognitive Function Three Months After Stroke: The Nor-COAST Study†. Phys Ther 2023; 103:pzad092. [PMID: 37440440 PMCID: PMC10733132 DOI: 10.1093/ptj/pzad092] [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: 06/06/2022] [Revised: 03/15/2023] [Accepted: 05/22/2023] [Indexed: 07/15/2023]
Abstract
OBJECTIVE The purposes of this study were to determine the association between physical activity (PA) behavior and global cognitive function 3 months after stroke and to explore the role of physical capacity as a mediating factor. METHODS Participants with stroke were successively recruited at 5 different hospitals in Norway. PA was measured using accelerometers, with a follow-up period of 7 consecutive days, and global cognitive function was assessed using the Montreal Cognitive Assessment (MoCA). The general pattern of PA and the percentage of participants adhering to World Health Organization PA recommendations (at least 150 minutes of moderate-intensity aerobic PA per week) were investigated using descriptive statistics. Multiple regression and mediator analyses were used to examine the relationship between PA behavior and MoCA scores; physical capacity, measured with the Short Physical Performance Battery, served as the mediating variable. RESULTS A total of 193 women (42.6%) and 260 men (57.4%) with a median age of 73.7 years (25th and 75th percentiles = 65.8 and 80.4, respectively) and a median MoCA score of 25 points (25th and 75th percentiles = 22 and 27, respectively) were included. Mean total time spent walking at moderate intensity was 251.7 (SD = 164.6) min/wk (mean bout length = 20.9 [SD = 7.3] seconds), which indicated 69.3% adherence to World Health Organization guidelines. With each point decrease in the MoCA score, there was an expected 8.6% increase in the odds of nonadherence to PA recommendations. Physical capacity was identified as an important mediating factor, explaining the strength of the association between cognition and PA behavior. CONCLUSIONS In contrast to previous research, in the present study, most participants adhered to the updated global PA guidelines. However, people who had survived stroke and had reduced cognitive function were at higher risk of inactivity, an association mediated by physical capacity. IMPACT A better understanding of the association between cognition and PA behavior after stroke might help for developing more targeted early-onset interventions.
Collapse
Affiliation(s)
- Geske Luzum
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Science, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Mari Gunnes
- Department of Health Research, SINTEF, Trondheim, Norway
| | - Stian Lydersen
- Department of Mental Health, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Ingvild Saltvedt
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Science, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- Department of Geriatric Medicine, St. Olavs hospital, Trondheim University Hospital, Trondheim, Norway
| | - Xiangchun Tan
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Science, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Pernille Thingstad
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Science, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
- Department of Health and Welfare Services, City of Trondheim, Trondheim, Norway
| | - Gyrd Thrane
- Department of Health and Care Science, Faculty of Health, The Arctic University of Norway, Tromsø, Norway
| | - Torunn Askim
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Science, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
4
|
Su C, Miao J, Guo J. The relationship between TGF-β1 and cognitive function in the brain. Brain Res Bull 2023; 205:110820. [PMID: 37979810 DOI: 10.1016/j.brainresbull.2023.110820] [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: 09/13/2023] [Revised: 11/05/2023] [Accepted: 11/15/2023] [Indexed: 11/20/2023]
Abstract
Transforming growth factor-β1 (TGF-β1), a multifunctional cytokine, plays a pivotal role in synaptic formation, plasticity, and neurovascular unit regulation. This review highlights TGF-β1's potential impact on cognitive function, particularly in the context of neurodegenerative disorders. However, despite the growing body of evidence, a comprehensive understanding of TGF-β1's precise role remains elusive. Further research is essential to unravel the complex mechanisms through which TGF-β1 influences cognitive function and to explore therapeutic avenues for targeting TGF-β1 in neurodegenerative conditions. This investigation sheds light on TGF-β1's contribution to cognitive function and offers prospects for innovative treatments and interventions. This review delves into the intricate relationship between TGF-β1 and cognitive function.
Collapse
Affiliation(s)
- Chen Su
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province 030000, China
| | - Jie Miao
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province 030000, China
| | - Junhong Guo
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province 030000, China.
| |
Collapse
|
5
|
Bangen KJ, Calcetas AT, Thomas KR, Wierenga C, Smith CN, Bordyug M, Brenner EK, Wing D, Chen C, Liu TT, Zlatar ZZ. Greater accelerometer-measured physical activity is associated with better cognition and cerebrovascular health in older adults. J Int Neuropsychol Soc 2023; 29:859-869. [PMID: 36789631 PMCID: PMC10425574 DOI: 10.1017/s1355617723000140] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
OBJECTIVES Physical activity (PA) may help maintain brain structure and function in aging. Since the intensity of PA needed to effect cognition and cerebrovascular health remains unknown, we examined associations between PA and cognition, regional white matter hyperintensities (WMH), and regional cerebral blood flow (CBF) in older adults. METHOD Forty-three older adults without cognitive impairment underwent magnetic resonance imaging (MRI) and comprehensive neuropsychological assessment. Waist-worn accelerometers objectively measured PA for approximately one week. RESULTS Higher time spent in moderate to vigorous PA (MVPA) was uniquely associated with better memory and executive functioning after adjusting for all light PA. Higher MVPA was also uniquely associated with lower frontal WMH volume although the finding was no longer significant after additionally adjusting for age and accelerometer wear time. MVPA was not associated with CBF. Higher time spent in all light PA was uniquely associated with higher CBF but not with cognitive performance or WMH volume. CONCLUSIONS Engaging in PA may be beneficial for cerebrovascular health, and MVPA in particular may help preserve memory and executive function in otherwise cognitively healthy older adults. There may be differential effects of engaging in lighter PA and MVPA on MRI markers of cerebrovascular health although this needs to be confirmed in future studies with larger samples. Future randomized controlled trials that increase PA are needed to elucidate cause-effect associations between PA and cerebrovascular health.
Collapse
Affiliation(s)
- Katherine J Bangen
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Amanda T Calcetas
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Kelsey R Thomas
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Christina Wierenga
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Christine N Smith
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA, USA
| | - Maria Bordyug
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Einat K Brenner
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - David Wing
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA, USA
| | - Conan Chen
- Center for Functional MRI and Department of Radiology, University of California, San Diego, La Jolla, CA, USA
| | - Thomas T Liu
- Center for Functional MRI and Department of Radiology, University of California, San Diego, La Jolla, CA, USA
| | - Zvinka Z Zlatar
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
6
|
Werneck AO, Araujo RHO, Silva DR, Vancampfort D. Handgrip strength, physical activity and incident mild cognitive impairment and dementia. Maturitas 2023; 176:107789. [PMID: 37354742 DOI: 10.1016/j.maturitas.2023.107789] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/15/2023] [Accepted: 06/10/2023] [Indexed: 06/26/2023]
Abstract
OBJECTIVE We analyzed the mediating role of handgrip strength in the association between moderate to vigorous physical activity and incident mild cognitive impairment and dementia. METHODS We used prospective data from 14 European countries participating in the Survey of Health, Ageing, and Retirement in Europe. 19,686 participants free of dementia and mild cognitive impairment (64.9 ± 8.7 years) were followed up for a mean of 10.2 years. Moderate to vigorous physical activity was self-reported, and handgrip strength was assessed with a dynamometer. Mild cognitive impairment was defined as 1.5 standard deviations below the mean of the standardized global cognition score, while dementia was determined by physician diagnosis. Gender, age, country, education, presence of chronic diseases, depressive symptoms, limitations in activities of daily living, body mass index, and baseline cognitive levels were used as covariates. Cox proportional hazards as well as mediation models were used. RESULTS Moderate to vigorous physical activity for at least 1 day per week was independently associated with lower incident mild cognitive impairment (HR: 0.85; 95%CI: 0.74-0.98). A 10 % increase in handgrip strength was associated a 6 % lower hazard for incident mild cognitive impairment (0.94; 0.92-0.97) and 5 % lower hazard for incident dementia (0.95; 0.93-0.98). Handgrip strength partly mediated the association of moderate to vigorous physical activity with mild cognitive impairment (Coefficient: 0.03; 95%CI: 0.01-0.05; 17.9 %). CONCLUSIONS Physical activity is independently associated with a lower incidence of mild cognitive impairment.
Collapse
Affiliation(s)
- André O Werneck
- Center for Epidemiological Research in Nutrition and Health, Department of Nutrition, School of Public Health, Universidade de São Paulo, São Paulo, Brazil.
| | - Raphael H O Araujo
- Graduation Program in Health Sciences, Londrina State University, Londrina, Brazil
| | - Danilo R Silva
- Department of Physical Education, Federal University of Sergipe - UFS, São Cristóvão, Brazil; Faculty of Health Sciences, Universidad Autónoma de Chile, Providencia, Chile
| | - Davy Vancampfort
- Department of Rehabilitation Sciences, University of Leuven and University Psychiatric Center, Katholieke Universiteit Leuven, Belgium
| |
Collapse
|
7
|
Pinto-Hernandez P, Castilla-Silgado J, Coto-Vilcapoma A, Fernández-Sanjurjo M, Fernández-García B, Tomás-Zapico C, Iglesias-Gutiérrez E. Modulation of microRNAs through Lifestyle Changes in Alzheimer's Disease. Nutrients 2023; 15:3688. [PMID: 37686720 PMCID: PMC10490103 DOI: 10.3390/nu15173688] [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: 07/28/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Lifestyle factors, including diet and physical activity (PA), are known beneficial strategies to prevent and delay Alzheimer's disease (AD) development. Recently, microRNAs have emerged as potential biomarkers in multiple diseases, including AD. The aim of this review was to analyze the available information on the modulatory effect of lifestyle on microRNA expression in AD. Few studies have addressed this question, leaving important gaps and limitations: (1) in human studies, only circulating microRNAs were analyzed; (2) in mice studies, microRNA expression was only analyzed in brain tissue; (3) a limited number of microRNAs was analyzed; (4) no human nutritional intervention studies were conducted; and (5) PA interventions in humans and mice were poorly detailed and only included aerobic training. Despite this, some conclusions could be drawn. Circulating levels of let-7g-5p, miR-107, and miR-144-3p were associated with overall diet quality in mild cognitive impairment patients. In silico analysis showed that these microRNAs are implicated in synapse formation, microglia activation, amyloid beta accumulation, and pro-inflammatory pathways, the latter also being targeted by miR-129-5p and miR-192-5p, whose circulating levels are modified by PA in AD patients. PA also modifies miR-132, miR-15b-5p, miR-148b-3p, and miR-130a-5p expression in mice brains, which targets are related to the regulation of neuronal activity, ageing, and pro-inflammatory pathways. This supports the need to further explore lifestyle-related miRNA changes in AD, both as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Paola Pinto-Hernandez
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| | - Juan Castilla-Silgado
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| | - Almudena Coto-Vilcapoma
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| | - Manuel Fernández-Sanjurjo
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| | - Benjamín Fernández-García
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
- Department of Morphology and Cell Biology, Anatomy, University of Oviedo, 33006 Asturias, Spain
| | - Cristina Tomás-Zapico
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| | - Eduardo Iglesias-Gutiérrez
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| |
Collapse
|
8
|
Wang Y, Wu J, Wang J, He L, Lai H, Zhang T, Wang X, Li W. Mitochondrial oxidative stress in brain microvascular endothelial cells: Triggering blood-brain barrier disruption. Mitochondrion 2023; 69:71-82. [PMID: 36709855 DOI: 10.1016/j.mito.2023.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/02/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023]
Abstract
Blood-brain barrier disruption plays an important role in central nervous system diseases. This review provides information on the role of mitochondrial oxidative stress in brain microvascular endothelial cells in cellular dysfunction, the disruption of intercellular junctions, transporter dysfunction, abnormal angiogenesis, neurovascular decoupling, and the involvement and aggravation of vascular inflammation and illustrates related molecular mechanisms. In addition, recent drug and nondrug therapies targeting cerebral vascular endothelial cell mitochondria to repair the blood-brain barrier are discussed. This review shows that mitochondrial oxidative stress disorder in brain microvascular endothelial cells plays a key role in the occurrence and development of blood-brain barrier damage and may be critical in various pathological mechanisms of blood-brain barrier damage. These new findings suggest a potential new strategy for the treatment of central nervous system diseases through mitochondrial modulation of cerebral vascular endothelial cells.
