1
|
Ibañez S, Sengupta N, Luebke JI, Wimmer K, Weaver CM. Myelin dystrophy impairs signal transmission and working memory in a multiscale model of the aging prefrontal cortex. eLife 2024; 12:RP90964. [PMID: 39028036 PMCID: PMC11259433 DOI: 10.7554/elife.90964] [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] [Indexed: 07/20/2024] Open
Abstract
Normal aging leads to myelin alterations in the rhesus monkey dorsolateral prefrontal cortex (dlPFC), which are positively correlated with degree of cognitive impairment. It is hypothesized that remyelination with shorter and thinner myelin sheaths partially compensates for myelin degradation, but computational modeling has not yet explored these two phenomena together systematically. Here, we used a two-pronged modeling approach to determine how age-related myelin changes affect a core cognitive function: spatial working memory. First, we built a multicompartment pyramidal neuron model fit to monkey dlPFC empirical data, with an axon including myelinated segments having paranodes, juxtaparanodes, internodes, and tight junctions. This model was used to quantify conduction velocity (CV) changes and action potential (AP) failures after demyelination and subsequent remyelination. Next, we incorporated the single neuron results into a spiking neural network model of working memory. While complete remyelination nearly recovered axonal transmission and network function to unperturbed levels, our models predict that biologically plausible levels of myelin dystrophy, if uncompensated by other factors, can account for substantial working memory impairment with aging. The present computational study unites empirical data from ultrastructure up to behavior during normal aging, and has broader implications for many demyelinating conditions, such as multiple sclerosis or schizophrenia.
Collapse
Affiliation(s)
- Sara Ibañez
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of MedicineBostonUnited States
- Centre de Recerca Matemàtica, Edifici C, Campus BellaterraBellaterraSpain
- Departament de Matemàtiques, Universitat Autònoma de Barcelona, Edifici CBellaterraSpain
| | - Nilapratim Sengupta
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of MedicineBostonUnited States
- Department of Mathematics, Franklin and Marshall CollegeLancasterUnited States
| | - Jennifer I Luebke
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of MedicineBostonUnited States
| | - Klaus Wimmer
- Centre de Recerca Matemàtica, Edifici C, Campus BellaterraBellaterraSpain
- Departament de Matemàtiques, Universitat Autònoma de Barcelona, Edifici CBellaterraSpain
| | - Christina M Weaver
- Department of Mathematics, Franklin and Marshall CollegeLancasterUnited States
| |
Collapse
|
2
|
Wang Q, Qi L, He C, Feng H, Xie C. Age- and gender-related dispersion of brain networks across the lifespan. GeroScience 2024; 46:1303-1318. [PMID: 37542582 PMCID: PMC10828139 DOI: 10.1007/s11357-023-00900-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 07/30/2023] [Indexed: 08/07/2023] Open
Abstract
The effects of age and gender on large-scale resting-state networks (RSNs) reflecting within- and between-network connectivity in the healthy brain remain unclear. This study investigated how age and gender influence the brain network roles and topological properties underlying the ageing process. Ten RSNs were constructed based on 998 participants from the REST-meta-MDD cohort. Multivariate linear regression analysis was used to examine the independent and interactive influences of age and gender on large-scale RSNs and their topological properties. A support vector regression model integrating whole-brain network features was used to predict brain age across the lifespan and cognitive decline in an Alzheimer's disease spectrum (ADS) sample. Differential effects of age and gender on brain network roles were demonstrated across the lifespan. Specifically, cingulo-opercular, auditory, and visual (VIS) networks showed more incohesive features reflected by decreased intra-network connectivity with ageing. Further, females displayed distinctive brain network trajectory patterns in middle-early age, showing enhanced network connectivity within the fronto-parietal network (FPN) and salience network (SAN) and weakened network connectivity between the FPN-somatomotor, FPN-VIS, and SAN-VIS networks. Age - but not gender - induced widespread decrease in topological properties of brain networks. Importantly, these differential network features predicted brain age and cognitive impairment in the ADS sample. By showing that age and gender exert specific dispersion of dynamic network roles and trajectories across the lifespan, this study has expanded our understanding of age- and gender-related brain changes with ageing. Moreover, the findings may be useful for detecting early-stage dementia.
Collapse
Affiliation(s)
- Qing Wang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Lingyu Qi
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Cancan He
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Haixia Feng
- Department of Nursing, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Chunming Xie
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China.
- Institute of Neuropsychiatry, Affiliated ZhongDa Hospital, Southeast University, Nanjing, Jiangsu, 210009, China.
- The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu, 210096, China.
| |
Collapse
|
3
|
Rivi V, Batabyal A, Lukowiak K. The multifaceted effects of flavonoids on neuroplasticity. Restor Neurol Neurosci 2024; 42:93-111. [PMID: 38995810 DOI: 10.3233/rnn-230150] [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] [Indexed: 07/14/2024]
Abstract
There has been a significant increase in the incidence of multiple neurodegenerative and terminal diseases in the human population with life expectancy increasing in the current times. This highlights the urgent need for a more comprehensive understanding of how different aspects of lifestyle, in particular diet, may affect neural functioning and consequently cognitive performance as well as in enhancing overall health. Flavonoids, found in a variety of fruits, vegetables, and derived beverages, provide a new avenue of research that shows a promising influence on different aspects of brain function. However, despite the promising evidence, most bioactive compounds lack strong clinical research efficacy. In the current scoping review, we highlight the effects of Flavonoids on cognition and neural plasticity across vertebrates and invertebrates with special emphasis on the studies conducted in the pond snail, Lymnaea stagnalis, which has emerged to be a functionally dynamic model for studies on learning and memory. In conclusion, we suggest future research directions and discuss the social, cultural, and ethnic dependencies of bioactive compounds that influence how these compounds are used and accepted globally. Bridging the gap between preclinical and clinical studies about the effects of bioactive natural compounds on brain health will surely lead to lifestyle choices such as dietary Flavonoids being used complementarily rather than as replacements to classical drugs bringing about a healthier future.
Collapse
Affiliation(s)
- Veronica Rivi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Anuradha Batabyal
- Department of Physical and Natural Sciences, FLAME University, India
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Ken Lukowiak
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
4
|
Wang J, Li N, Meng Z, Li Q. Change point detection for high dimensional data via kernel measure with application to human aging brain data. Stat Med 2023; 42:4644-4663. [PMID: 37649243 DOI: 10.1002/sim.9881] [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: 03/31/2023] [Revised: 08/07/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023]
Abstract
Identifying the existence and locations of change points has been a broadly encountered task in many statistical application areas. The existing change point detection methods may produce unsatisfactory results for high-dimensional data since certain distributional assumptions are made on data, which are hard to verify in practice. Moreover, some parameters (such as the number of change points) need to be estimated beforehand for some methods, making their powers sensitive to these values. Here, we propose a kernel-basedU $$ U $$ -statistic to identify change points (KUCP) for high dimensional data, which is free of distributional assumptions and sup-parameter estimations. Specifically, we employ a kernel function to describe similarities among the subjects and construct aU $$ U $$ -statistic to test the existence of change point for a given location. The asymptotic properties of theU $$ U $$ -statistic are deduced. We also develop a procedure to locate the change points sequentially via a dichotomy algorithm. Extensive simulations demonstrate that KUCP has higher sensitivity in identifying existence of change points and higher accuracy in locating these change points than its counterparts. We further illustrate its practical utility by analyzing a gene expression data of human brain to detect the time point when gene expression profiles begin to change, which has been reported to be closely related with aging brain.