Collapse
Affiliation(s)
- Yi Wang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Jing Wu
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Jiexin Wang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Linxi He
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Han Lai
- School of Foreign Languages, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Tian Zhang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Xin Wang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Weihong Li
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| |
Collapse
|
9
|
Abstract
BackgroundWomen in many cohorts have a higher risk for Alzheimer's disease (AD), the most common form of dementia. Sex is a biological construct whereby differences in disease manifestation and prevalence are rooted in genetic differences between XX and XY combinations of chromosomes. This chapter focuses specifically on sex-driven differences in dementia, as opposed to differences driven by gender - a social construct referring to the societal norms that influence people's roles, relationships, and positional power throughout their lifetime.MethodsUsing a narrative review, this chapter explored the characteristics and risk factors for the dementias, alongside a discussion of sex differences including loss of sex steroid hormones in middle-aged women, differences in the prevalence of cardiovascular diseases and engagement in lifestyle protective factors for dementia.ResultsThe sex difference in AD prevalence may exist because of systematic and historic differences in risk and protective factors for dementia, including level of education obtained and socioeconomic status differences, which can impact on health and dementia risk.Levels of sex steroids decline significantly after menopause in women, whereas this is more gradual in men with age. Animal and cell culture studies show strong biological plausibility for sex steroids to protect the ageing brain against dementia. Sex steroid hormone replacement therapy has in some observational studies shown to protect against AD, but treatment studies in humans have mainly shown disappointing results. Cardiovascular disease (CVD) shares midlife medical risk (e.g. hypertension, hyperlipidaemia, obesity etc.) factors with AD and other forms of dementia, but also with related lifestyle risk - and protective factors (e.g. exercise, not smoking etc.). Men tend to die earlier of CVD, so fewer survive to develop AD at an older age. Those who do survive may have healthier lifestyles and fewer risk factors for both CVD and AD. An earlier age at menopause also confers great risk for both without hormone treatment.DiscussionIt could be the case that the decline in sex steroids around the menopause make women more susceptible to lifestyle-related risk factors associated with dementia and CVD, but this remains to be further investigated. Combining hormone treatment with lifestyle changes in midlife (e.g. exercise) could be an important preventative treatment for dementia and CVD in later life, but this also requires further research.
Collapse
|
10
|
Cheah YK, Lim KK, Ismail H, Mohd Yusoff MF, Kee CC. Can the association between hypertension and physical activity be moderated by age? J Taibah Univ Med Sci 2023; 18:844-854. [PMID: 36852251 PMCID: PMC9957764 DOI: 10.1016/j.jtumed.2022.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 12/01/2022] [Accepted: 12/24/2022] [Indexed: 01/05/2023] Open
Abstract
Objectives Physical inactivity, hypertension and non-communicable diseases are major public concerns across the globe. To our knowledge, there is a lack of research that has investigated the moderating effect of age on the relationship between hypertension and physical activity in developing countries. This study had two objectives: (1) investigating hypertension and sociodemographic factors associated with physical activity and (2) investigating whether age moderates the relationship between hypertension and physical activity. Methods Nationally representative data of Malaysia were used to generate cross-sectional evidence. The sample size was 2156 respondents. An ordered probit regression was utilized to assess factors associated with the practice of physical activity. Results Respondents aged 40-49 years with hypertension were 7.3% less likely to participate in high-level physical activity when compared to those without hypertension. The probability of having a low level of physical activity was 12.3% higher among hypertensive patients aged ≥60. Males, married individuals, less-educated adults, low-income earners, and individuals who were aware of their BMI, had a higher tendency to indulge in a highly active lifestyle than others. Conclusion The effect of hypertension on physical activity was moderated by age. Factors influencing physical activity levels among adults were income, gender, marital status, education, employment status, and BMI awareness.
Collapse
Affiliation(s)
- Yong Kang Cheah
- School of Economics, Finance and Banking, College of Business, Universiti Utara Malaysia, Sintok, Kedah, Malaysia,Corresponding address. School of Economics, Finance and Banking, College of Business, Universiti Utara Malaysia, 06010, Sintok, Kedah, Malaysia
| | - Kuang Kuay Lim
- Centre for Occupational Health Research, Institute for Public Health, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Selangor, Malaysia
| | - Hasimah Ismail
- Centre for Non-Communicable Diseases Research, Institute for Public Health, National Institutes of Health, Ministry of Health Malaysia Shah Alam, Selangor, Malaysia
| | - Muhammad Fadhli Mohd Yusoff
- Centre for Non-Communicable Diseases Research, Institute for Public Health, National Institutes of Health, Ministry of Health Malaysia Shah Alam, Selangor, Malaysia
| | - Chee Cheong Kee
- Sector for Biostatistics and Data Repository, National Institutes of Health, Ministry of Health Malaysia Shah Alam, Selangor, Malaysia
| |
Collapse
|
11
|
Verrall CE, Tran DL, Yang JYM, Lubans DR, Winlaw DS, Ayer J, Celermajer D, Cordina R. Exercise as therapy for neurodevelopmental and cognitive dysfunction in people with a Fontan circulation: A narrative review. Front Pediatr 2023; 11:1111785. [PMID: 36861078 PMCID: PMC9969110 DOI: 10.3389/fped.2023.1111785] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/25/2023] [Indexed: 02/15/2023] Open
Abstract
People with a Fontan circulation are at risk of neurodevelopmental delay and disability, and cognitive dysfunction, that has significant implications for academic and occupational attainment, psychosocial functioning, and overall quality of life. Interventions for improving these outcomes are lacking. This review article discusses current intervention practices and explores the evidence supporting exercise as a potential intervention for improving cognitive functioning in people living with a Fontan circulation. Proposed pathophysiological mechanisms underpinning these associations are discussed in the context of Fontan physiology and avenues for future research are recommended.
Collapse
Affiliation(s)
- Charlotte Elizabeth Verrall
- Heart Centre for Children, The Children's Hospital at Westmead, Sydney, NSW, Australia.,Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Derek Lee Tran
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.,Central Clinical School, The University of Sydney School of Medicine, Sydney, NSW, Australia.,Charles Perkins Centre, Heart Research Institute, Sydney, NSW, Australia
| | - Joseph Yuan-Mou Yang
- Developmental Imaging, Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Neuroscience Research, Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia.,Department of Neurosurgery, Neuroscience Advanced Clinical Imaging Service (NACIS), Royal Children's Hospital, Melbourne, VIC, Australia
| | - David Revalds Lubans
- Centre for Active Living and Learning, College of Human and Social Futures, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - David Scott Winlaw
- Cardiothoracic Surgery, the Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Julian Ayer
- Heart Centre for Children, The Children's Hospital at Westmead, Sydney, NSW, Australia.,Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - David Celermajer
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.,Central Clinical School, The University of Sydney School of Medicine, Sydney, NSW, Australia.,Charles Perkins Centre, Heart Research Institute, Sydney, NSW, Australia
| | - Rachael Cordina
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.,Central Clinical School, The University of Sydney School of Medicine, Sydney, NSW, Australia.,Charles Perkins Centre, Heart Research Institute, Sydney, NSW, Australia.,Heart Research Group, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| |
Collapse
|
12
|
Adams JA, Uryash A, Lopez JR. Non-Invasive Pulsatile Shear Stress Modifies Endothelial Activation; A Narrative Review. Biomedicines 2022; 10:biomedicines10123050. [PMID: 36551807 PMCID: PMC9775985 DOI: 10.3390/biomedicines10123050] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
The monolayer of cells that line both the heart and the entire vasculature is the endothelial cell (EC). These cells respond to external and internal signals, producing a wide array of primary or secondary messengers involved in coagulation, vascular tone, inflammation, and cell-to-cell signaling. Endothelial cell activation is the process by which EC changes from a quiescent cell phenotype, which maintains cellular integrity, antithrombotic, and anti-inflammatory properties, to a phenotype that is prothrombotic, pro-inflammatory, and permeable, in addition to repair and leukocyte trafficking at the site of injury or infection. Pathological activation of EC leads to increased vascular permeability, thrombosis, and an uncontrolled inflammatory response that leads to endothelial dysfunction. This pathological activation can be observed during ischemia reperfusion injury (IRI) and sepsis. Shear stress (SS) and pulsatile shear stress (PSS) are produced by mechanical frictional forces of blood flow and contraction of the heart, respectively, and are well-known mechanical signals that affect EC function, morphology, and gene expression. PSS promotes EC homeostasis and cardiovascular health. The archetype of inducing PSS is exercise (i.e., jogging, which introduces pulsations to the body as a function of the foot striking the pavement), or mechanical devices which induce external pulsations to the body (Enhanced External Pulsation (EECP), Whole-body vibration (WBV), and Whole-body periodic acceleration (WBPA aka pGz)). The purpose of this narrative review is to focus on the aforementioned noninvasive methods to increase PSS, review how each of these modify specific diseases that have been shown to induce endothelial activation and microcirculatory dysfunction (Ischemia reperfusion injury-myocardial infarction and cardiac arrest and resuscitation), sepsis, and lipopolysaccharide-induced sepsis syndrome (LPS)), and review current evidence and insight into how each may modify endothelial activation and how these may be beneficial in the acute and chronic setting of endothelial activation and microvascular dysfunction.
Collapse
Affiliation(s)
- Jose A. Adams
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
- Correspondence:
| | - Arkady Uryash
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
| | - Jose R. Lopez
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
| |
Collapse
|
13
|
Cabral DF, Bigliassi M, Cattaneo G, Rundek T, Pascual-Leone A, Cahalin LP, Gomes-Osman J. Exploring the interplay between mechanisms of neuroplasticity and cardiovascular health in aging adults: A multiple linear regression analysis study. Auton Neurosci 2022; 242:103023. [PMID: 36087362 PMCID: PMC11012134 DOI: 10.1016/j.autneu.2022.103023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 06/13/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Neuroplasticity and cardiovascular health behavior are critically important factors for optimal brain health. OBJECTIVE To assess the association between the efficacy of the mechanisms of neuroplasticity and metrics of cardiovascular heath in sedentary aging adults. METHODS We included thirty sedentary individuals (age = 60.6 ± 3.8 y; 63 % female). All underwent assessments of neuroplasticity, measured by the change in amplitude of motor evoked potentials elicited by single-pulse Transcranial Magnetic Stimulation (TMS) at baseline and following intermittent Theta-Burst (iTBS) at regular intervals. Cardiovascular health measures were derived from the Incremental Shuttle Walking Test and included Heart Rate Recovery (HRR) at 1-min/2-min after test cessation. We also collected plasma levels of brain-derived neurotrophic factor (BDNF), vascular endothelial growth factor (VEGF), and c-reactive protein. RESULTS We revealed moderate but significant relationships between TMS-iTBS neuroplasticity, and the predictors of cardiovascular health (|r| = 0.38 to 0.53, p < .05). HRR1 was the best predictor of neuroplasticity (β = 0.019, p = .002). The best fit model (Likelihood ratio = 5.83, p = .016) of the association between neuroplasticity and HRR1 (β = 0.043, p = .002) was selected when controlling for demographics and health status. VEGF and BDNF plasma levels augmented the association between neuroplasticity and HRR1. CONCLUSIONS Our findings build on existing data demonstrating that TMS may provide insight into neuroplasticity and the role cardiovascular health have on its mechanisms. These implications serve as theoretical framework for future longitudinal and interventional studies aiming to improve cardiovascular and brain health. HRR1 is a potential prognostic measure of cardiovascular health and a surrogate marker of brain health in aging adults.
Collapse
Affiliation(s)
- Danylo F Cabral
- Department of Physical Therapy, University of Miami Miller School of Medicine, Coral Gables, FL, USA.
| | - Marcelo Bigliassi
- Department of Teaching and Learning, Florida International University, Miami, FL, USA
| | - Gabriele Cattaneo
- Institut Guttmann, Institut Universitari de Neurorehabilitació, Badalona, Spain; Department of Medicine, Universitat Autónoma de Barcelona, Bellaterra, Spain
| | - Tatjana Rundek
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA; Evelyn F. McKnight Brain Institute, University of Miami, Miami, FL, USA
| | - Alvaro Pascual-Leone
- Institut Guttmann, Institut Universitari de Neurorehabilitació, Badalona, Spain; Hinda and Arthur Marcus Institute for Aging Research and Deanna and Sidney Wolk Center for Memory Health, Hebrew SeniorLife, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Lawrence P Cahalin
- Department of Physical Therapy, University of Miami Miller School of Medicine, Coral Gables, FL, USA
| | - Joyce Gomes-Osman
- Department of Physical Therapy, University of Miami Miller School of Medicine, Coral Gables, FL, USA; Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
14
|
Soldan A, Alfini A, Pettigrew C, Faria A, Hou X, Lim C, Lu H, Spira AP, Zipunnikov V, Albert M. Actigraphy-estimated physical activity is associated with functional and structural brain connectivity among older adults. Neurobiol Aging 2022; 116:32-40. [PMID: 35551019 PMCID: PMC10167793 DOI: 10.1016/j.neurobiolaging.2022.04.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 02/04/2022] [Accepted: 04/09/2022] [Indexed: 12/20/2022]
Abstract
Higher physical activity levels are associated with reduced cognitive decline among older adults; however, current understanding of underlying brain mechanisms is limited. This cross-sectional study investigated the relationship between actigraphy-estimated total volume of physical activity (TVPA) and magnetic resonance imaging (MRI) measures of white matter hyperintensities (WMH), and functional and structural brain connectivity, measured by resting-state functional MRI and diffusion tensor imaging. Study participants (N = 156, mean age = 71 years) included 136 with normal cognition and 20 with Mild Cognitive Impairment. Higher TVPA was associated with greater functional connectivity within the default-mode network and greater network modularity (a measure of network specialization), as well as with greater anisotropy and lower radial diffusion in white matter, suggesting better structural connectivity. These associations with functional and structural connectivity were independent of one another and independent of the level of vascular risk, APOE-ε4 status, cognitive reserve, and WMH volume, which were not associated with TVPA. Findings suggest that physical activity is beneficial for brain connectivity among older individuals with varying levels of risk for cognitive decline.