Collapse
Affiliation(s)
- Jinjuan Wang
- School of Mathematics and Statistics, Beijing Institute of Technology, Beijing, China
| | - Na Li
- School of Applied Science, Beijing Information Science and Technology University, Beijing, China
| | - Zhen Meng
- School of Statistics, Capital University of Economics and Business, Beijing, China
| | - Qizhai Li
- LSC Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
5
|
Zhang T, Liu W, Bai Q, Gao S. Virtual reality technology in the rehabilitation of post-stroke cognitive impairment: an opinion article on recent findings. Front Psychol 2023; 14:1271458. [PMID: 37849482 PMCID: PMC10577207 DOI: 10.3389/fpsyg.2023.1271458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023] Open
Affiliation(s)
- Ting Zhang
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
- Department of Traditional Chinese Medicine, University Hospital, Zhejiang Normal University, Jinhua, China
| | - Wei Liu
- Physical Education College, Guangxi University of Science and Technology, Liuzhou, China
| | - Qingping Bai
- Physical Education College, Guangxi University of Science and Technology, Liuzhou, China
| | - Song Gao
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
| |
Collapse
|
6
|
Ibañez S, Sengupta N, Luebke JI, Wimmer K, Weaver CM. Myelin dystrophy in the aging prefrontal cortex leads to impaired signal transmission and working memory decline: a multiscale computational study. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.30.555476. [PMID: 37693412 PMCID: PMC10491254 DOI: 10.1101/2023.08.30.555476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Normal aging leads to myelin alternations in the rhesus monkey dorsolateral prefrontal cortex (dlPFC), which are often correlated with cognitive impairment. It is hypothesized that remyelination with shorter and thinner myelin sheaths partially compensates for myelin degradation, but computational modeling has not yet explored these two phenomena together systematically. Here, we used a two-pronged modeling approach to determine how age-related myelin changes affect a core cognitive function: spatial working memory. First we built a multicompartment pyramidal neuron model fit to monkey dlPFC data, with axon including myelinated segments having paranodes, juxtaparanodes, internodes, and tight junctions, to quantify conduction velocity (CV) changes and action potential (AP) failures after demyelination and subsequent remyelination in a population of neurons. Lasso regression identified distinctive parameter sets likely to modulate an axon's susceptibility to CV changes following demyelination versus remyelination. Next we incorporated the single neuron results into a spiking neural network model of working memory. While complete remyelination nearly recovered axonal transmission and network function to unperturbed levels, our models predict that biologically plausible levels of myelin dystrophy, if uncompensated by other factors, can account for substantial working memory impairment with aging. The present computational study unites empirical data from electron microscopy up to behavior on aging, and has broader implications for many demyelinating conditions, such as multiple sclerosis or schizophrenia.
Collapse
Affiliation(s)
- Sara Ibañez
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA 02118
- Centre de Recerca Matemàtica, Edifici C, Campus Bellaterra, 08193 Bellaterra, Spain
| | - Nilapratim Sengupta
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA 02118
- Department of Mathematics, Franklin and Marshall College, Lancaster, PA, USA 17604
| | - Jennifer I Luebke
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA 02118
| | - Klaus Wimmer
- Centre de Recerca Matemàtica, Edifici C, Campus Bellaterra, 08193 Bellaterra, Spain
| | - Christina M Weaver
- Department of Mathematics, Franklin and Marshall College, Lancaster, PA, USA 17604
| |
Collapse
|
7
|
Tian J, Guo L, Wang T, Jia K, Swerdlow RH, Zigman JM, Du H. Liver-expressed antimicrobial peptide 2 elevation contributes to age-associated cognitive decline. JCI Insight 2023; 8:166175. [PMID: 37212281 DOI: 10.1172/jci.insight.166175] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 03/31/2023] [Indexed: 05/23/2023] Open
Abstract
Elderly individuals frequently report cognitive decline, while various studies indicate hippocampal functional declines with advancing age. Hippocampal function is influenced by ghrelin through hippocampus-expressed growth hormone secretagogue receptor (GHSR). Liver-expressed antimicrobial peptide 2 (LEAP2) is an endogenous GHSR antagonist that attenuates ghrelin signaling. Here, we measured plasma ghrelin and LEAP2 levels in a cohort of cognitively normal individuals older than 60 and found that LEAP2 increased with age while ghrelin (also referred to in literature as "acyl-ghrelin") marginally declined. In this cohort, plasma LEAP2/ghrelin molar ratios were inversely associated with Mini-Mental State Examination scores. Studies in mice showed an age-dependent inverse relationship between plasma LEAP2/ghrelin molar ratio and hippocampal lesions. In aged mice, restoration of the LEAP2/ghrelin balance to youth-associated levels with lentiviral shRNA Leap2 downregulation improved cognitive performance and mitigated various age-related hippocampal deficiencies such as CA1 region synaptic loss, declines in neurogenesis, and neuroinflammation. Our data collectively suggest that LEAP2/ghrelin molar ratio elevation may adversely affect hippocampal function and, consequently, cognitive performance; thus, it may serve as a biomarker of age-related cognitive decline. Moreover, targeting LEAP2 and ghrelin in a manner that lowers the plasma LEAP2/ghrelin molar ratio could benefit cognitive performance in elderly individuals for rejuvenation of memory.
Collapse
Affiliation(s)
- Jing Tian
- Department of Pharmacology and Toxicology and
| | - Lan Guo
- Higuchi Biosciences Center, University of Kansas, Lawrence, Kansas, USA
| | - Tienju Wang
- Department of Pharmacology and Toxicology and
| | - Kun Jia
- Department of Pharmacology and Toxicology and
| | - Russell H Swerdlow
- Alzheimer's Disease Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jeffrey M Zigman
- Departments of Internal Medicine and Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Heng Du
- Department of Pharmacology and Toxicology and
- Higuchi Biosciences Center, University of Kansas, Lawrence, Kansas, USA
- Alzheimer's Disease Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
8
|
Kumar A, Konar A. Editorial: Neuromodulating bioactive compounds as potential cognitive therapeutics. Front Aging Neurosci 2023; 15:1143193. [PMID: 36819727 PMCID: PMC9936231 DOI: 10.3389/fnagi.2023.1143193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Affiliation(s)
- Ashish Kumar
- Brain Science Research Institute, AriBio Co., Ltd., Seongnam-si, Gyeonggi-do, Republic of Korea,*Correspondence: Ashish Kumar ✉
| | - Arpita Konar
- Institute of Health Sciences, Presidency University, Kolkata, India,Arpita Konar ✉
| |
Collapse
|
9
|
Kassis A, Fichot MC, Horcajada MN, Horstman AMH, Duncan P, Bergonzelli G, Preitner N, Zimmermann D, Bosco N, Vidal K, Donato-Capel L. Nutritional and lifestyle management of the aging journey: A narrative review. Front Nutr 2023; 9:1087505. [PMID: 36761987 PMCID: PMC9903079 DOI: 10.3389/fnut.2022.1087505] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/28/2022] [Indexed: 01/25/2023] Open
Abstract
With age, the physiological responses to occasional or regular stressors from a broad range of functions tend to change and adjust at a different pace and restoring these functions in the normal healthy range becomes increasingly challenging. Even if this natural decline is somehow unavoidable, opportunities exist to slow down and attenuate the impact of advancing age on major physiological processes which, when weakened, constitute the hallmarks of aging. This narrative review revisits the current knowledge related to the aging process and its impact on key metabolic functions including immune, digestive, nervous, musculoskeletal, and cardiovascular functions; and revisits insights into the important biological targets that could inspire effective strategies to promote healthy aging.
Collapse
Affiliation(s)
- Amira Kassis
- Whiteboard Nutrition Science, Beaconsfield, QC, Canada,Amira Kassis,
| | | | | | | | - Peter Duncan
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | | | - Nicolas Preitner
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Diane Zimmermann
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Nabil Bosco
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Karine Vidal
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Laurence Donato-Capel
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland,*Correspondence: Laurence Donato-Capel,
| |
Collapse
|
10
|
Zangerolamo L, Carvalho M, Barssotti L, Soares GM, Marmentini C, Boschero AC, Barbosa HCL. The bile acid TUDCA reduces age-related hyperinsulinemia in mice. Sci Rep 2022; 12:22273. [PMID: 36564463 PMCID: PMC9789133 DOI: 10.1038/s41598-022-26915-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
Aging is associated with glucose metabolism disturbances, such as insulin resistance and hyperinsulinemia, which contribute to the increased prevalence of type 2 diabetes (T2D) and its complications in the elderly population. In this sense, some bile acids have emerged as new therapeutic targets to treat TD2, as well as associated metabolic disorders. The taurine conjugated bile acid, tauroursodeoxycholic acid (TUDCA) improves glucose homeostasis in T2D, obesity, and Alzheimer's disease mice model. However, its effects in aged mice have not been explored yet. Here, we evaluated the actions of TUDCA upon glucose-insulin homeostasis in aged C57BL/6 male mice (18-month-old) treated with 300 mg/kg of TUDCA or its vehicle. TUDCA attenuated hyperinsulinemia and improved glucose homeostasis in aged mice, by enhancing liver insulin-degrading enzyme (IDE) expression and insulin clearance. Furthermore, the improvement in glucose-insulin homeostasis in these mice was accompanied by a reduction in adiposity, associated with adipocyte hypertrophy, and lipids accumulation in the liver. TUDCA-treated aged mice also displayed increased energy expenditure and metabolic flexibility, as well as a better cognitive ability. Taken together, our data highlight TUDCA as an interesting target for the attenuation of age-related hyperinsulinemia and its deleterious effects on metabolism.