Collapse
Affiliation(s)
- Anja Soldan
- Division of Cognitive Neuroscience, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Alfonso Alfini
- National Center on Sleep Disorders Research, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Corinne Pettigrew
- Division of Cognitive Neuroscience, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Andreia Faria
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xirui Hou
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chantelle Lim
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hanzhang Lu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Adam P Spira
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Vadim Zipunnikov
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Marilyn Albert
- Division of Cognitive Neuroscience, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
15
|
Trigiani LJ, Bourourou M, Lacalle-Aurioles M, Lecrux C, Hynes A, Spring S, Fernandes DJ, Sled JG, Lesage F, Schwaninger M, Hamel E. A functional cerebral endothelium is necessary to protect against cognitive decline. J Cereb Blood Flow Metab 2022; 42:74-89. [PMID: 34515549 PMCID: PMC8721775 DOI: 10.1177/0271678x211045438] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 12/30/2022]
Abstract
A vascular insult occurring early in disease onset may initiate cognitive decline leading to dementia, while pharmacological and lifestyle interventions can prevent this progression. Mice with a selective, tamoxifen-inducible deletion of NF-κB essential modulator (Nemo) in brain endothelial cells were studied as a model of vascular cognitive impairment. Groups included NemoFl controls and three NemobeKO groups: One untreated, and two treated with simvastatin or exercise. Social preference and nesting were impaired in NemobeKO mice and were not countered by treatments. Cerebrovascular function was compromised in NemobeKO groups regardless of treatment, with decreased changes in sensory-evoked cerebral blood flow and total hemoglobin levels, and impaired endothelium-dependent vasodilation. NemobeKO mice had increased string vessel pathology, blood-brain barrier disruption, neuroinflammation, and reduced cortical somatostatin-containing interneurons. These alterations were reversed when endothelial function was recovered. Findings strongly suggest that damage to the cerebral endothelium can trigger pathologies associated with dementia and its functional integrity should be an effective target in future therapeutic efforts.
Collapse
Affiliation(s)
- Lianne J Trigiani
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Canada
| | - Miled Bourourou
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Canada
| | - María Lacalle-Aurioles
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Canada
| | - Clotilde Lecrux
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Canada
| | - Amy Hynes
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Canada
| | - Shoshana Spring
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Canada
| | - Darren J Fernandes
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Canada
| | - John G Sled
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Canada
| | - Frédéric Lesage
- Biomedical Engineering Institute, École Polytechnique de Montréal, Montréal, Canada
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Canada
| |
Collapse
|
16
|
Borek-Dorosz A, Pieczara A, Czamara K, Stojak M, Matuszyk E, Majzner K, Brzozowski K, Bresci A, Polli D, Baranska M. What is the ability of inflamed endothelium to uptake exogenous saturated fatty acids? A proof-of-concept study using spontaneous Raman, SRS and CARS microscopy. Cell Mol Life Sci 2022; 79:593. [PMID: 36380212 PMCID: PMC9666316 DOI: 10.1007/s00018-022-04616-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/16/2022] [Accepted: 10/27/2022] [Indexed: 11/17/2022]
Abstract
Endothelial cells (EC) in vivo buffer and regulate the transfer of plasma fatty acid (FA) to the underlying tissues. We hypothesize that inflammation could alter the functionality of the EC, i.e., their capacity and uptake of different FA. The aim of this work is to verify the functionality of inflamed cells by analyzing their ability to uptake and accumulate exogenous saturated FA. Control and inflammatory human microvascular endothelial cells stimulated in vitro with two deuterium-labeled saturated FA (D-FA), i.e., palmitic (D31-PA) and myristic (D27-MA) acids. Cells were measured both by spontaneous and stimulated Raman imaging to extract detailed information about uptaken FA, whereas coherent anti-Stokes Raman scattering and fluorescence imaging showed the global content of FA in cells. Additionally, we employed atomic force microscopy to obtain a morphological image of the cells. The results indicate that the uptake of D-FA in inflamed cells is dependent on their concentration and type. Cells accumulated D-FA when treated with a low concentration, and the effect was more pronounced for D27-MA, in normal cells, but even more so, in inflamed cells. In the case of D31-PA, a slightly increased uptake was observed for inflamed cells when administered at higher concentration. The results provide a better understanding of the EC inflammation and indicate the impact of the pathological state of the EC on their capacity to buffer fat. All the microscopic methods used showed complementarity in the analysis of FA uptake by EC, but each method recognized this process from a different perspective.
Collapse
Affiliation(s)
| | - Anna Pieczara
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Krzysztof Czamara
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Marta Stojak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Ewelina Matuszyk
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Katarzyna Majzner
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland ,Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Krzysztof Brzozowski
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland
| | - Arianna Bresci
- Physics Department, Politecnico di Milano, Piazza Leonardo da Vinci, 32, 20133 Milan, Italy
| | - Dario Polli
- Physics Department, Politecnico di Milano, Piazza Leonardo da Vinci, 32, 20133 Milan, Italy ,Institute for Photonics and Nanotechnology at CNR (CNR-IFN), Piazza Leonardo da Vinci, 32, 20133 Milan, Italy
| | - Malgorzata Baranska
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland ,Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| |
Collapse
|
17
|
Lourenço CF, Laranjinha J. Nitric Oxide Pathways in Neurovascular Coupling Under Normal and Stress Conditions in the Brain: Strategies to Rescue Aberrant Coupling and Improve Cerebral Blood Flow. Front Physiol 2021; 12:729201. [PMID: 34744769 PMCID: PMC8569710 DOI: 10.3389/fphys.2021.729201] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/20/2021] [Indexed: 01/04/2023] Open
Abstract
The brain has impressive energy requirements and paradoxically, very limited energy reserves, implying its huge dependency on continuous blood supply. Aditionally, cerebral blood flow must be dynamically regulated to the areas of increased neuronal activity and thus, of increased metabolic demands. The coupling between neuronal activity and cerebral blood flow (CBF) is supported by a mechanism called neurovascular coupling (NVC). Among the several vasoactive molecules released by glutamatergic activation, nitric oxide (•NO) is recognized to be a key player in the process and essential for the development of the neurovascular response. Classically, •NO is produced in neurons upon the activation of the glutamatergic N-methyl-D-aspartate (NMDA) receptor by the neuronal isoform of nitric oxide synthase and promotes vasodilation by activating soluble guanylate cyclase in the smooth muscle cells of the adjacent arterioles. This pathway is part of a more complex network in which other molecular and cellular intervenients, as well as other sources of •NO, are involved. The elucidation of these interacting mechanisms is fundamental in understanding how the brain manages its energy requirements and how the failure of this process translates into neuronal dysfunction. Here, we aimed to provide an integrated and updated perspective of the role of •NO in the NVC, incorporating the most recent evidence that reinforces its central role in the process from both viewpoints, as a physiological mediator and a pathological stressor. First, we described the glutamate-NMDA receptor-nNOS axis as a central pathway in NVC, then we reviewed the link between the derailment of the NVC and neuronal dysfunction associated with neurodegeneration (with a focus on Alzheimer's disease). We further discussed the role of oxidative stress in the NVC dysfunction, specifically by decreasing the •NO bioavailability and diverting its bioactivity toward cytotoxicity. Finally, we highlighted some strategies targeting the rescue or maintenance of •NO bioavailability that could be explored to mitigate the NVC dysfunction associated with neurodegenerative conditions. In line with this, the potential modulatory effects of dietary nitrate and polyphenols on •NO-dependent NVC, in association with physical exercise, may be used as effective non-pharmacological strategies to promote the •NO bioavailability and to manage NVC dysfunction in neuropathological conditions.
Collapse
Affiliation(s)
- Cátia F Lourenço
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - João Laranjinha
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
18
|
Tan ZX, Dong F, Wu LY, Feng YS, Zhang F. The Beneficial Role of Exercise on Treating Alzheimer's Disease by Inhibiting β-Amyloid Peptide. Mol Neurobiol 2021; 58:5890-5906. [PMID: 34415486 DOI: 10.1007/s12035-021-02514-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is associated with a very large burden on global healthcare systems. Thus, it is imperative to find effective treatments of the disease. One feature of AD is the accumulation of neurotoxic β-amyloid peptide (Aβ). Aβ induces multiple pathological processes that are deleterious to nerve cells. Despite the development of medications that target the reduction of Aβ to treat AD, none has proven to be effective to date. Non-pharmacological interventions, such as physical exercise, are also being studied. The benefits of exercise on AD are widely recognized. Experimental and clinical studies have been performed to verify the role that exercise plays in reducing Aβ deposition to alleviate AD. This paper reviewed the various mechanisms involved in the exercise-induced reduction of Aβ, including the regulation of amyloid precursor protein cleaved proteases, the glymphatic system, brain-blood transport proteins, degrading enzymes and autophagy, which is beneficial to promote exercise therapy as a means of prevention and treatment of AD and indicates that exercise may provide new therapeutic targets for the treatment of AD.
Collapse
Affiliation(s)
- Zi-Xuan Tan
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, People's Republic of China
| | - Lin-Yu Wu
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Ya-Shuo Feng
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, People's Republic of China. .,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050051, People's Republic of China.
| |
Collapse
|
19
|
Querfurth H, Lee HK. Mammalian/mechanistic target of rapamycin (mTOR) complexes in neurodegeneration. Mol Neurodegener 2021; 16:44. [PMID: 34215308 PMCID: PMC8252260 DOI: 10.1186/s13024-021-00428-5] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Novel targets to arrest neurodegeneration in several dementing conditions involving misfolded protein accumulations may be found in the diverse signaling pathways of the Mammalian/mechanistic target of rapamycin (mTOR). As a nutrient sensor, mTOR has important homeostatic functions to regulate energy metabolism and support neuronal growth and plasticity. However, in Alzheimer's disease (AD), mTOR alternately plays important pathogenic roles by inhibiting both insulin signaling and autophagic removal of β-amyloid (Aβ) and phospho-tau (ptau) aggregates. It also plays a role in the cerebrovascular dysfunction of AD. mTOR is a serine/threonine kinase residing at the core in either of two multiprotein complexes termed mTORC1 and mTORC2. Recent data suggest that their balanced actions also have implications for Parkinson's disease (PD) and Huntington's disease (HD), Frontotemporal dementia (FTD) and Amyotrophic Lateral Sclerosis (ALS). Beyond rapamycin; an mTOR inhibitor, there are rapalogs having greater tolerability and micro delivery modes, that hold promise in arresting these age dependent conditions.
Collapse
Affiliation(s)
- Henry Querfurth
- Department of Neurology, Tufts Medical Center, Boston, Massachusetts, USA.
| | - Han-Kyu Lee
- Department of Neurology, Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Biomarkers for evaluating the effects of exercise interventions in patients with MCI or dementia: A systematic review and meta-analysis. Exp Gerontol 2021; 151:111424. [PMID: 34051283 DOI: 10.1016/j.exger.2021.111424] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/13/2021] [Accepted: 05/23/2021] [Indexed: 11/20/2022]
Abstract
OBJECTIVES To summarize the biomarkers for evaluating the effects of exercise interventions in patients with cognitive impairment associated with aging, as well as their responses to exercise interventions. DESIGN A systematic review and meta-analysis METHODS: We systematically searched different electronic database, including PubMed, Cochrane Central Register of Controlled Trials, Embase, Web of Science, PsycINFO, SPORTDiscus up to April 2020. Clinical controlled trials with exercise interventions in patients with cognitive impairment were included. The main outcomes included all the biomarkers used to evaluate the effects of exercise interventions. If data for certain biomarkers was enough (more than 2 studies), meta-analyses would be performed to estimate the effect sizes by calculating the standard mean differences (SMDs) and 95% confidence intervals (CIs). RESULTS Finally, we included 33 articles from 26 trials. The biomarkers included neurotrophic factors, inflammatory factors, oxidative stress markers, neuropathological hallmarks, metabolic biomarkers and genotypes. The meta-analyses indicated that exercise significantly decreased the levels of IL-6 (SMD = -0.45; 95% CI: -0.72, -0.18) and low-density lipoprotein (SMD = -0.26; 95% CI: -0.50, -0.01). Subgroup analyses showed that aerobic exercise also could decrease the levels of TNF-α (SMD = -1.21; 95% CI: -2.29, -0.14). There were some important cognition-related biomarkers which were rarely measured, such as Aβ, tau and IGF-1. CONCLUSION Regular exercise showed positive effects on reducing inflammation and regulating lipid metabolism. But the available evidence is limited and more studies with different exercise interventions should be conducted to test the effects of exercise on other important cognition-related biomarkers in patients with cognitive dysfunction.
Collapse
|
21
|
Pedrinolla A, Magliozzi R, Colosio AL, Danese E, Gelati M, Rossi S, Pogliaghi S, Calabrese M, Muti E, Cè E, Longo S, Esposito F, Lippi G, Schena F, Venturelli M. Repeated passive mobilization to stimulate vascular function in individuals of advanced age who are chronically bedridden. A randomized controlled trial. J Gerontol A Biol Sci Med Sci 2021; 77:588-596. [PMID: 34036337 DOI: 10.1093/gerona/glab148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Vascular dysfunction and associated disorders are major side effects of chronic bed rest, yet passive mobilization as a potential treatment has only been theorized so far. This study investigated the effects of passive mobilization treatment on vascular function in older, chronically bedridden people. METHODS The study sample was 45 chronically bedridden people of advanced age (mean age 87 years; 56% female; mean bed rest 4 years) randomly assigned to a treatment (n=23) or a control group (CTRL, n=22). The treatment group received passive mobilization twice daily (30 min, 5 times/week) for 4 weeks. A kinesiologist performed passive mobilization by passive knee flexion/extension at 1 Hz in one leg (treated leg, T-leg vs ctrl-leg). The CTRL group received routine treatment. The primary outcome was changes in peak blood flow (∆Peak) as measured with the single passive leg movement test (sPLM) at the common femoral artery. RESULTS ∆Peak was increased in both legs in the Treatment group (+90.9 ml/min, p<0.001, in T-leg and +25.7 ml/min, p=0.039 in ctrl-leg). No difference in peak blood flow after routine treatment was found in the CTRL group. CONCLUSION Improvement in vascular function after 4 weeks of passive mobilization was recorded in the treatment group. Passive mobilization may be advantageously included in standard clinical practice as an effective strategy to treat vascular dysfunction in persons with severely limited mobility.
Collapse
Affiliation(s)
- Anna Pedrinolla
- Department of Neuroscience, Biomedicine, and Movement Science, Section of Movement Science, University of Verona, Verona, Italy
| | - Roberta Magliozzi
- Department of Neuroscience, Biomedicine, and Movement Science, Section of Movement Science, University of Verona, Verona, Italy
| | - Alessandro L Colosio
- Department of Neuroscience, Biomedicine, and Movement Science, Section of Movement Science, University of Verona, Verona, Italy
| | - Elisa Danese
- Department of Life and Reproduction Sciences, Laboratory of Clinical Biochemistry, University of Verona, Verona, Italy
| | - Matteo Gelati
- Department of Life and Reproduction Sciences, Laboratory of Clinical Biochemistry, University of Verona, Verona, Italy
| | - Stefania Rossi
- Department of Neuroscience, Biomedicine, and Movement Science, Section of Movement Science, University of Verona, Verona, Italy
| | - Silvia Pogliaghi
- Department of Neuroscience, Biomedicine, and Movement Science, Section of Movement Science, University of Verona, Verona, Italy
| | - Massimiliano Calabrese
- Department of Neuroscience, Biomedicine, and Movement Science, Section of Movement Science, University of Verona, Verona, Italy
| | | | - Emiliano Cè
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy.,IRCSS Galeazzi Orthopaedic Institute, Milano, Italy
| | - Stefano Longo
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Fabio Esposito
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy.,IRCSS Galeazzi Orthopaedic Institute, Milano, Italy
| | - Giuseppe Lippi
- Department of Life and Reproduction Sciences, Laboratory of Clinical Biochemistry, University of Verona, Verona, Italy
| | - Federico Schena
- Department of Neuroscience, Biomedicine, and Movement Science, Section of Movement Science, University of Verona, Verona, Italy
| | - Massimo Venturelli
- Department of Neuroscience, Biomedicine, and Movement Science, Section of Movement Science, University of Verona, Verona, Italy.,Department of Internal Medicine section of Geriatrics, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
22
|
Zhu HY, Hong FF, Yang SL. The Roles of Nitric Oxide Synthase/Nitric Oxide Pathway in the Pathology of Vascular Dementia and Related Therapeutic Approaches. Int J Mol Sci 2021; 22:ijms22094540. [PMID: 33926146 PMCID: PMC8123648 DOI: 10.3390/ijms22094540] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/18/2021] [Accepted: 04/21/2021] [Indexed: 12/16/2022] Open
Abstract
Vascular dementia (VaD) is the second most common form of dementia worldwide. It is caused by cerebrovascular disease, and patients often show severe impairments of advanced cognitive abilities. Nitric oxide synthase (NOS) and nitric oxide (NO) play vital roles in the pathogenesis of VaD. The functions of NO are determined by its concentration and bioavailability, which are regulated by NOS activity. The activities of different NOS subtypes in the brain are partitioned. Pathologically, endothelial NOS is inactivated, which causes insufficient NO production and aggravates oxidative stress before inducing cerebrovascular endothelial dysfunction, while neuronal NOS is overactive and can produce excessive NO to cause neurotoxicity. Meanwhile, inflammation stimulates the massive expression of inducible NOS, which also produces excessive NO and then induces neuroinflammation. The vicious circle of these kinds of damage having impacts on each other finally leads to VaD. This review summarizes the roles of the NOS/NO pathway in the pathology of VaD and also proposes some potential therapeutic methods that target this pathway in the hope of inspiring novel ideas for VaD therapeutic approaches.
Collapse
Affiliation(s)
- Han-Yan Zhu
- Department of Physiology, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China;
- Queen Marry College, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China
| | - Fen-Fang Hong
- Teaching Center, Department of Experimental, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China
- Correspondence: (F.-F.H.); (S.-L.Y.)
| | - Shu-Long Yang
- Department of Physiology, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China;
- Correspondence: (F.-F.H.); (S.-L.Y.)
| |
Collapse
|
23
|
Jiang T, Luo J, Pan X, Zheng H, Yang H, Zhang L, Hu X. Physical exercise modulates the astrocytes polarization, promotes myelin debris clearance and remyelination in chronic cerebral hypoperfusion rats. Life Sci 2021; 278:119526. [PMID: 33894268 DOI: 10.1016/j.lfs.2021.119526] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/01/2021] [Accepted: 04/13/2021] [Indexed: 10/21/2022]
Abstract
AIMS White matter damage is the main pathological feature of chronic cerebral hypoperfusion (CCH) and glial activation is crucial in this process. Physical exercise has protective effects on CCH, but the mechanism is unclear. Therefore, this study focuses on investigating the influence of physical exercise on activated astrocytes polarization and its role in CCH. MAIN METHODS Rats were given wheel running 48 h after 2VO (2 vessel occlusion) surgery. The cognitive function was evaluated by Morris water maze and novel object recognition test. Inflammatory cytokines expressions were detected by ELISA. Astrocytes polarization was analyzed by immunofluorescence. Myelin debris clearance and remyelination were detected by immunofluorescence and transmission electron microscopy. KEY FINDINGS Astrocytes were activated and mainly switched to A1 phenotype in rats 2 and 3 months after 2VO. Myelin debris deposition and limited remyelination can be observed at the corresponding time. Whereas physical exercise can improve the cognitive function of 2VO rats, downregulate the expression of inflammatory factors IL-1α, C1q and TNF, upregulate the release of TGFβ, and promote activated astrocytes transformation from A1 to A2 phenotype. In addition, it can also enhance myelin debris removal and remyelination. SIGNIFICANCE These findings suggest that the benefits of physical exercise on white matter repair and cognition improvement may be related to its regulation of astrocytes polarization, which contributes to myelin debris clearance and effective remyelination in CCH.
Collapse
Affiliation(s)
- Ting Jiang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China; Department of Neurorehabilitation, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Jing Luo
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Xiaona Pan
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Haiqing Zheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Huaichun Yang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Liying Zhang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China.
| | - Xiquan Hu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China.
| |
Collapse
|
24
|
Carvalho J, Borges-Machado F, Barros D, Sampaio A, Marques-Aleixo I, Bohn L, Pizarro A, Teixeira L, Magalhães J, Ribeiro Ó. "Body & Brain": effects of a multicomponent exercise intervention on physical and cognitive function of adults with dementia - study protocol for a quasi-experimental controlled trial. BMC Geriatr 2021; 21:156. [PMID: 33663414 PMCID: PMC7934383 DOI: 10.1186/s12877-021-02104-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Dementia is a leading cause of death and disability that was declared as one of the greatest health and social care challenges of the twenty-first century. Regular physical activity and exercise have been proposed as a non-pharmacological strategy in disease prevention and management. Multicomponent Training (MT) combines aerobic, strength, balance and postural exercises and might be an effective training to improve both functional capacity and cognitive function in individuals with dementia (IwD). Nevertheless, data on the effects of MT in IwD are still limited and the extent to which IwD can retain improvements after an exercise intervention still needs to be elucidated. The aim of "Body & Brain" study is to investigate the effects of a 6-month MT intervention and 3-month detraining on the physical and cognitive function of IwD. Additionally, we aim to explore the impact of this intervention on psychosocial factors and physiologic markers related to dementia. METHODS This study is a quasi-experimental controlled trial using a parallel-group design. The study sample consists of community-dwelling individuals aged ≥60 years who are clinically diagnosed with dementia or major neurocognitive disorder. Participants will be either allocated into the intervention group or the control group. The intervention group will participate in MT biweekly exercise sessions, whereas the control group will receive monthly sessions regarding physical activity and health-related topics for 6 months. The main outcomes will be physical function as measured by the Short Physical Performance Battery (SPPB) and cognitive function evaluated using the Alzheimer Disease Assessment Scale - Cognitive (ADAS-Cog) at baseline, after 6-months and 3-months after the end of intervention. Secondary outcomes will be body composition, physical fitness, daily functionality, quality of life, neuropsychiatric symptoms and caregiver's burden. Cardiovascular, inflammatory and neurotrophic blood-based biomarkers, and arterial stiffness will also be evaluated in subsamples. DISCUSSION If our hypothesis is correct, this project will provide evidence regarding the efficacy of MT training in improving physical and cognitive function and give insights about its impact on novel molecular biomarkers related to dementia. This project may also contribute to provide guidelines on exercise prescription for IwD. TRIAL REGISTRATION ClinicalTrials.gov - identifier number NCT04095962 ; retrospectively registered on 19 September 2019.
Collapse
Affiliation(s)
- Joana Carvalho
- Faculdade de Desporto da Universidade do Porto, Rua Dr. Plácido Costa 91, 4200-450, Porto, Portugal
- CIAFEL, Centro de Investigação em Atividade Física, Saúde e Lazer, Universidade do Porto, Rua Dr. Plácido Costa 91, 4200-450, Porto, Portugal
| | - Flávia Borges-Machado
- CIAFEL, Centro de Investigação em Atividade Física, Saúde e Lazer, Universidade do Porto, Rua Dr. Plácido Costa 91, 4200-450, Porto, Portugal.
| | - Duarte Barros
- CIAFEL, Centro de Investigação em Atividade Física, Saúde e Lazer, Universidade do Porto, Rua Dr. Plácido Costa 91, 4200-450, Porto, Portugal
| | - Arnaldina Sampaio
- CIAFEL, Centro de Investigação em Atividade Física, Saúde e Lazer, Universidade do Porto, Rua Dr. Plácido Costa 91, 4200-450, Porto, Portugal
| | - Inês Marques-Aleixo
- CIAFEL, Centro de Investigação em Atividade Física, Saúde e Lazer, Universidade do Porto, Rua Dr. Plácido Costa 91, 4200-450, Porto, Portugal
- Faculdade de Educação Física e Desporto, Universidade Lusófona, Rua de Augusto Rosa 24, 4000-098, Porto, Portugal
| | - Lucimere Bohn
- CIAFEL, Centro de Investigação em Atividade Física, Saúde e Lazer, Universidade do Porto, Rua Dr. Plácido Costa 91, 4200-450, Porto, Portugal
- Faculdade de Educação Física e Desporto, Universidade Lusófona, Rua de Augusto Rosa 24, 4000-098, Porto, Portugal
| | - Andreia Pizarro
- CIAFEL, Centro de Investigação em Atividade Física, Saúde e Lazer, Universidade do Porto, Rua Dr. Plácido Costa 91, 4200-450, Porto, Portugal
| | - Laetitia Teixeira
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - José Magalhães
- Faculdade de Desporto da Universidade do Porto, Rua Dr. Plácido Costa 91, 4200-450, Porto, Portugal
- CIAFEL, Centro de Investigação em Atividade Física, Saúde e Lazer, Universidade do Porto, Rua Dr. Plácido Costa 91, 4200-450, Porto, Portugal
| | - Óscar Ribeiro
- CINTESIS, Centro de Investigação em Tecnologias e Serviços de Saúde, Departamento de Educação e Psicologia, Universidade de Aveiro - Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
25
|
Bliss ES, Wong RHX, Howe PRC, Mills DE. Benefits of exercise training on cerebrovascular and cognitive function in ageing. J Cereb Blood Flow Metab 2021; 41:447-470. [PMID: 32954902 PMCID: PMC7907999 DOI: 10.1177/0271678x20957807] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Derangements in cerebrovascular structure and function can impair cognitive performance throughout ageing and in cardiometabolic disease states, thus increasing dementia risk. Modifiable lifestyle factors that cause a decline in cardiometabolic health, such as physical inactivity, exacerbate these changes beyond those that are associated with normal ageing. The purpose of this review was to examine cerebrovascular, cognitive and neuroanatomical adaptations to ageing and the potential benefits of exercise training on these outcomes in adults 50 years or older. We systematically searched for cross-sectional or intervention studies that included exercise (aerobic, resistance or multimodal) and its effect on cerebrovascular function, cognition and neuroanatomical adaptations in this age demographic. The included studies were tabulated and described narratively. Aerobic exercise training was the predominant focus of the studies identified; there were limited studies exploring the effects of resistance exercise training and multimodal training on cerebrovascular function and cognition. Collectively, the evidence indicated that exercise can improve cerebrovascular function, cognition and neuroplasticity through areas of the brain associated with executive function and memory in adults 50 years or older, irrespective of their health status. However, more research is required to ascertain the mechanisms of action.