Collapse
Affiliation(s)
- Lucas Zangerolamo
- grid.411087.b0000 0001 0723 2494Obesity and Comorbidities Research Center, Department of Structural and Functional Biology, University of Campinas, UNICAMP, Campinas, Sao Paulo CEP: 13083-864 Brazil
| | - Marina Carvalho
- grid.411087.b0000 0001 0723 2494Obesity and Comorbidities Research Center, Department of Structural and Functional Biology, University of Campinas, UNICAMP, Campinas, Sao Paulo CEP: 13083-864 Brazil
| | - Leticia Barssotti
- grid.411087.b0000 0001 0723 2494Obesity and Comorbidities Research Center, Department of Structural and Functional Biology, University of Campinas, UNICAMP, Campinas, Sao Paulo CEP: 13083-864 Brazil
| | - Gabriela M. Soares
- grid.411087.b0000 0001 0723 2494Obesity and Comorbidities Research Center, Department of Structural and Functional Biology, University of Campinas, UNICAMP, Campinas, Sao Paulo CEP: 13083-864 Brazil
| | - Carine Marmentini
- grid.411087.b0000 0001 0723 2494Obesity and Comorbidities Research Center, Department of Structural and Functional Biology, University of Campinas, UNICAMP, Campinas, Sao Paulo CEP: 13083-864 Brazil
| | - Antonio C. Boschero
- grid.411087.b0000 0001 0723 2494Obesity and Comorbidities Research Center, Department of Structural and Functional Biology, University of Campinas, UNICAMP, Campinas, Sao Paulo CEP: 13083-864 Brazil
| | - Helena Cristina L. Barbosa
- grid.411087.b0000 0001 0723 2494Obesity and Comorbidities Research Center, Department of Structural and Functional Biology, University of Campinas, UNICAMP, Campinas, Sao Paulo CEP: 13083-864 Brazil
| |
Collapse
|
11
|
Shimizu S, Kasai S, Yamazaki H, Tatara Y, Mimura J, Engler MJ, Tanji K, Nikaido Y, Inoue T, Suganuma H, Wakabayashi K, Itoh K. Sulforaphane Increase Mitochondrial Biogenesis-Related Gene Expression in the Hippocampus and Suppresses Age-Related Cognitive Decline in Mice. Int J Mol Sci 2022; 23:ijms23158433. [PMID: 35955572 PMCID: PMC9369397 DOI: 10.3390/ijms23158433] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 11/22/2022] Open
Abstract
Sulforaphane (SFN) is a potent activator of the transcriptional factor, Nuclear Factor Erythroid 2 (NF-E2)-Related factor 2 (NRF2). SFN and its precursor, glucoraphanin (sulforaphane glucosinolate, SGS), have been shown to ameliorate cognitive function in clinical trials and in vivo studies. However, the effects of SGS on age-related cognitive decline in Senescence-Accelerated Mouse Prone 8 (SAMP8) is unknown. In this study, we determined the preventive potential of SGS on age-related cognitive decline. One-month old SAMP8 mice or control SAM resistance 1 (SAMR1) mice were fed an ad libitum diet with or without SGS-containing broccoli sprout powder (0.3% w/w SGS in diet) until 13 months of age. SGS significantly improved long-term memory in SAMP8 at 12 months of age. Interestingly, SGS increased hippocampal mRNA and protein levels of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC1α) and mitochondrial transcription factor A (TFAM), which are master regulators of mitochondrial biogenesis, both in SAMR1 and SAMP8 at 13 months of age. Furthermore, mRNAs for nuclear respiratory factor-1 (NRF-1) and mitochondrial DNA-encoded respiratory complex enzymes, but not mitochondrial DNA itself, were increased by SGS in SAMP8 mice. These results suggest that SGS prevents age-related cognitive decline by maintaining mitochondrial function in senescence-accelerated mice.
Collapse
Affiliation(s)
- Sunao Shimizu
- Innovation Division, KAGOME Co., Ltd., 17 Nishitomiyama, Nasushiobara 329-2762, Tochigi, Japan; (S.S.); (T.I.); (H.S.)
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan; (S.K.); (H.Y.); (Y.T.); (J.M.)
- Department of Stress Response Science, Center for Advanced Medical Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan;
| | - Shuya Kasai
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan; (S.K.); (H.Y.); (Y.T.); (J.M.)
- Department of Stress Response Science, Center for Advanced Medical Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan;
| | - Hiromi Yamazaki
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan; (S.K.); (H.Y.); (Y.T.); (J.M.)
- Department of Stress Response Science, Center for Advanced Medical Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan;
| | - Yota Tatara
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan; (S.K.); (H.Y.); (Y.T.); (J.M.)
- Department of Stress Response Science, Center for Advanced Medical Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan;
| | - Junsei Mimura
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan; (S.K.); (H.Y.); (Y.T.); (J.M.)
- Department of Stress Response Science, Center for Advanced Medical Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan;
| | - Máté János Engler
- Department of Stress Response Science, Center for Advanced Medical Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan;
| | - Kunikazu Tanji
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan; (K.T.); (K.W.)
| | - Yoshikazu Nikaido
- Department of Metabolomics Innovation, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan;
- Department of Anesthesiology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan
| | - Takuro Inoue
- Innovation Division, KAGOME Co., Ltd., 17 Nishitomiyama, Nasushiobara 329-2762, Tochigi, Japan; (S.S.); (T.I.); (H.S.)
| | - Hiroyuki Suganuma
- Innovation Division, KAGOME Co., Ltd., 17 Nishitomiyama, Nasushiobara 329-2762, Tochigi, Japan; (S.S.); (T.I.); (H.S.)
| | - Koichi Wakabayashi
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan; (K.T.); (K.W.)
| | - Ken Itoh
- Department of Vegetable Life Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan; (S.K.); (H.Y.); (Y.T.); (J.M.)
- Department of Stress Response Science, Center for Advanced Medical Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Aomori, Japan;
- Correspondence:
| |
Collapse
|
12
|
Genetic Expression between Ageing and Exercise: Secreted Protein Acidic and Rich in Cysteine as a Potential “Exercise Substitute” Antiageing Therapy. Genes (Basel) 2022; 13:genes13060950. [PMID: 35741712 PMCID: PMC9223223 DOI: 10.3390/genes13060950] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
Ageing is the effect of time on biological entities. It represents a risk factor for a variety of diseases and health disorders; thus, therapeutic options are required to tackle ageing issues. Modern geriatric medicine prescribes exercise to counteract ageing effects. This work presents secreted protein acidic and rich in cysteine (SPARC) as a potential antiageing therapy. Indeed, SPARC declines with ageing, exercise induces SPARC, and SPARC overexpression in mice mimics exercise. Thus, we hypothesize that SPARC is an exercise-induced factor that is beyond—at least part of—the antiageing effects induced by exercise. This could become a potential antiageing therapy for the elderly that counteracts ageing by mimicking the effects of exercise without needing to perform exercise. This is of particular importance because ageing usually reduces mobility and age-related diseases can reduce the ability to perform the required physical activity. On the other hand, the possibilities of mimicking exercise benefits via SPARC are not limited to ageing, and can be applied in various contexts in which exercise cannot be performed because of physical disabilities, health disorders, or limited mobility.