Collapse
Affiliation(s)
- Edward S Bliss
- Respiratory and Exercise Physiology Research Group, School of
Health and Wellbeing, University of Southern Queensland, Ipswich, Queensland,
Australia
- Edward S Bliss, School of Health and
Wellbeing, University of Southern Queensland, Toowoomba Campus, West St,
Toowoomba QLD 4350, Australia.
| | - Rachel HX Wong
- Centre for Health, Informatics, and Economic Research, Institute
for Resilient Regions, University of Southern Queensland, Ipswich, Queensland,
Australia
- School of Biomedical Sciences and Pharmacy, Clinical Nutrition
Research Centre, University of Newcastle, Callaghan, New South Wales,
Australia
| | - Peter RC Howe
- Centre for Health, Informatics, and Economic Research, Institute
for Resilient Regions, University of Southern Queensland, Ipswich, Queensland,
Australia
- School of Biomedical Sciences and Pharmacy, Clinical Nutrition
Research Centre, University of Newcastle, Callaghan, New South Wales,
Australia
- Allied Health and Human Performance, University of South
Australia, Adelaide, South Australia, Australia
| | - Dean E Mills
- Respiratory and Exercise Physiology Research Group, School of
Health and Wellbeing, University of Southern Queensland, Ipswich, Queensland,
Australia
| |
Collapse
|
26
|
Adams JA, Uryash A, Lopez JR, Sackner MA. The Endothelium as a Therapeutic Target in Diabetes: A Narrative Review and Perspective. Front Physiol 2021; 12:638491. [PMID: 33708143 PMCID: PMC7940370 DOI: 10.3389/fphys.2021.638491] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 01/29/2021] [Indexed: 12/18/2022] Open
Abstract
Diabetes has reached worldwide epidemic proportions, and threatens to be a significant economic burden to both patients and healthcare systems, and an important driver of cardiovascular mortality and morbidity. Improvement in lifestyle interventions (which includes increase in physical activity via exercise) can reduce diabetes and cardiovascular disease mortality and morbidity. Encouraging a population to increase physical activity and exercise is not a simple feat particularly in individuals with co-morbidities (obesity, heart disease, stroke, peripheral vascular disease, and those with cognitive and physical limitations). Translation of the physiological benefits of exercise within that vulnerable population would be an important step for improving physical activity goals and a stopgap measure to exercise. In large part many of the beneficial effects of exercise are due to the introduction of pulsatile shear stress (PSS) to the vascular endothelium. PSS is a well-known stimulus for endothelial homeostasis, and induction of a myriad of pathways which include vasoreactivity, paracrine/endocrine function, fibrinolysis, inflammation, barrier function, and vessel growth and formation. The endothelial cell mediates the balance between vasoconstriction and relaxation via the major vasodilator endothelial derived nitric oxide (eNO). eNO is critical for vasorelaxation, increasing blood flow, and an important signaling molecule that downregulates the inflammatory cascade. A salient feature of diabetes, is endothelial dysfunction which is characterized by a reduction of the bioavailability of vasodilators, particularly nitric oxide (NO). Cellular derangements in diabetes are also related to dysregulation in Ca2+ handling with increased intracellular Ca2+overload, and oxidative stress. PSS increases eNO bioavailability, reduces inflammatory phenotype, decreases intracellular Ca2+ overload, and increases antioxidant capacity. This narrative review and perspective will outline four methods to non-invasively increase PSS; Exercise (the prototype for increasing PSS), Enhanced External Counterpulsation (EECP), Whole Body Vibration (WBV), Passive Simulated Jogging and its predicate device Whole Body Periodic Acceleration, and will discuss current knowledge on their use in diabetes.
Collapse
Affiliation(s)
- Jose A Adams
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, United States
| | - Arkady Uryash
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, United States
| | - Jose R Lopez
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL, United States
| | - Marvin A Sackner
- Department of Medicine, Mount Sinai Medical Center, Miami Beach, FL, United States
| |
Collapse
|
27
|
Vergoossen LWM, Jansen JFA, de Jong JJA, Stehouwer CDA, Schaper NC, Savelberg HHCM, Koster A, Backes WH, Schram MT. Association of physical activity and sedentary time with structural brain networks-The Maastricht Study. GeroScience 2021; 43:239-252. [PMID: 33034792 PMCID: PMC8050169 DOI: 10.1007/s11357-020-00276-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/22/2020] [Indexed: 12/25/2022] Open
Abstract
We assessed whether objectively measured low- and high-intensity physical activity (LPA and HPA) and sedentary time (ST) were associated with white matter connectivity, both throughout the whole brain and in brain regions involved in motor function. In the large population-based Maastricht Study (n = 1715, age 59.6 ± 8.1 (mean ± standard deviation) years, and 48% women), the amounts of LPA, HPA, and ST were objectively measured during 7 days by an activPAL accelerometer. In addition, using 3T structural and diffusion MRI, we calculated whole brain node degree and node degree of the basal ganglia and primary motor cortex. Multivariable linear regression analysis was performed, and we report standardized regression coefficients (stβ) adjusted for age, sex, education level, wake time, diabetes status, BMI, office systolic blood pressure, antihypertensive medication, total-cholesterol-to-HDL-cholesterol ratio, lipid-modifying medication, alcohol use, smoking status, and history of cardiovascular disease. Lower HPA was associated with lower whole brain node degree after full adjustment (stβ [95%CI] = - 0.062 [- 0.101, - 0.013]; p = 0.014), whereas lower LPA (stβ [95%CI] = - 0.013 [- 0.061, 0.034]; p = 0.580) and higher ST (stβ [95%CI] = - 0.030 [- 0.081, 0.021]; p = 0.250) was not. In addition, lower HPA was associated with lower node degree of the basal ganglia after full adjustment (stβ [95%CI] = - 0.070 [- 0.121, - 0.018]; p = 0.009). Objectively measured lower HPA, but not lower LPA and higher ST, was associated with lower whole brain node degree and node degree in specific brain regions highly specialized in motor function. Further research is needed to establish whether more HPA may preserve structural brain connectivity.
Collapse
Affiliation(s)
- Laura W M Vergoossen
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- School for Cardiovascular Disease (CARIM), Maastricht University, Maastricht, The Netherlands
| | - J F A Jansen
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - J J A de Jong
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - C D A Stehouwer
- School for Cardiovascular Disease (CARIM), Maastricht University, Maastricht, The Netherlands
- Department of Internal Medicine, Maastricht University Medical Center+, PO Box 5800, AZ, 6202, Maastricht, The Netherlands
| | - N C Schaper
- School for Cardiovascular Disease (CARIM), Maastricht University, Maastricht, The Netherlands
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
- Department of Internal Medicine, Maastricht University Medical Center+, PO Box 5800, AZ, 6202, Maastricht, The Netherlands
| | - H H C M Savelberg
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - A Koster
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
- Department of Social Medicine, Maastricht University, Maastricht, The Netherlands
| | - W H Backes
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- School for Cardiovascular Disease (CARIM), Maastricht University, Maastricht, The Netherlands
| | - M T Schram
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands.
- School for Cardiovascular Disease (CARIM), Maastricht University, Maastricht, The Netherlands.
- Heart and Vascular Centre, Maastricht University Medical Center+, Maastricht, the Netherlands.
- Department of Internal Medicine, Maastricht University Medical Center+, PO Box 5800, AZ, 6202, Maastricht, The Netherlands.
| |
Collapse
|
28
|
Boltze J, Aronowski JA, Badaut J, Buckwalter MS, Caleo M, Chopp M, Dave KR, Didwischus N, Dijkhuizen RM, Doeppner TR, Dreier JP, Fouad K, Gelderblom M, Gertz K, Golubczyk D, Gregson BA, Hamel E, Hanley DF, Härtig W, Hummel FC, Ikhsan M, Janowski M, Jolkkonen J, Karuppagounder SS, Keep RF, Koerte IK, Kokaia Z, Li P, Liu F, Lizasoain I, Ludewig P, Metz GAS, Montagne A, Obenaus A, Palumbo A, Pearl M, Perez-Pinzon M, Planas AM, Plesnila N, Raval AP, Rueger MA, Sansing LH, Sohrabji F, Stagg CJ, Stetler RA, Stowe AM, Sun D, Taguchi A, Tanter M, Vay SU, Vemuganti R, Vivien D, Walczak P, Wang J, Xiong Y, Zille M. New Mechanistic Insights, Novel Treatment Paradigms, and Clinical Progress in Cerebrovascular Diseases. Front Aging Neurosci 2021; 13:623751. [PMID: 33584250 PMCID: PMC7876251 DOI: 10.3389/fnagi.2021.623751] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/04/2021] [Indexed: 12/13/2022] Open
Abstract
The past decade has brought tremendous progress in diagnostic and therapeutic options for cerebrovascular diseases as exemplified by the advent of thrombectomy in ischemic stroke, benefitting a steeply increasing number of stroke patients and potentially paving the way for a renaissance of neuroprotectants. Progress in basic science has been equally impressive. Based on a deeper understanding of pathomechanisms underlying cerebrovascular diseases, new therapeutic targets have been identified and novel treatment strategies such as pre- and post-conditioning methods were developed. Moreover, translationally relevant aspects are increasingly recognized in basic science studies, which is believed to increase their predictive value and the relevance of obtained findings for clinical application.This review reports key results from some of the most remarkable and encouraging achievements in neurovascular research that have been reported at the 10th International Symposium on Neuroprotection and Neurorepair. Basic science topics discussed herein focus on aspects such as neuroinflammation, extracellular vesicles, and the role of sex and age on stroke recovery. Translational reports highlighted endovascular techniques and targeted delivery methods, neurorehabilitation, advanced functional testing approaches for experimental studies, pre-and post-conditioning approaches as well as novel imaging and treatment strategies. Beyond ischemic stroke, particular emphasis was given on activities in the fields of traumatic brain injury and cerebral hemorrhage in which promising preclinical and clinical results have been reported. Although the number of neutral outcomes in clinical trials is still remarkably high when targeting cerebrovascular diseases, we begin to evidence stepwise but continuous progress towards novel treatment options. Advances in preclinical and translational research as reported herein are believed to have formed a solid foundation for this progress.