Collapse
|
13
|
Olaniran AF, Taiwo AE, Bamidele OP, Iranloye YM, Malomo AA, Olaniran OD. The role of nutraceutical fruit drink on neurodegenerative diseases: a review. Int J Food Sci Technol 2022. [DOI: 10.1111/ijfs.15106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Abiola Folakemi Olaniran
- Landmark University SDG 12 (Responsible Consumption and Production Group) Department of Food Science and Nutrition College of Agriculture Landmark University P.M.B. 1001 Omu‐Aran Kwara State Nigeria
| | - Abiola Ezekiel Taiwo
- Department of Chemical Engineering College of Engineering Landmark University PMB 1001 Omu Aran Nigeria
| | | | - Yetunde Mary Iranloye
- Landmark University SDG 12 (Responsible Consumption and Production Group) Department of Food Science and Nutrition College of Agriculture Landmark University P.M.B. 1001 Omu‐Aran Kwara State Nigeria
| | - Adekunbi Adetola Malomo
- Department of Food Science and Technology Faculty of Technology Obafemi Awolowo University Ile‐Ife Nigeria
| | | |
Collapse
|
14
|
Kim CK, Sachdev PS, Braidy N. Recent Neurotherapeutic Strategies to Promote Healthy Brain Aging: Are we there yet? Aging Dis 2022; 13:175-214. [PMID: 35111369 PMCID: PMC8782556 DOI: 10.14336/ad.2021.0705] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/05/2021] [Indexed: 12/21/2022] Open
Abstract
Owing to the global exponential increase in population ageing, there is an urgent unmet need to develop reliable strategies to slow down and delay the ageing process. Age-related neurodegenerative diseases are among the main causes of morbidity and mortality in our contemporary society and represent a major socio-economic burden. There are several controversial factors that are thought to play a causal role in brain ageing which are continuously being examined in experimental models. Among them are oxidative stress and brain inflammation which are empirical to brain ageing. Although some candidate drugs have been developed which reduce the ageing phenotype, their clinical translation is limited. There are several strategies currently in development to improve brain ageing. These include strategies such as caloric restriction, ketogenic diet, promotion of cellular nicotinamide adenine dinucleotide (NAD+) levels, removal of senescent cells, 'young blood' transfusions, enhancement of adult neurogenesis, stem cell therapy, vascular risk reduction, and non-pharmacological lifestyle strategies. Several studies have shown that these strategies can not only improve brain ageing by attenuating age-related neurodegenerative disease mechanisms, but also maintain cognitive function in a variety of pre-clinical experimental murine models. However, clinical evidence is limited and many of these strategies are awaiting findings from large-scale clinical trials which are nascent in the current literature. Further studies are needed to determine their long-term efficacy and lack of adverse effects in various tissues and organs to gain a greater understanding of their potential beneficial effects on brain ageing and health span in humans.
Collapse
Affiliation(s)
- Chul-Kyu Kim
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Perminder S Sachdev
- Neuropsychiatric Institute, Euroa Centre, Prince of Wales Hospital, Sydney, Australia
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| |
Collapse
|
15
|
Fathi A, Mathivanan S, Kong L, Petersen AJ, Harder CK, Block J, Miller JM, Bhattacharyya A, Wang D, Zhang S. Chemically induced senescence in human stem cell-derived neurons promotes phenotypic presentation of neurodegeneration. Aging Cell 2022; 21:e13541. [PMID: 34953016 PMCID: PMC8761019 DOI: 10.1111/acel.13541] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 11/18/2021] [Accepted: 12/10/2021] [Indexed: 01/10/2023] Open
Abstract
Modeling age‐related neurodegenerative disorders with human stem cells are difficult due to the embryonic nature of stem cell‐derived neurons. We developed a chemical cocktail to induce senescence of iPSC‐derived neurons to address this challenge. We first screened small molecules that induce embryonic fibroblasts to exhibit features characteristic of aged fibroblasts. We then optimized a cocktail of small molecules that induced senescence in fibroblasts and cortical neurons without causing DNA damage. The utility of the “senescence cocktail” was validated in motor neurons derived from ALS patient iPSCs which exhibited protein aggregation and axonal degeneration substantially earlier than those without cocktail treatment. Our “senescence cocktail” will likely enhance the manifestation of disease‐related phenotypes in neurons derived from iPSCs, enabling the generation of reliable drug discovery platforms.
Collapse
Affiliation(s)
- Ali Fathi
- Waisman Center University of Wisconsin‐Madison Madison Wisconsin USA
| | | | - Linghai Kong
- Waisman Center University of Wisconsin‐Madison Madison Wisconsin USA
| | | | - Cole R. K. Harder
- Waisman Center University of Wisconsin‐Madison Madison Wisconsin USA
| | - Jasper Block
- Waisman Center University of Wisconsin‐Madison Madison Wisconsin USA
| | | | - Anita Bhattacharyya
- Waisman Center University of Wisconsin‐Madison Madison Wisconsin USA
- Department of Cell and Regenerative Biology School of Medicine and Public Health University of Wisconsin‐Madison Madison Wisconsin USA
| | - Daifeng Wang
- Waisman Center University of Wisconsin‐Madison Madison Wisconsin USA
| | - Su‐Chun Zhang
- Waisman Center University of Wisconsin‐Madison Madison Wisconsin USA
- Department of Neuroscience School of Medicine and Public Health University of Wisconsin Madison Wisconsin USA
- Department of Neurology School of Medicine and Public Health University of Wisconsin Madison Wisconsin USA
- Program in Neuroscience and Behavioral Disorders Duke‐NUS Medical School Singapore Singapore
| |
Collapse
|
16
|
Mavrikaki M, Lee JD, Solomon IH, Slack FJ. Severe COVID-19 induces molecular signatures of aging in the human brain. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.11.24.21266779. [PMID: 34845457 PMCID: PMC8629201 DOI: 10.1101/2021.11.24.21266779] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is predominantly an acute respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and remains a significant threat to public health. COVID-19 is accompanied by neurological symptoms and cognitive decline, but the molecular mechanisms underlying this effect remain unclear. As aging induces distinct molecular signatures in the brain associated with cognitive decline in healthy populations, we hypothesized that COVID-19 may induce molecular signatures of aging. Here, we performed whole transcriptomic analysis of human frontal cortex, a critical area for cognitive function, in 12 COVID-19 cases and age- and sex-matched uninfected controls. COVID-19 induces profound changes in gene expression, despite the absence of detectable virus in brain tissue. Pathway analysis shows downregulation of genes involved in synaptic function and cognition and upregulation of genes involved in immune processes. Comparison with five independent transcriptomic datasets of aging human frontal cortex reveals striking similarities between aged individuals and severe COVID-19 patients. Critically, individuals below 65 years of age exhibit profound transcriptomic changes not observed among older individuals in our patient cohort. Our data indicate that severe COVID-19 induces molecular signatures of aging in the human brain and emphasize the value of neurological follow-up in recovered individuals.
Collapse
Affiliation(s)
- Maria Mavrikaki
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- These authors contributed equally: Maria Mavrikaki and Jonathan D. Lee
- Correspondence to M.M. and F.J.S.: ;
| | - Jonathan D. Lee
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- These authors contributed equally: Maria Mavrikaki and Jonathan D. Lee
| | - Isaac H. Solomon
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Frank J. Slack
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02215, USA
- Correspondence to M.M. and F.J.S.: ;
| |
Collapse
|
17
|
Sinha P, Rani A, Kumar A, Riva A, Brant JO, Foster TC. Examination of CA1 Hippocampal DNA Methylation as a Mechanism for Closing of Estrogen's Critical Window. Front Aging Neurosci 2021; 13:717032. [PMID: 34421577 PMCID: PMC8371553 DOI: 10.3389/fnagi.2021.717032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/15/2021] [Indexed: 02/01/2023] Open
Abstract
There is a critical window for estrogen replacement therapy, beyond which estradiol (E2) fails to enhance cognition and N-methyl-D-aspartate (NMDA) receptor function, and E2-responsive transcription decreases. Much less attention has been given to the mechanism for closing of the critical window, which is thought to involve the decline in estrogen signaling cascades, possibly involving epigenetic mechanisms, including DNA methylation. This study investigated changes in DNA methylation in region CA1 of the hippocampus of ovariectomized female rats over the course of brain aging and in response to E2-treatment, using whole genome bisulfite sequencing. Differential methylation of CpG and non-CpG (CHG and CHH) sites and associated genes were characterized in aged controls (AC), middle-age controls (MC), and young controls (YC) and differential methylation in response to E2-treatment (T) was examined in each age group (AT-AC, MT-MC, and YT-YC). Possible candidate genes for the closing of the critical window were defined as those that were hypomethylated by E2-treatment in younger animals, but were unresponsive in aged animals. Gene ontology categories for possible critical window genes were linked to response to hormones (Adcyap1, Agtr2, Apob, Ahr, Andpro, Calm2, Cyp4a2, Htr1b, Nr3c2, Pitx2, Pth, Pdk4, Slc2a2, Tnc, and Wnt5a), including G-protein receptor signaling (Gpr22 and Rgs4). Other possible critical window genes were linked to glutamate synapses (Nedd4, Grm1, Grm7, and Grin3a). These results suggest that decreased E2 signaling with advanced age, and/or prolonged E2 deprivation, results in methylation of E2-responsive genes, including those involved in rapid E2 signaling, which may limit subsequent transcription.