Collapse
Affiliation(s)
- Johannes Boltze
- School of Life Sciences, University of Warwick, Warwick, United Kingdom
| | - Jaroslaw A. Aronowski
- Institute for Stroke and Cerebrovascular Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jerome Badaut
- NRS UMR 5287, INCIA, Brain Molecular Imaging Team, University of Bordeaux, Bordeaux cedex, France
| | - Marion S. Buckwalter
- Departments of Neurology and Neurological Sciences, and Neurosurgery, Wu Tsai Neurosciences Institute, Stanford School of Medicine, Stanford, CA, United States
| | - Mateo Caleo
- Neuroscience Institute, National Research Council, Pisa, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
- Department of Physics, Oakland University, Rochester, MI, United States
| | - Kunjan R. Dave
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Nadine Didwischus
- School of Life Sciences, University of Warwick, Warwick, United Kingdom
| | - Rick M. Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Thorsten R. Doeppner
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Jens P. Dreier
- Department of Neurology, Center for Stroke Research Berlin, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
| | - Karim Fouad
- Faculty of Rehabilitation Medicine and Institute for Neuroscience and Mental Health, University of Alberta, Edmonton, AB, Canada
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karen Gertz
- Department of Neurology, Center for Stroke Research Berlin, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Dominika Golubczyk
- Department of Neurosurgery, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Barbara A. Gregson
- Neurosurgical Trials Group, Institute of Neuroscience, The University of Newcastle upon Tyne, Newcastle upon Tyne, United Kingdom
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Daniel F. Hanley
- Division of Brain Injury Outcomes, Johns Hopkins University, Baltimore, MD, United States
| | - Wolfgang Härtig
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Friedhelm C. Hummel
- Clinical Neuroengineering, Center for Neuroprosthetics and Brain Mind Institute, Swiss Federal Institute of Technology Valais, Clinique Romande de Réadaptation, Sion, Switzerland
- Clinical Neuroscience, University of Geneva Medical School, Geneva, Switzerland
| | - Maulana Ikhsan
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
- Fraunhofer Research Institution for Marine Biotechnology and Cell Technology, Lübeck, Germany
- Institute for Medical and Marine Biotechnology, University of Lübeck, Lübeck, Germany
| | - Miroslaw Janowski
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD, United States
| | - Jukka Jolkkonen
- Department of Neurology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Saravanan S. Karuppagounder
- Burke Neurological Institute, White Plains, NY, United States
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Richard F. Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, United States
| | - Inga K. Koerte
- Psychiatric Neuroimaging Laboratory, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Department of Child and Adolescent Psychiatry, Psychosomatic, and Psychotherapy, Ludwig Maximilians University, Munich, Germany
| | - Zaal Kokaia
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Peiying Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Fudong Liu
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, United States
| | - Ignacio Lizasoain
- Unidad de Investigación Neurovascular, Departamento Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Madrid, Spain
| | - Peter Ludewig
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerlinde A. S. Metz
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Axel Montagne
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Andre Obenaus
- Department of Pediatrics, University of California, Irvine, Irvine, CA, United States
| | - Alex Palumbo
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
- Fraunhofer Research Institution for Marine Biotechnology and Cell Technology, Lübeck, Germany
- Institute for Medical and Marine Biotechnology, University of Lübeck, Lübeck, Germany
| | - Monica Pearl
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Miguel Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Anna M. Planas
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Àrea de Neurociències, Barcelona, Spain
- Department d’Isquèmia Cerebral I Neurodegeneració, Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Munich University Hospital, Munich, Germany
- Munich Cluster of Systems Neurology (Synergy), Munich, Germany
| | - Ami P. Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Maria A. Rueger
- Faculty of Medicine and University Hospital, Department of Neurology, University of Cologne, Cologne, Germany
| | - Lauren H. Sansing
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - Farida Sohrabji
- Women’s Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M College of Medicine, Bryan, TX, United States
| | - Charlotte J. Stagg
- Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
- MRC Brain Network Dynamics Unit, University of Oxford, Oxford, United Kingdom
| | - R. Anne Stetler
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ann M. Stowe
- Department of Neurology and Neurotherapeutics, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, United States
| | - Dandan Sun
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, PA, United States
| | - Akihiko Taguchi
- Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Kobe, Japan
| | - Mickael Tanter
- Institute of Physics for Medicine Paris, INSERM U1273, ESPCI Paris, CNRS FRE 2031, PSL University, Paris, France
| | - Sabine U. Vay
- Faculty of Medicine and University Hospital, Department of Neurology, University of Cologne, Cologne, Germany
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, United States
| | - Denis Vivien
- UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging for Neurological Disorders (PhIND), Normandy University, Caen, France
- CHU Caen, Clinical Research Department, CHU de Caen Côte de Nacre, Caen, France
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD, United States
| | - Jian Wang
- Department of Human Anatomy, College of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ye Xiong
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, United States
| | - Marietta Zille
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
- Fraunhofer Research Institution for Marine Biotechnology and Cell Technology, Lübeck, Germany
- Institute for Medical and Marine Biotechnology, University of Lübeck, Lübeck, Germany
| |
Collapse
|
29
|
Zhang J, Sun P, Zhou C, Zhang X, Ma F, Xu Y, Hamblin MH, Yin K. Regulatory microRNAs and vascular cognitive impairment and dementia. CNS Neurosci Ther 2020; 26:1207-1218. [PMID: 33459504 PMCID: PMC7702235 DOI: 10.1111/cns.13472] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular cognitive impairment and dementia (VCID) is defined as a progressive dementia disease related to cerebrovascular injury and often occurs in aged populations. Despite decades of research, effective treatment for VCID is still absent. The pathological processes of VCID are mediated by the molecular mechanisms that are partly modulated at the post-transcriptional level. As small endogenous non-coding RNAs, microRNAs (miRs) can regulate target gene expression through post-transcriptional gene silencing. miRs have been reported to play an important role in the pathology of VCID and have recently been suggested as potential novel pharmacological targets for the development of new diagnosis and treatment strategies in VCID. In this review, we summarize the current understanding of VCID, the possible role of miRs in the regulation of VCID and attempt to envision future therapeutic strategies. Since manipulation of miR levels by either pharmacological or genetic approaches has shown therapeutic effects in experimental VCID models, we also emphasize the potential therapeutic value of miRs in clinical settings.
Collapse
Affiliation(s)
- Jing Zhang
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Ping Sun
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Chao Zhou
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Xuejing Zhang
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Feifei Ma
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Yang Xu
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Milton H. Hamblin
- Department of PharmacologyTulane University School of MedicineNew OrleansLAUSA
| | - Ke‐Jie Yin
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
- Geriatric ResearchEducation and Clinical CenterVeterans Affairs Pittsburgh Healthcare SystemPittsburghPAUSA
| |
Collapse
|
30
|
Pedrinolla A, Venturelli M, Fonte C, Tamburin S, Di Baldassarre A, Naro F, Varalta V, Giuriato G, Ghinassi B, Muti E, Smania N, Schena F. Exercise training improves vascular function in patients with Alzheimer's disease. Eur J Appl Physiol 2020; 120:2233-2245. [PMID: 32728820 PMCID: PMC7502067 DOI: 10.1007/s00421-020-04447-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/19/2020] [Indexed: 11/25/2022]
Abstract
PURPOSE Vascular dysfunction has been demonstrated in patients with Alzheimer's disease (AD). Exercise is known to positively affect vascular function. Thus, the aim of our study was to investigate exercise-induced effects on vascular function in AD. METHODS Thirty-nine patients with AD (79 ± 8 years) were recruited and randomly assigned to exercise training (EX, n = 20) or control group (CTRL, n = 19). All subjects performed 72 treatment sessions (90 min, 3 t/w). EX included moderate-high-intensity aerobic and strength training. CTRL included cognitive stimuli (visual, verbal, auditive). Before and after the 6-month treatment, the vascular function was measured by passive-leg movement test (PLM, calculating the variation in blood flow: ∆peak; and area under the curve: AUC) tests, and flow-mediated dilation (FMD, %). A blood sample was analyzed for vascular endothelial growth factor (VEGF). Arterial blood flow (BF) and shear rate (SR) were measured during EX and CTRL during a typical treatment session. RESULTS EX group has increased FMD% (+ 3.725%, p < 0.001), PLM ∆peak (+ 99.056 ml/min, p = 0.004), AUC (+ 37.359AU, p = 0.037) and VEGF (+ 8.825 pg/ml, p = 0.004). In the CTRL group, no difference between pre- and post-treatment was found for any variable. Increase in BF and SR was demonstrated during EX (BF + 123%, p < 0.05; SR + 134%, p < 0.05), but not during CTRL treatment. CONCLUSION Exercise training improves peripheral vascular function in AD. These ameliorations may be due to the repetitive increase in SR during exercise which triggers NO and VEGF upregulation. This approach might be included in standard AD clinical practice as an effective strategy to treat vascular dysfunction in this population.
Collapse
Affiliation(s)
- Anna Pedrinolla
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Via Casorati 43, 37131, Verona, Italy
| | - Massimo Venturelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Via Casorati 43, 37131, Verona, Italy.
- Department of Internal Medicine, University of Utah, Salt Lake, Utah, USA.
| | - Cristina Fonte
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Via Casorati 43, 37131, Verona, Italy
- Department of Neurosciences, Biomedicine and Movement Sciences, Neuromotor and Cognitive Rehabilitation Research Centre, University of Verona, Verona, Italy
| | - Stefano Tamburin
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Via Casorati 43, 37131, Verona, Italy
| | - Angela Di Baldassarre
- Department of Medicine and Aging Sciences, University G. D'Annunzio, Chieti-Pescara, Chieti, Italy
| | - Fabio Naro
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Science, Rome, Italy
| | - Valentina Varalta
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Via Casorati 43, 37131, Verona, Italy
- Department of Neurosciences, Biomedicine and Movement Sciences, Neuromotor and Cognitive Rehabilitation Research Centre, University of Verona, Verona, Italy
| | - Gaia Giuriato
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Via Casorati 43, 37131, Verona, Italy
| | - Barbara Ghinassi
- Department of Medicine and Aging Sciences, University G. D'Annunzio, Chieti-Pescara, Chieti, Italy
| | | | - Nicola Smania
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Via Casorati 43, 37131, Verona, Italy
- Department of Neurosciences, Biomedicine and Movement Sciences, Neuromotor and Cognitive Rehabilitation Research Centre, University of Verona, Verona, Italy
| | - Federico Schena
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Via Casorati 43, 37131, Verona, Italy
| |
Collapse
|
31
|
Chandran R, Li W, Ahmed HA, Dong G, Ward RA, He L, Doueiry C, Ergul A. Diabetic rats are more susceptible to cognitive decline in a model of microemboli-mediated vascular contributions to cognitive impairment and dementia. Brain Res 2020; 1749:147132. [PMID: 33002484 DOI: 10.1016/j.brainres.2020.147132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/26/2020] [Accepted: 09/15/2020] [Indexed: 12/17/2022]
Abstract
Vascular disease plays an important role in all kinds of cognitive impairment and dementia. Diabetes increases the risk of vascular disease and dementia. However, it is not clear how existing vascular disease in the brain accelerates the development of small vessel disease and promotes cognitive dysfunction in diabetes. We used microemboli (ME) injection model in the current study to test the hypothesis that cerebrovascular dysfunction in diabetes facilitates entrapment of ME leading to inflammation and cognitive decline. We investigated cognitive function, axonal/white matter (WM) changes, neurovascular coupling, and microglial activation in control and diabetic male and female Wistar rats subjected to sham or low/high dose ME injection. Diabetic male animals had cognitive deficits, WM demyelination and greater microglial activation than the control animals even at baseline. Functional hyperemia gradually declined in diabetic male animals after ME injection. Both low and high ME injection worsened WM damage and increased microglial activation in diabetic male and female animals. Low ME did not cause cognitive decline in controls, while promoting learning/memory deficits in diabetic female rats and no further decline in diabetic male animals. High ME led to cognitive decline in control male rats and exacerbated the deficits in diabetic cohort. These results suggest that the existing cerebrovascular dysfunction in diabetes may facilitate ME-mediated demyelination leading to cognitive decline. It is important to integrate comorbidities/sex as a biological variable into experimental models for the development of preventive or therapeutic targets.
Collapse
Affiliation(s)
- Raghavendar Chandran
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Weiguo Li
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States.
| | - Heba A Ahmed
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Guangkuo Dong
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA, United States
| | - Rebecca A Ward
- Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Lianying He
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Caren Doueiry
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Adviye Ergul
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, United States
| |
Collapse
|
32
|
Kandasamy M, Anusuyadevi M, Aigner KM, Unger MS, Kniewallner KM, de Sousa DMB, Altendorfer B, Mrowetz H, Bogdahn U, Aigner L. TGF-β Signaling: A Therapeutic Target to Reinstate Regenerative Plasticity in Vascular Dementia? Aging Dis 2020; 11:828-850. [PMID: 32765949 PMCID: PMC7390515 DOI: 10.14336/ad.2020.0222] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/22/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular dementia (VaD) is the second leading form of memory loss after Alzheimer's disease (AD). Currently, there is no cure available. The etiology, pathophysiology and clinical manifestations of VaD are extremely heterogeneous, but the impaired cerebral blood flow (CBF) represents a common denominator of VaD. The latter might be the result of atherosclerosis, amyloid angiopathy, microbleeding and micro-strokes, together causing blood-brain barrier (BBB) dysfunction and vessel leakage, collectively originating from the consequence of hypertension, one of the main risk factors for VaD. At the histopathological level, VaD displays abnormal vascular remodeling, endothelial cell death, string vessel formation, pericyte responses, fibrosis, astrogliosis, sclerosis, microglia activation, neuroinflammation, demyelination, white matter lesions, deprivation of synapses and neuronal loss. The transforming growth factor (TGF) β has been identified as one of the key molecular factors involved in the aforementioned various pathological aspects. Thus, targeting TGF-β signaling in the brain might be a promising therapeutic strategy to mitigate vascular pathology and improve cognitive functions in patients with VaD. This review revisits the recent understanding of the role of TGF-β in VaD and associated pathological hallmarks. It further explores the potential to modulate certain aspects of VaD pathology by targeting TGF-β signaling.
Collapse
Affiliation(s)
- Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India.
- Faculty Recharge Programme, University Grants Commission (UGC-FRP), New Delhi, India.
| | - Muthuswamy Anusuyadevi
- Molecular Gerontology Group, Department of Biochemistry, School of Life Sciences, Bharathidhasan University, Tiruchirappalli, Tamil Nadu, India.
| | - Kiera M Aigner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Michael S Unger
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Kathrin M Kniewallner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Diana M Bessa de Sousa
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Barbara Altendorfer
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Heike Mrowetz
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Ulrich Bogdahn
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
- Velvio GmbH, Regensburg, Germany.
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
33
|
Uddin MS, Rahman MA, Kabir MT, Behl T, Mathew B, Perveen A, Barreto GE, Bin-Jumah MN, Abdel-Daim MM, Ashraf GM. Multifarious roles of mTOR signaling in cognitive aging and cerebrovascular dysfunction of Alzheimer's disease. IUBMB Life 2020; 72:1843-1855. [PMID: 32472959 DOI: 10.1002/iub.2324] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/10/2020] [Accepted: 05/13/2020] [Indexed: 12/19/2022]
Abstract
Age-related cognitive failure is a main devastating incident affecting even healthy people. Alzheimer's disease (AD) is the utmost common form of dementia among the geriatric community. In the pathogenesis of AD, cerebrovascular dysfunction is revealed before the beginning of the cognitive decline. Mounting proof shows a precarious impact of cerebrovascular dysregulation in the development of AD pathology. Recent studies document that the mammalian target of rapamycin (mTOR) acts as a crucial effector of cerebrovascular dysregulation in AD. The mTOR contributes to brain vascular dysfunction and subsequence cerebral blood flow deficits as well as cognitive impairment. Furthermore, mTOR causes the blood-brain barrier (BBB) breakdown in AD models. Inhibition of mTOR hyperactivity protects the BBB integrity in AD. Furthermore, mTOR drives cognitive defect and cerebrovascular dysfunction, which are greatly prevalent in AD, but the central molecular mechanisms underlying these alterations are obscure. This review represents the crucial and current research findings regarding the role of mTOR signaling in cognitive aging and cerebrovascular dysfunction in the pathogenesis of AD.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Md Ataur Rahman
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | | | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Patiala, India
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - May N Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mohamed M Abdel-Daim
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia.,Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
34
|
Hong J, Hong SG, Lee J, Park JY, Eriksen JL, Rooney BV, Park Y. Exercise training ameliorates cerebrovascular dysfunction in a murine model of Alzheimer's disease: role of the P2Y2 receptor and endoplasmic reticulum stress. Am J Physiol Heart Circ Physiol 2020; 318:H1559-H1569. [PMID: 32383993 DOI: 10.1152/ajpheart.00129.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cerebrovascular dysfunction is a critical risk factor for the pathogenesis of Alzheimer's disease (AD). The purinergic P2Y2 receptor and endoplasmic reticulum (ER) stress are tightly associated with vascular dysfunction and the pathogenesis of AD. However, the protective effects of exercise training on P2Y2 receptor- and ER stress-associated cerebrovascular dysfunction in AD are mostly unknown. Control (C57BL/6, CON) and AD (APP/PS1dE9, AD) mice underwent treadmill exercise training (EX). 2-MeS-ATP-induced dose-dependent vasoreactivity was determined by using a pressurized posterior cerebral artery (PCA) from 10-12-mo-old mice. Human brain microvascular endothelial cells (HBMECs) were exposed to laminar shear stress (LSS) at 20 dyn/cm2 for 30 min, 2 h, and 24 h. The expression of P2Y2 receptors, endothelial nitric oxide synthase (eNOS), and ER stress signaling were quantified by Western blot analysis. Notably, exercise converted ATP-induced vasoconstriction in the PCA from AD mice to vasodilation in AD+EX mice to a degree commensurate to the vascular reactivity observed in CON mice. Exercise reduced the expression of amyloid peptide precursor (APP) and increased the P2Y2 receptor and Akt/eNOS expression in AD mice brain. Mechanistically, LSS increased the expression of both P2Y2 receptor and eNOS protein in HBMECs, but these increases were blunted by a P2Y2 receptor antagonist in HBMECs. Exercise also reduced the expression of aberrant ER stress markers p-IRE1, p/t-eIF2α, and CHOP, as well as Bax/Bcl-2, in AD mice brain. Collectively, our results demonstrate for the first time that exercise mitigates cerebrovascular dysfunction in AD through modulating P2Y2 receptor- and ER stress-dependent endothelial dysfunction.NEW & NOTEWORTHY A limited study has investigated whether exercise training can improve cerebrovascular function in Alzheimer's disease. The novel findings of the study are that exercise training improves cerebrovascular dysfunction through enhancing P2Y2 receptor-mediated eNOS signaling and reducing ER stress-associated pathways in AD. These data suggest that exercise training, which regulates P2Y2 receptor and ER stress in AD brain, is a potential therapeutic strategy for Alzheimer's disease.
Collapse
Affiliation(s)
- Junyoung Hong
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, Houston, Texas
| | - Soon-Gook Hong
- Department of Kinesiology and Cardiovascular Research Center, Temple University, Philadelphia, Pennsylvania
| | - Jonghae Lee
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, Houston, Texas
| | - Joon-Young Park
- Department of Kinesiology and Cardiovascular Research Center, Temple University, Philadelphia, Pennsylvania
| | - Jason L Eriksen
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas
| | - Bridgette V Rooney
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, Houston, Texas.,Geocontrol Systems, Incorporated, Johnson Space Center, National Aeronautics and Space Administration, Houston, Texas
| | - Yoonjung Park
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, Houston, Texas
| |
Collapse
|
35
|
de Montgolfier O, Thorin-Trescases N, Thorin E. Pathological Continuum From the Rise in Pulse Pressure to Impaired Neurovascular Coupling and Cognitive Decline. Am J Hypertens 2020; 33:375-390. [PMID: 32202623 PMCID: PMC7188799 DOI: 10.1093/ajh/hpaa001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/11/2019] [Accepted: 03/03/2020] [Indexed: 12/12/2022] Open
Abstract
The "biomechanical hypothesis" stipulates that with aging, the cumulative mechanical damages to the cerebral microvasculature, magnified by risk factors for vascular diseases, contribute to a breach in cerebral homeostasis producing neuronal losses. In other words, vascular dysfunction affects brain structure and function, and leads to cognitive failure. This is gathered under the term Vascular Cognitive Impairment and Dementia (VCID). One of the main culprits in the occurrence of cognitive decline could be the inevitable rise in arterial pulse pressure due to the age-dependent stiffening of large conductance arteries like the carotids, which in turn, could accentuate the penetration of the pulse pressure wave deeper into the fragile microvasculature of the brain and damage it. In this review, we will discuss how and why the vascular and brain cells communicate and are interdependent, describe the deleterious impact of a vascular dysfunction on brain function in various neurodegenerative diseases and even of psychiatric disorders, and the potential chronic deleterious effects of the pulsatile blood pressure on the cerebral microcirculation. We will also briefly review data from antihypertensive clinical trial aiming at improving or delaying dementia. Finally, we will debate how the aging process, starting early in life, could determine our sensitivity to risk factors for vascular diseases, including cerebral diseases, and the trajectory to VCID.
Collapse
Affiliation(s)
- Olivia de Montgolfier
- Faculty of Medicine, Department of Pharmacology and Physiology, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | | | - Eric Thorin
- Faculty of Medicine, Department of Pharmacology and Physiology, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montreal, Quebec, Canada
- Correspondence: Eric Thorin ()
| |
Collapse
|
36
|
Luca M, Luca A. Oxidative Stress-Related Endothelial Damage in Vascular Depression and Vascular Cognitive Impairment: Beneficial Effects of Aerobic Physical Exercise. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8067045. [PMID: 31929857 PMCID: PMC6939429 DOI: 10.1155/2019/8067045] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 12/11/2019] [Indexed: 01/08/2023]
Abstract
Oxidative stress- (OS-) related endothelial damage is involved in the occurrence and progression of several disorders, such as vascular depression and dementia. It has been reported that moderate, aerobic, physical exercise could reduce OS and inflammation, thus limiting the cardiovascular risk factors while improving endothelial homeostasis, mood, and cognition. In this review, we will discuss about the role of OS and OS-related endothelial damage in vascular depression and vascular cognitive impairment. Then, we will comment on the effects of physical exercise on both disorders.
Collapse
Affiliation(s)
- Maria Luca
- Department of Medical, Surgical Sciences and Advanced Technologies “GF Ingrassia”, University of Catania, Italy
| | - Antonina Luca
- Department of Medical, Surgical Sciences and Advanced Technologies “GF Ingrassia”, University of Catania, Italy
| |
Collapse
|
37
|
Trigiani LJ, Royea J, Tong XK, Hamel E. Comparative benefits of simvastatin and exercise in a mouse model of vascular cognitive impairment and dementia. FASEB J 2019; 33:13280-13293. [PMID: 31557051 PMCID: PMC6894065 DOI: 10.1096/fj.201901002r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Aerobic physical exercise (EX) and controlling cardiovascular risk factors in midlife can improve and protect cognitive function in healthy individuals and are considered to be effective at reducing late-onset dementia incidence. By investigating commonalities between these preventative approaches, we sought to identify possible targets for effective interventions. We compared the efficacy of EX and simvastatin (SV) pharmacotherapy to counteract cognitive deficits induced by a high-cholesterol diet (2%, HCD) in mice overexpressing TGF-β1 (TGF mice), a model of vascular cognitive impairment and dementia. Cognitive deficits were found in hypercholesterolemic mice for object recognition memory, and both SV and EX prevented this decline. EX improved stimulus-evoked cerebral blood flow responses and was as effective as SV in normalizing endothelium-dependent vasodilatory responses in cerebral arteries. The up-regulation of galectin-3-positive microglial cells in white matter (WM) of HCD-fed TGF mice with cognitive deficits was significantly reduced by both SV and EX concurrently with cognitive recovery. Altered hippocampal neurogenesis, gray matter astrogliosis, or microgliosis did not correlate with cognitive deficits or benefits. Overall, results indicate that SV and EX prevented cognitive decline in hypercholesterolemic mice and that they share common sites of action in preventing endothelial cell dysfunction and reducing WM inflammation.-Trigiani, L. J., Royea, J., Tong, X.-K., Hamel, E. Comparative benefits of simvastatin and exercise in a mouse model of vascular cognitive impairment and dementia.
Collapse
Affiliation(s)
- Lianne J Trigiani
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Jessika Royea
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Xin-Kang Tong
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
38
|
Cabral DF, Rice J, Morris TP, Rundek T, Pascual-Leone A, Gomes-Osman J. Exercise for Brain Health: An Investigation into the Underlying Mechanisms Guided by Dose. Neurotherapeutics 2019; 16:580-599. [PMID: 31197642 PMCID: PMC6694330 DOI: 10.1007/s13311-019-00749-w] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
There is a strong link between the practice of regular physical exercise and maintenance of cognitive brain health. Animal and human studies have shown that exercise exerts positive effects on cognition through a variety of mechanisms, such as changes in brain volume and connectivity, cerebral perfusion, synaptic plasticity, neurogenesis, and regulation of trophic factors. However, much of this data has been conducted in young humans and animals, raising questions regarding the generalizability of these findings to aging adults. Furthermore, it is not clear at which doses these effects might take place, and if effects would differ with varying exercise modes (such as aerobic, resistance training, combinations, or other). The purpose of this review is to summarize the evidence on the effects of exercise interventions on various mechanisms believed to support cognitive improvements: cerebral perfusion, synaptic neuroplasticity, brain volume and connectivity, neurogenesis, and regulation of trophic factors. We synthesized the findings according to exposure to exercise (short- [1 day-16 weeks], medium- [24-40 weeks], and long-term exercise [52 weeks and beyond]) and have limited our discussion of dose effects to studies in aging adults and aged animals (when human data was not available).
Collapse
Affiliation(s)
- Danylo F Cabral
- Department of Physical Therapy, University of Miami Miller School of Medicine, 5915 Ponce de Leon Boulevard, 5th Floor, Coral Gables, Florida, 33146, USA
- Evelyn McKnight Brain Institute, University of Miami Miller School of Medicine, 1150 Northwest 14th Street, Suite 309, Miami, Florida, 33136, USA
| | - Jordyn Rice
- Department of Physical Therapy, University of Miami Miller School of Medicine, 5915 Ponce de Leon Boulevard, 5th Floor, Coral Gables, Florida, 33146, USA
- Evelyn McKnight Brain Institute, University of Miami Miller School of Medicine, 1150 Northwest 14th Street, Suite 309, Miami, Florida, 33136, USA
| | - Timothy P Morris
- Berenson-Allen Center for Noninvasive Brain Stimulation and Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, USA
| | - Tatjana Rundek
- Evelyn McKnight Brain Institute, University of Miami Miller School of Medicine, 1150 Northwest 14th Street, Suite 309, Miami, Florida, 33136, USA
- Department of Neurology, University of Miami Miller School of Medicine, 1150 Northwest 14th Street, Suite 309, Miami, Florida, 33136, USA
| | - Alvaro Pascual-Leone
- Berenson-Allen Center for Noninvasive Brain Stimulation and Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, USA
| | - Joyce Gomes-Osman
- Department of Physical Therapy, University of Miami Miller School of Medicine, 5915 Ponce de Leon Boulevard, 5th Floor, Coral Gables, Florida, 33146, USA.
- Evelyn McKnight Brain Institute, University of Miami Miller School of Medicine, 1150 Northwest 14th Street, Suite 309, Miami, Florida, 33136, USA.
- Berenson-Allen Center for Noninvasive Brain Stimulation and Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, USA.