Collapse
Affiliation(s)
- Puja Sinha
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Asha Rani
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Alberto Riva
- Bioinformatics Core, Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL, United States
| | - Jason Orr Brant
- Department of Biostatistics, University of Florida, Gainesville, FL, United States
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States.,Genetics and Genomics Program, University of Florida, Gainesville, FL, United States
| |
Collapse
|
18
|
Khairy EY, Attia MM. Protective effects of vitamin D on neurophysiologic alterations in brain aging: role of brain-derived neurotrophic factor (BDNF). Nutr Neurosci 2021; 24:650-659. [PMID: 31524100 DOI: 10.1080/1028415x.2019.1665854] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Background/aim: Vitamin D has been hypothesized to be main regulator of the aging rate, alongside evidences support its role in neuroprotection. However, data about the protective role of vitamin D against neurophysiologic alterations associated with brain aging is limited. This study investigated the possible protective effects that vitamin D has on brain-derived neurotrophic factor (BDNF), cholinergic function, oxidative stress and apoptosis in aging rat brain.Methods: Male Wister albino rats aged 5 months (young), 12 months (middle aged) and 24 months (old) (n = 20 each) were used. Each age group subdivided to either vitamin D3 supplementation (500 IU/kg/day orally for 5 weeks) or no supplementation (control) group (n = 10 each). Serum 25-hydroxyvitamin D [25(OH)D], brain BDNF and malondialdehyde levels and activities of acetylcholinesterase (AChE), antioxidant enzymes (glutathione reductase, glutathione peroxidase and superoxide dismutase) and caspase-3 were quantified.Results: Vitamin D supplementation significantly mitigated the observed aging-related reduction in brain BDNF level and activities of AChE and antioxidant enzymes and elevation in malondialdehyde level and caspase-3 activity compared to control groups. Brain BDNF level correlated positively with serum 25(OH) D level and brain AChE activity and negatively with brain malondialdehyde level and caspase-3 activity in supplemented groups.Conclusion: Restoring vitamin D levels may, therefore, represent a useful strategy for healthy brain aging. Augmenting brain BDNF seems to be a key mechanism through which vitamin D counteracts age-related brain dysfunction.
Collapse
Affiliation(s)
- Eman Y Khairy
- Department of Physiology, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Maha M Attia
- Department of Physiology, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
19
|
Zheng M. Cellular Tropism of SARS-CoV-2 across Human Tissues and Age-related Expression of ACE2 and TMPRSS2 in Immune-inflammatory Stromal Cells. Aging Dis 2021; 12:718-725. [PMID: 34094637 PMCID: PMC8139212 DOI: 10.14336/ad.2021.0429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 04/29/2021] [Indexed: 01/08/2023] Open
Abstract
Recently, emerging evidence has indicated that COVID-19 represents a major threat to older populations, but the underlying mechanisms remain unclear. The pathogen causing COVID-19 is acute respiratory syndrome coronavirus 2 (SARS-CoV-2). SARS-CoV-2 infection depends on the key entry factors, angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2). Recognizing the importance of ACE2 and TMPRSS2 for the cellular tropism of SARS-CoV-2, we analyzed and presented the landscape of cell-type identities for ACE2+ TMPRSS2+ cells across different human tissues and the age-related alterations in ACE2 and TMPRSS2 expression across different cell types. Additionally, most of the post-acute COVID-19 sequelae could attribute to the ACE2-expressing organ systems. Therefore, these SARS-CoV-2 tropism data should be an essential resource for guiding clinical treatment and pathological studies, which should draw attention toward the prioritization of COVID-19 research in the future. Notably, we discovered the age-related expression of ACE2 and TMPRSS2 in the immune-inflammatory stromal cells, implying the potential interplay between COVID-19, stromal cells, and aging. In this study, we developed a novel and practical analysis framework for mapping the cellular tropism of SARS-CoV-2. This approach was built to aid the identification of viral-specific cell types and age-related alterations of viral tropism, highlighting the power of single-cell RNA sequencing (scRNA-seq) to address viral pathogenesis systematically. With the rapid accumulation of scRNA-seq data and the continuously increasing insight into viral entry factors, we anticipate that this scRNA-seq-based approach will attract broader interest in the virus research communities.
Collapse
Affiliation(s)
- Ming Zheng
- 1Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China.,2Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing, China
| |
Collapse
|
20
|
Guida JL, Agurs-Collins T, Ahles TA, Campisi J, Dale W, Demark-Wahnefried W, Dietrich J, Fuldner R, Gallicchio L, Green PA, Hurria A, Janelsins MC, Jhappan C, Kirkland JL, Kohanski R, Longo V, Meydani S, Mohile S, Niedernhofer LJ, Nelson C, Perna F, Schadler K, Scott JM, Schrack JA, Tracy RP, van Deursen J, Ness KK. Strategies to Prevent or Remediate Cancer and Treatment-Related Aging. J Natl Cancer Inst 2021; 113:112-122. [PMID: 32348501 PMCID: PMC7850536 DOI: 10.1093/jnci/djaa060] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/20/2020] [Accepted: 04/17/2020] [Indexed: 12/15/2022] Open
Abstract
Up to 85% of adult cancer survivors and 99% of adult survivors of childhood cancer live with an accumulation of chronic conditions, frailty, and/or cognitive impairments resulting from cancer and its treatment. Thus, survivors often show an accelerated development of multiple geriatric syndromes and need therapeutic interventions. To advance progress in this area, the National Cancer Institute convened the second of 2 think tanks under the auspices of the Cancer and Accelerated Aging: Advancing Research for Healthy Survivors initiative. Experts assembled to share evidence of promising strategies to prevent, slow, or reverse the aging consequences of cancer and its treatment. The meeting identified research and resource needs, including geroscience-guided clinical trials; comprehensive assessments of functional, cognitive, and psychosocial vulnerabilities to assess and predict age-related outcomes; preclinical and clinical research to determine the optimal dosing for behavioral (eg, diet, exercise) and pharmacologic (eg, senolytic) therapies; health-care delivery research to evaluate the efficacy of integrated cancer care delivery models; optimization of intervention implementation, delivery, and uptake; and patient and provider education on cancer and treatment-related late and long-term adverse effects. Addressing these needs will expand knowledge of aging-related consequences of cancer and cancer treatment and inform strategies to promote healthy aging of cancer survivors.
Collapse
Affiliation(s)
- Jennifer L Guida
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Tanya Agurs-Collins
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Tim A Ahles
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | | | | | - Jorg Dietrich
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Rebecca Fuldner
- Division of Aging Biology, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Lisa Gallicchio
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Paige A Green
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | | | - Michelle C Janelsins
- Department of Surgery and Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
| | - Chamelli Jhappan
- Division of Cancer Biology, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Ronald Kohanski
- Division of Aging Biology, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Valter Longo
- University of Southern California, Los Angeles, California, USA
- IFOM Institute, Milan, Italy
| | - Simin Meydani
- Jean Mayer USDA Human Nutritional Research Center on Aging, Tufts University, Boston, MA, USA
| | - Supriya Mohile
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Laura J Niedernhofer
- Department of Biochemistry, Molecular Biology, and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Christian Nelson
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Frank Perna
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Keri Schadler
- Department of Pediatrics, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Jennifer A Schrack
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Russell P Tracy
- Departments of Pathology & Laboratory Medicine, and Biochemistry, Larner College of Medicine, University of Vermont, Colchester, VT, USA
| | | | - Kirsten K Ness
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
21
|
Marois G, Aktas A. Projecting health-ageing trajectories in Europe using a dynamic microsimulation model. Sci Rep 2021; 11:1785. [PMID: 33469046 PMCID: PMC7815779 DOI: 10.1038/s41598-021-81092-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 01/01/2021] [Indexed: 02/07/2023] Open
Abstract
The extent of the challenges and opportunities that population ageing presents depends heavily on the population's health. Hence, for the development of appropriate strategies that enable countries to adopt the emerging demographic and epidemiological realities, information on future health trajectories of elderly population is a natural requirement. This study presents an innovative methodological framework for projecting the health of individuals using a dynamic microsimulation model that considers interactions between sociodemographic characteristics, health, mortality, bio-medical and behavioral risk factors. The model developed, called ATHLOS-Mic, is used to project the health of cohorts born before 1960 for the period 2015-2060 for selected European Countries using SHARE data to illustrate the possible effects of some selected risk factors and education on future health trajectories. Results show that, driven by a better educational attainment, each generation will be healthier than the previous one at same age. Also, we see that, on average, an individual of our base population will live about 18 more years since the start of the projection period, but only 5 years in good health. Finally, we find that a scenario that removes the effect of having a low level of education on individual health has the largest impact on the projected average health, the average number of years lived per person, and the average number of years lived in good health.
Collapse
Affiliation(s)
- Guillaume Marois
- Asian Demographic Research Institute, School of Sociology and Political Sciences, Shanghai University, 99 Shangda Rd., Shanghai, 200444, China. .,Wittgenstein Centre for Demography and Global Human Capital (IIASA, OeAW, University of Vienna), International Institute for Applied Systems Analysis, Schlossplatz 1, 2361, Laxenburg, Austria.
| | - Arda Aktas
- Wittgenstein Centre for Demography and Global Human Capital (IIASA, OeAW, University of Vienna), International Institute for Applied Systems Analysis, Schlossplatz 1, 2361, Laxenburg, Austria
| |
Collapse
|
22
|
Singh P, Sivanandam TM, Konar A, Thakur MK. Role of nutraceuticals in cognition during aging and related disorders. Neurochem Int 2020; 143:104928. [PMID: 33285273 DOI: 10.1016/j.neuint.2020.104928] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/05/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022]
Abstract
Cognitive abilities are compromised with advancing age posing a great risk for the development of dementia and other related brain disorders. Genetic susceptibility as well as environmental exposures determine the fate of cognitive aging and its transition to pathological states. Emerging epidemiological and observational studies have revealed the importance of lifestyle factors including dietary patterns and nutritional intake in the maintenance of cognitive health and reducing the risk of neurodegenerative disorders. In this context, nutraceutical interventions have gained considerable attention in preventing age-related cognitive deficits and counteracting pathological processes. Nutraceuticals include dietary plants and derivatives, food supplements and processed foods with nutritional and pharmaceutical values. The present review highlights the importance of nutraceuticals in attenuating cognitive aging and its progression to dementia, with specific emphasis on chemical constituents, neurocognitive properties and mechanism of action.
Collapse
Affiliation(s)
- Padmanabh Singh
- Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Thamil Mani Sivanandam
- Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Arpita Konar
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India.
| | - M K Thakur
- Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
23
|
Fenyves BG, Arnold A, Gharat VG, Haab C, Tishinov K, Peter F, de Quervain D, Papassotiropoulos A, Stetak A. Dual Role of an mps-2/KCNE-Dependent Pathway in Long-Term Memory and Age-Dependent Memory Decline. Curr Biol 2020; 31:527-539.e7. [PMID: 33259792 DOI: 10.1016/j.cub.2020.10.069] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/14/2020] [Accepted: 10/21/2020] [Indexed: 01/24/2023]
Abstract
Activity-dependent persistent changes in neuronal intrinsic excitability and synaptic strength are underlying learning and memory. Voltage-gated potassium (Kv) channels are potential regulators of memory and may be linked to age-dependent neuronal disfunction. MinK-related peptides (MiRPs) are conserved transmembrane proteins modulating Kv channels; however, their possible role in the regulation of memory and age-dependent memory decline are unknown. Here, we show that, in C. elegans, mps-2 is the sole member of the MiRP family that controls exclusively long-term associative memory (LTAM) in AVA neuron. In addition, we demonstrate that mps-2 also plays a critical role in age-dependent memory decline. In young adult worms, mps-2 is transcriptionally upregulated by CRH-1/cyclic AMP (cAMP)-response-binding protein (CREB) during LTAM, although the mps-2 baseline expression is CREB independent and instead, during aging, relies on nhr-66, which acts as an age-dependent repressor. Deletion of nhr-66 or its binding element in the mps-2 promoter prevents age-dependent transcriptional repression of mps-2 and memory decline. Finally, MPS-2 acts through the modulation of the Kv2.1/KVS-3 and Kv2.2/KVS-4 heteromeric potassium channels. Altogether, we describe a conserved MPS-2/KVS-3/KVS-4 pathway essential for LTAM and also for a programmed control of physiological age-dependent memory decline.
Collapse
Affiliation(s)
- Bank G Fenyves
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Division of Molecular Neuroscience, Department of Psychology, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Department of Molecular Biology, Semmelweis University, Tűzoltó u. 37-47, 1094 Budapest, Hungary
| | - Andreas Arnold
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Division of Molecular Neuroscience, Department of Psychology, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland
| | - Vaibhav G Gharat
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Division of Molecular Neuroscience, Department of Psychology, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland
| | - Carmen Haab
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland
| | - Kiril Tishinov
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Fabian Peter
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Division of Molecular Neuroscience, Department of Psychology, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland
| | - Dominique de Quervain
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Division of Cognitive Neuroscience, Department of Psychology, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; University Psychiatric Clinics, University of Basel, Wilhelm Klein-Strasse 27, 4055 Basel, Switzerland
| | - Andreas Papassotiropoulos
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Division of Molecular Neuroscience, Department of Psychology, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Biozentrum, Life Sciences Training Facility, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland; University Psychiatric Clinics, University of Basel, Wilhelm Klein-Strasse 27, 4055 Basel, Switzerland
| | - Attila Stetak
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Division of Molecular Neuroscience, Department of Psychology, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; University Psychiatric Clinics, University of Basel, Wilhelm Klein-Strasse 27, 4055 Basel, Switzerland.
| |
Collapse
|
24
|
Konar A, Kalra RS, Chaudhary A, Nayak A, Guruprasad KP, Satyamoorthy K, Ishida Y, Terao K, Kaul SC, Wadhwa R. Identification of Caffeic Acid Phenethyl Ester (CAPE) as a Potent Neurodifferentiating Natural Compound That Improves Cognitive and Physiological Functions in Animal Models of Neurodegenerative Diseases. Front Aging Neurosci 2020; 12:561925. [PMID: 33244299 PMCID: PMC7685006 DOI: 10.3389/fnagi.2020.561925] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/14/2020] [Indexed: 12/22/2022] Open
Abstract
Cell-based screening of bioactive compounds has served as an important gateway in drug discovery. In the present report, using human neuroblastoma cells and enrolling an extensive three-step screening of 57 phytochemicals, we have identified caffeic acid phenethyl ester (CAPE) as a potent neurodifferentiating natural compound. Analyses of control and CAPE-induced neurodifferentiated cells revealed: (i) modulation of several key proteins (NF200, MAP-2, NeuN, PSD95, Tuj1, GAP43, and GFAP) involved in neurodifferentiation process; and (ii) attenuation of neuronal stemness (HOXD13, WNT3, and Msh-2) and proliferation-promoting (CDC-20, CDK-7, and BubR1) proteins. We anticipated that the neurodifferentiation potential of CAPE may be beneficial for the treatment of neurodegenerative diseases and tested it using the Drosophila model of Alzheimer’s disease (AD) and mice model of amnesia/loss of memory. In both models, CAPE exhibited improved disease symptoms and activation of physiological functions. Remarkably, CAPE-treated mice showed increased levels of neurotrophin-BDNF, neural progenitor marker-Nestin, and differentiation marker-NeuN, both in the cerebral cortex and hippocampus. Taken together, we demonstrate the differentiation-inducing and therapeutic potential of CAPE for neurodegenerative diseases.