- Department of Neurology, University of Miami Miller School of Medicine, 1150 Northwest 14th Street, Suite 309, Miami, Florida, 33136, USA.
| |
Collapse
|
39
|
Rêgo MLM, Cabral DAR, Costa EC, Fontes EB. Physical Exercise for Individuals with Hypertension: It Is Time to Emphasize its Benefits on the Brain and Cognition. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2019; 13:1179546819839411. [PMID: 30967748 PMCID: PMC6444761 DOI: 10.1177/1179546819839411] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/27/2019] [Indexed: 01/01/2023]
Abstract
Hypertension affects more than 40% of adults worldwide and is associated with stroke, myocardial infarction, heart failure, and other cardiovascular diseases. It has also been shown to cause severe functional and structural damage to the brain, leading to cognitive impairment and dementia. Furthermore, it is believed that these cognitive impairments affect the mental ability to maintain productivity at work, ultimately causing social and economic problems. Because hypertension is a chronic condition that requires clinical treatment, strategies with fewer side effects and less-invasive procedures are needed. Physical exercise (PE) has proven to be an efficient and complementary tool for hypertension management, and its peripheral benefits have been widely supported by related studies. However, few studies have specifically examined the potential positive effects of PE on the brain in hypertensive individuals. This narrative review discusses the pathophysiological mechanisms that hypertension promotes in the brain, and suggests PE as an important tool to prevent and reduce cognitive damage caused by hypertension. We also provide PE recommendations for hypertensive individuals, as well as suggestions for promoting PE as a method for increasing cognitive abilities in the brain, particularly for hypertensive individuals.
Collapse
Affiliation(s)
- Maria LM Rêgo
- NEUROEX-Research Group in Physical
Activity, Cognition and Behavior, Health Science Center, Federal University of Rio
Grande do Norte, Natal/RN, Brazil
| | - Daniel AR Cabral
- NEUROEX-Research Group in Physical
Activity, Cognition and Behavior, Health Science Center, Federal University of Rio
Grande do Norte, Natal/RN, Brazil
| | - Eduardo C Costa
- GPEACE-Research Group on Acute and
Chronic Effects of Exercise, Health Science Center, Federal University of Rio Grande
do Norte, Natal/RN, Brazil
| | - Eduardo B Fontes
- NEUROEX-Research Group in Physical
Activity, Cognition and Behavior, Health Science Center, Federal University of Rio
Grande do Norte, Natal/RN, Brazil
| |
Collapse
|
40
|
Santisteban MM, Iadecola C. Hypertension, dietary salt and cognitive impairment. J Cereb Blood Flow Metab 2018; 38:2112-2128. [PMID: 30295560 PMCID: PMC6282225 DOI: 10.1177/0271678x18803374] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/31/2018] [Indexed: 12/12/2022]
Abstract
Dementia is growing at an alarming rate worldwide. Although Alzheimer disease is the leading cause, over 50% of individuals diagnosed with Alzheimer disease have vascular lesions at autopsy. There has been an increasing appreciation of the pathogenic role of vascular risk factors in cognitive impairment caused by neurodegeneration. Midlife hypertension is a leading risk factor for late-life dementia. Hypertension alters cerebrovascular structure, impairs the major factors regulating the cerebral microcirculation, and promotes Alzheimer pathology. Experimental studies have identified brain perivascular macrophages as the major free radical source mediating neurovascular dysfunction of hypertension. Recent evidence indicates that high dietary salt may also induce cognitive impairment. Contrary to previous belief, the effect is not necessarily associated with hypertension and is mediated by a deficit in endothelial nitric oxide. Collectively, the evidence suggests a remarkable cellular diversity of the impact of vascular risk factors on the cerebral vasculature and cognition. Whereas long-term longitudinal epidemiological studies are needed to resolve the temporal relationships between vascular risk factors and cognitive dysfunction, single-cell molecular studies of the vasculature in animal models will provide a fuller mechanistic understanding. This knowledge is critical for developing new preventive, diagnostic, and therapeutic approaches for these devastating diseases of the mind.
Collapse
Affiliation(s)
- Monica M Santisteban
- Feil Family Brain and Mind Research Institute Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
41
|
Tsuneki H, Wada T, Sasaoka T. Chronopathophysiological implications of orexin in sleep disturbances and lifestyle-related disorders. Pharmacol Ther 2018; 186:25-44. [DOI: 10.1016/j.pharmthera.2017.12.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
42
|
Physical Activity and Exercise Therapy Benefit More Than Just Symptoms and Impairments in People With Hip and Knee Osteoarthritis. J Orthop Sports Phys Ther 2018; 48:439-447. [PMID: 29669488 DOI: 10.2519/jospt.2018.7877] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Synopsis Osteoarthritis (OA) of the hip and knee is among the leading causes of global disability, highlighting the need for early, targeted, and effective treatment. The benefits of exercise therapy in people with hip and knee OA are substantial and supported by high-quality evidence, underlining that it should be part of first-line treatment in clinical practice. Furthermore, unlike other treatments for OA, such as analgesia and surgery, exercise therapy is not associated with risk of serious harm. Helping people with OA become more physically active, along with structured exercise therapy targeting symptoms and impairments, is crucial, considering that the majority of people with hip and knee OA do not meet physical activity recommendations. Osteoarthritis is associated with a range of chronic comorbidities, including type 2 diabetes, cardiovascular disease, and dementia, all of which are associated with chronic low-grade inflammation. Physical activity and exercise therapy not only improve symptoms and impairments of OA, but are also effective in preventing at least 35 chronic conditions and treating at least 26 chronic conditions, with one of the potential working mechanisms being exercise-induced anti-inflammatory effects. Patient education may be crucial to ensure long-term adherence and sustained positive effects on symptoms, impairments, physical activity levels, and comorbidities. J Orthop Sports Phys Ther 2018;48(6):439-447. Epub 18 Apr 2018. doi:10.2519/jospt.2018.7877.
Collapse
|
43
|
Niu Y, Wan C, Zhou B, Wang J, Wang J, Chen X, Li R, Wang X, Liu W, Wang Y. Aerobic exercise relieved vascular cognitive impairment via NF-κB/miR-503/BDNF pathway. Am J Transl Res 2018; 10:753-761. [PMID: 29636865 PMCID: PMC5883116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 12/24/2017] [Indexed: 06/08/2023]
Abstract
OBJECTIVE To investigate the mechanism of aerobic exercise in the relief of vascular cognitive impairment (VCI). MATERIALS AND METHODS Latency of Water Maze test was measured at sham, 2VO, 2VO+EX groups. miR-503 and BDNF mRNA levels were detected by quantitative real-time PCR. Protein levels of NF-κB and BDNF were detected by Western blot. Hippocampal neuron cell apoptosis was detected by flow cytometry. Luciferase reporter assay was conducted to investigate the effect of miR-503 on BDNF. RESULTS Latency of Water Maze test in 2VO group was longer than Sham group, while exercise shortened the latency. The expressions of NF-κB and miR-503 in 2VO group were higher than Sham group, while exercise downregulated the expressions. BDNF level in 2VO group were downregulated than Sham group, while exercise upregulated the levels. We also found NF-κB, miR-503 levels were upregulated and BDNF level was downregulated in OGD-treated hippocampal neuron cells. In addition, OGD increased the expression of NF-κB and miR-503, and the expression of miR-503 was downregulated when treated with NF-κB inhibitor (PDTC). Moreover, we confirmed BDNF was a direct target of miR-503. OGD decreased the expression of BDNF, while miR-503 inhibitor reversed this effect. And we proved OGD induced cell apoptosis via NF-κB/miR-503/BDNF. Finally, in rats injected with miR-503 inhibitor, latency of Water Maze test was shortened, miR-503 expression was downregulated, and BDNF level was upregulated. While in rats injected with miR-503 mimic, the results were the opposite. CONCLUSION Aerobic exercise relieved VCI via NF-κB/miR-503/BDNF pathway.
Collapse
Affiliation(s)
- Yali Niu
- Department of Rehabilitation, The General Hospital, Tianjin Medical UniversityTianjin 300052, China
| | - Chunxiao Wan
- Department of Rehabilitation, The General Hospital, Tianjin Medical UniversityTianjin 300052, China
| | - Bo Zhou
- Basic Medicine College, Tianjin Traditional Chinese Medicine UniversityTianjin 300193, China
| | - Junli Wang
- Department of Rehabilitation, The General Hospital, Tianjin Medical UniversityTianjin 300052, China
| | - Jing Wang
- Department of Rehabilitation, The General Hospital, Tianjin Medical UniversityTianjin 300052, China
| | - Xiaona Chen
- Department of Rehabilitation, The General Hospital, Tianjin Medical UniversityTianjin 300052, China
| | - Ruoying Li
- Department of Rehabilitation, The General Hospital, Tianjin Medical UniversityTianjin 300052, China
| | - Xue Wang
- Department of Rehabilitation, The General Hospital, Tianjin Medical UniversityTianjin 300052, China
| | - Wenjing Liu
- Department of Rehabilitation, The General Hospital, Tianjin Medical UniversityTianjin 300052, China
| | - Yueyun Wang
- Department of Rehabilitation, The General Hospital, Tianjin Medical UniversityTianjin 300052, China
| |
Collapse
|
44
|
Ongali B, Nicolakakis N, Tong XK, Lecrux C, Imboden H, Hamel E. Transforming growth factor-β1 induces cerebrovascular dysfunction and astrogliosis through angiotensin II type 1 receptor-mediated signaling pathways. Can J Physiol Pharmacol 2018; 96:527-534. [PMID: 29505736 DOI: 10.1139/cjpp-2017-0640] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Transgenic mice constitutively overexpressing the cytokine transforming growth factor-β1 (TGF-β1) (TGF mice) display cerebrovascular alterations as seen in Alzheimer's disease (AD) and vascular cognitive impairment and dementia (VCID), but no or only subtle cognitive deficits. TGF-β1 may exert part of its deleterious effects through interactions with angiotensin II (AngII) type 1 receptor (AT1R) signaling pathways. We test such interactions in the brain and cerebral vessels of TGF mice by measuring cerebrovascular reactivity, levels of protein markers of vascular fibrosis, nitric oxide synthase activity, astrogliosis, and mnemonic performance in mice treated (6 months) with the AT1R blocker losartan (10 mg/kg per day) or the angiotensin converting enzyme inhibitor enalapril (3 mg/kg per day). Both treatments restored the severely impaired cerebrovascular reactivity to acetylcholine, calcitonin gene-related peptide, endothelin-1, and the baseline availability of nitric oxide in aged TGF mice. Losartan, but not enalapril, significantly reduced astrogliosis and cerebrovascular levels of profibrotic protein connective tissue growth factor while raising levels of antifibrotic enzyme matrix metallopeptidase-9. Memory was unaffected by aging and treatments. The results suggest a pivotal role for AngII in TGF-β1-induced cerebrovascular dysfunction and neuroinflammation through AT1R-mediated mechanisms. Further, they suggest that AngII blockers could be appropriate against vasculopathies and astrogliosis associated with AD and VCID.
Collapse
Affiliation(s)
- Brice Ongali
- a Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| | - Nektaria Nicolakakis
- a Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| | - Xin-Kang Tong
- a Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| | - Clotilde Lecrux
- a Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| | - Hans Imboden
- b Institute of Cell Biology, University of Bern Baltzerstrasse 43012 Bern, Switzerland
| | - Edith Hamel
- a Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| |
Collapse
|
45
|
Van Skike CE, Galvan V. A Perfect sTORm: The Role of the Mammalian Target of Rapamycin (mTOR) in Cerebrovascular Dysfunction of Alzheimer's Disease: A Mini-Review. Gerontology 2018; 64:205-211. [PMID: 29320772 PMCID: PMC5876078 DOI: 10.1159/000485381] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/16/2017] [Indexed: 12/14/2022] Open
Abstract
Cerebrovascular dysfunction is detected prior to the onset of cognitive and histopathological changes in Alzheimer's disease (AD). Increasing evidence indicates a critical role of cerebrovascular dysfunction in the initiation and progression of AD. Recent studies identified the mechanistic/mammalian target of rapamycin (mTOR) as a critical effector of cerebrovascular dysfunction in AD. mTOR has a key role in the regulation of metabolism, but some mTOR-dependent mechanisms are uniquely specific to the regulation of cerebrovascular function. These include the regulation of cerebral blood flow, blood-brain barrier integrity and maintenance, neurovascular coupling, and cerebrovascular reactivity. This article examines the available evidence for a role of mTOR-driven cerebrovascular dysfunction in the pathogenesis of AD and of vascular cognitive impairment and dementia (VCID) and highlights the therapeutic potential of targeting mTOR and/or specific downstream effectors for vasculoprotection in AD, VCID, and other age-associated neurological diseases with cerebrovascular etiology.
Collapse
Affiliation(s)
- Candice E Van Skike
- Department of Cellular and Integrative Physiology and The Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, USA
| | | |
Collapse
|
46
|
Hase Y, Horsburgh K, Ihara M, Kalaria RN. White matter degeneration in vascular and other ageing-related dementias. J Neurochem 2018; 144:617-633. [DOI: 10.1111/jnc.14271] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 10/20/2017] [Accepted: 11/20/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Yoshiki Hase
- Neurovascular Research Group; Institute of Neuroscience; Newcastle University; Newcastle Upon Tyne UK
| | - Karen Horsburgh
- Centre for Neuroregeneration; University of Edinburgh; Edinburgh UK
| | - Masafumi Ihara
- Department of Neurology; National Cerebral and Cardiovascular Center; Suita Osaka Japan
| | - Raj N. Kalaria
- Neurovascular Research Group; Institute of Neuroscience; Newcastle University; Newcastle Upon Tyne UK
| |
Collapse
|