Collapse
Affiliation(s)
- Arpita Konar
- AIST-INDIA DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan.,CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Rajkumar Singh Kalra
- AIST-INDIA DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Anupama Chaudhary
- AIST-INDIA DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Aashika Nayak
- DAILAB, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Kanive P Guruprasad
- DAILAB, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Kapaettu Satyamoorthy
- DAILAB, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal, India
| | | | | | - Sunil C Kaul
- AIST-INDIA DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan.,KAUL-Tech Co., Ltd., Tsuchiura, Japan
| | - Renu Wadhwa
- AIST-INDIA DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| |
Collapse
|
25
|
Qiao H, Chen M, Li S, Li Y, Sun Y, Wu Y. Poor lung function accelerates cognitive decline in middle-aged and older adults: Evidence from the English Longitudinal Study of Ageing. Arch Gerontol Geriatr 2020; 90:104129. [DOI: 10.1016/j.archger.2020.104129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/22/2020] [Accepted: 05/23/2020] [Indexed: 12/22/2022]
|
26
|
Zanini B, Simonetto A, Zubani M, Castellano M, Gilioli G. The Effects of Cow-Milk Protein Supplementation in Elderly Population: Systematic Review and Narrative Synthesis. Nutrients 2020; 12:E2548. [PMID: 32842497 PMCID: PMC7551861 DOI: 10.3390/nu12092548] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND To review currently available evidence on the effect of cow-milk proteins supplementation (CPS) on health in the elderly. METHODS Five electronic databases (Pubmed, Web of Science, Embase, Cochrane Library, ClinicalTrials.gov) were searched for studies about CPS among older people. All types of publications were included, with the exception of systematic reviews, meta-analyses, opinion letters, editorials, case reports, conference abstracts and comments. An additional search in Google Scholar and a manual review of the reference lists were performed. RESULTS Overall, 103 studies were included. Several studies explored the role of CPS in the preservation or improvement of muscle mass among healthy subjects (40 studies) and pre-frail, frail or sarcopenic patients (14), with evidence of beneficial effects. Other studies assessed the effect of CPS on bones (12), cardiovascular disease (8), inflamm-aging (7), chronic pulmonary disease (4), neurocognitive function (4), and vaccines (2), with weak evidence of positive effects. Seven studies in the field of protein metabolism investigated the role of CPS as an important contributor to nutritional needs. Other investigational areas are considered in the last five studies. CONCLUSIONS The beneficial effects of CPS in achieving aged-related nutritional goals, in preserving muscle mass and in recovering after hospitalization may be particularly relevant in the elderly.
Collapse
Affiliation(s)
- Barbara Zanini
- Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa, 11, I-25123 Brescia, Italy;
| | - Anna Simonetto
- AgroFood Lab, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa, 11, I-25123 Brescia, Italy; (A.S.); (M.Z.); (G.G.)
| | - Matilde Zubani
- AgroFood Lab, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa, 11, I-25123 Brescia, Italy; (A.S.); (M.Z.); (G.G.)
| | - Maurizio Castellano
- Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa, 11, I-25123 Brescia, Italy;
| | - Gianni Gilioli
- AgroFood Lab, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa, 11, I-25123 Brescia, Italy; (A.S.); (M.Z.); (G.G.)
| |
Collapse
|
27
|
Sadigh-Eteghad S, Vatandoust SM, Mahmoudi J, Rahigh Aghsan S, Majdi A. Cotinine ameliorates memory and learning impairment in senescent mice. Brain Res Bull 2020; 164:65-74. [PMID: 32818583 DOI: 10.1016/j.brainresbull.2020.08.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/29/2020] [Accepted: 08/10/2020] [Indexed: 10/23/2022]
Abstract
This study aimed to assess the effects of cotinine on age-induced memory and learning impairment and related downstream pathways in mice. Thirty aged (18-month old) and 10 young mice (8-week old) were randomly divided into 4 groups (n = 10 each) and subjected to cotinine at 5 mg/kg dose and/or methyllycaconitine (MLA) at 1 mg/kg, i.p. dose (α7 nAChRs antagonist) for 4 weeks. Morris water maze (MWM) and novel object recognition (NOR) tasks were used to assess spatial and recognition learning and memories of the mice, respectively. Levels of oxidative stress, apoptosis, neuroinflammation, and structural synaptic plasticity, and also neurotrophic factors and α7 nAChRs were assessed in the hippocampus using either ELISA or Western blotting. Aging was associated with learning and memory disabilities and dysregulation of the assessed pathways in the hippocampus of mice. Chronic cotinine treatment improved learning and memory in aged animals, indicated by decreased latency time, and increased time spent in the target quadrant and discrimination index (DI) in the MWM and NOR tasks. Also, chronic cotinine injection increased total antioxidant capacity (TAC), SOD and GSH-px activity, PSD-95, GAP-43, SYN, brain-derived neurotrophic factor, and neural growth factor levels and decreased malondialdehyde, TNF-α, and IL-1β in the hippocampus of aged mice. Conversely, MLA treatment reversed most of the mentioned effects via the blockade of α7 nAChRs. Cotinine improves age-induced memory and learning impairment via its modulatory effects on α7 nAChRs and subsequent activation/deactivation of the mentioned pathways in the hippocampus of aged mice.
Collapse
Affiliation(s)
- Saeed Sadigh-Eteghad
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Mehdi Vatandoust
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Rahigh Aghsan
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Majdi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
28
|
Behrens A, Graessel E, Pendergrass A, Donath C. Vitamin B-Can it prevent cognitive decline? A systematic review and meta-analysis. Syst Rev 2020; 9:111. [PMID: 32414424 PMCID: PMC7229605 DOI: 10.1186/s13643-020-01378-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 05/04/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Development of cognitive decline represents substantial issues in today's society, steadily gaining importance with increasing life expectancy. One potential approach to preventing cognitive decline is to lower homocysteine by administering vitamin B. In this systematic review and meta-analysis, we address this topic and investigate whether oral supplementation of vitamin B can successfully prevent cognitive decline in cognitively unimpaired individuals. METHODS A computerized systematic literature search was conducted using the electronic databases PubMed, Embase, and the Cochrane Library. Eligibility criteria included oral supplementation with vitamin B (B1, B6, folic acid, and B12) and the absence of cognitive impairment. A meta-analysis was conducted with "global cognition" as the primary outcome of this review. Secondary outcomes were changes in cognitive function in other cognitive domains reported in the included studies. Risk of bias was assessed according to the Cochrane Risk of Bias tool and the GRADE approach to establish the overall certainty of the evidence. RESULTS The meta-analysis did not yield a significant overall effect of supplementation with vitamin B on cognitive function (Z = 0.87; p = 0.39; SMD, 0.02; 95% CI, - 0.034, 0.08). A sensitivity analysis focusing on specific risk factors did not alter this result. Some studies reported isolated significant effects of the intervention on secondary outcomes. However, these findings were outnumbered by the number of cognitive tests that did not yield significant effects. DISCUSSION We found no overall evidence that oral vitamin B supplementation prevented cognitive decline. The isolated significant effects that were reported could be attributed to methodological issues. The results of this review do not provide evidence that population groups with certain risk factors would profit more from the intervention than others. Our findings do not apply to forms of administration other than oral supplementation nor do they offer information regarding the treatment of cognitively impaired individuals via the administration of vitamin B. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42017071692.
Collapse
Affiliation(s)
- Annika Behrens
- Center of Health Services Research in Medicine, Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany.
| | - Elmar Graessel
- Center of Health Services Research in Medicine, Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Anna Pendergrass
- Center of Health Services Research in Medicine, Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Carolin Donath
- Center of Health Services Research in Medicine, Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany
| |
Collapse
|
29
|
Soerensen M, Hozakowska-Roszkowska DM, Nygaard M, Larsen MJ, Schwämmle V, Christensen K, Christiansen L, Tan Q. A Genome-Wide Integrative Association Study of DNA Methylation and Gene Expression Data and Later Life Cognitive Functioning in Monozygotic Twins. Front Neurosci 2020; 14:233. [PMID: 32327964 PMCID: PMC7160301 DOI: 10.3389/fnins.2020.00233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/02/2020] [Indexed: 12/02/2022] Open
Abstract
Monozygotic twins are genetically identical but rarely phenotypically identical. Epigenetic and transcriptional variation could influence this phenotypic discordance. Investigation of intra-pair differences in molecular markers and a given phenotype in monozygotic twins controls most of the genetic contribution, enabling studies of the molecular features of the phenotype. This study aimed to identify genes associated with cognition in later life using integrated enrichment analyses of the results of blood-derived intra-pair epigenome-wide and transcriptome-wide association analyses of cognition in 452 middle-aged and old-aged monozygotic twins (56–80 years). Integrated analyses were performed with an unsupervised approach using KeyPathwayMiner, and a supervised approach using the KEGG and Reactome databases. The supervised approach identified several enriched gene sets, including “neuroactive ligand receptor interaction” (p-value = 1.62∗10-2), “Neurotrophin signaling” (p-value = 2.52∗10-3), “Alzheimer’s disease” (p-value = 1.20∗10-2), and “long-term depression” (p-value = 1.62∗10-2). The unsupervised approach resulted in a 238 gene network, including the Alzheimer’s disease gene APP (Amyloid Beta Precursor Protein) as an exception node, and several novel candidate genes. The strength of the unsupervised method is that it can reveal previously uncharacterized sub-pathways and detect interplay between biological processes, which remain undetected by the current supervised methods. In conclusion, this study identified several previously reported cognition genes and pathways and, additionally, puts forward novel candidates for further verification and validation.
Collapse
Affiliation(s)
- Mette Soerensen
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark.,Center for Individualized Medicine in Arterial Diseases, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark.,Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Dominika Marzena Hozakowska-Roszkowska
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark.,Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Marianne Nygaard
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark.,Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Martin J Larsen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark.,Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Veit Schwämmle
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Kaare Christensen
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark.,Department of Clinical Genetics, Odense University Hospital, Odense, Denmark.,Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Lene Christiansen
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark.,Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Qihua Tan
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark.,Department of Clinical Genetics, Odense University Hospital, Odense, Denmark.,Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
30
|
Balachandar R, Soundararajan S, Bagepally BS. Docosahexaenoic acid supplementation in age-related cognitive decline: a systematic review and meta-analysis. Eur J Clin Pharmacol 2020; 76:639-648. [PMID: 32060571 DOI: 10.1007/s00228-020-02843-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 02/01/2020] [Indexed: 12/27/2022]
Abstract
PURPOSE To investigate the role of DHA supplementation in preventing age-related cognitive decline (ARCD) in individual cognitive domains by conducting systematic review and meta-analysis. METHODS Relevant clinical trials were systematically searched at Medline, PubMed, Scopus, Cochrane, ProQuest, and Embase databases since inception to June 2018. The PRISMA guidelines were adhered for data abstraction, quality assessment, and validation of included studies. Study details such as participant characteristics, DHA supplementation, and cognitive function outcome measures, i.e., memory, attention, working memory, and executive function, were extracted to perform meta-analysis according to the Cochrane guidelines. Additional meta-regression and subgroup analyses were performed to detect confounding variables and sensitivity of results, respectively. RESULTS Ten studies including 2327 elderly individuals were part of the final results. Study exhibited minimal or no pooled incremental effects on memory (0.22, 95%CI = - 0.17 to 0.61, I2 = 94.36%), attention (0.1, 95%CI = - 0.04 to 0.25, I2 = 32.25%), working memory (0.01, 95%CI = - 0.10 to 0.12, I2 = 0%), and executive function (0.03, 95%CI = - 0.05 to 0.11, I2 = 78.48%) among the DHA-supplemented group. The results from standard mean difference between the groups, on memory (0.08, 95%CI = - 0.12 to 0.28, I2 = 76.82%), attention (0.04, 95%CI = - 0.09 to 0.23, I2 = 42.63%), working memory (- 0.08, 95%CI = - 0.26 to 0.10, I2 = 37.57%), and executive function (0.17, 95%CI = - 0.01 to 0.36, I2 = 78.48%) were similar to the results of pooled incremental analysis. Lastly, results remained unaffected by sensitivity and sub-group analyses. CONCLUSIONS Current pieces of evidence do not support the role of DHA supplementation, in preventing/retarding ARCD of memory, executive function, attention, and working memory. Protocol registered at PROSPERO (ID: PROSPERO 2018 CRD42018099401).
Collapse
Affiliation(s)
- Rakesh Balachandar
- Clinical Epidemiology, ICMR-National Institute of Occupational Health, Ahmedabad, Gujarat, India
| | - Soundarya Soundararajan
- Section on Human Psychopharmacology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda Maryland, USA
| | - Bhavani Shankara Bagepally
- Non-Communicable Diseases, ICMR-National Institute of Epidemiology, Ayapakkam, Chennai, Tamil Nadu, 600 077, India.
| |
Collapse
|
31
|
Guinobert I, Blondeau C, Colicchio B, Oudrhiri N, Dieterlen A, Jeandidier E, Deschenes G, Bardot V, Cotte C, Ripoche I, Carde P, Berthomier L, M’Kacher R. The Use of Natural Agents to Counteract Telomere Shortening: Effects of a Multi-Component Extract of Astragalus mongholicus Bunge and Danazol. Biomedicines 2020; 8:biomedicines8020031. [PMID: 32059353 PMCID: PMC7168059 DOI: 10.3390/biomedicines8020031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/16/2022] Open
Abstract
A link between telomere shortening and oxidative stress was found in aging people and patients with cancer or inflammatory diseases. Extracts of Astragalus spp. are known to stimulate telomerase activity, thereby compensating telomere shortening. We characterized a multi-component hydroethanolic root extract (HRE) of Astragalus mongholicus Bunge and assessed its effects on telomeres compared to those of danazol. Astragalosides I to IV, flavonoids, amino acids and sugars were detected in the HRE. Samples of peripheral blood lymphocytes with short telomeres from 18 healthy donors (mean age 63.5 years; range 32–86 years) were exposed to a single dose of 1 µg/mL HRE or danazol for three days. Telomere length and telomerase expression were then measured. Significant elongation of telomeres associated to a less toxicity was observed in lymphocytes from 13/18 donors following HRE treatment (0.54 kb (0.15–2.06 kb)) and in those from 9/18 donors after danazol treatment (0.95 kb (0.06–2.06 kb)). The rate of cells with short telomeres (<3 kb) decreased in lymphocytes from all donors after exposure to either HRE or danazol, telomere elongation being telomerase-dependent. These findings suggest that the HRE could be used for the management of age-related diseases.
Collapse
Affiliation(s)
- Isabelle Guinobert
- Groupe PiLeJe, 37 Quai de Grenelle, 75015 Paris Cedex 15, Naturopôle, Les Tiolans, 03800 Saint-Bonnet de Rochefort, France; (I.G.); (C.B.); (V.B.); (C.C.)
| | - Claude Blondeau
- Groupe PiLeJe, 37 Quai de Grenelle, 75015 Paris Cedex 15, Naturopôle, Les Tiolans, 03800 Saint-Bonnet de Rochefort, France; (I.G.); (C.B.); (V.B.); (C.C.)
| | - Bruno Colicchio
- IRIMAS, Institut de Recherche en Informatique, Mathématiques, Automatique et Signal, Université de Haute-Alsace, 68093 Mulhouse, France; (B.C.); (A.D.)
| | - Noufissa Oudrhiri
- Service d’Hématologie Moléculaire et Cytogénétique Paul Brousse CHU Paris Sud, Université Paris Sud, Inserm UMRS935, 94800 Villejuif, France;
| | - Alain Dieterlen
- IRIMAS, Institut de Recherche en Informatique, Mathématiques, Automatique et Signal, Université de Haute-Alsace, 68093 Mulhouse, France; (B.C.); (A.D.)
| | - Eric Jeandidier
- Service de Génétique Médicale, Groupe Hospitalier de la Région de Mulhouse et Sud-Alsace, 68070 Mulhouse, France;
| | - Georges Deschenes
- Service de Néphrologie, APHP-Hôpital Robert Debré, 75019 Paris, France;
| | - Valérie Bardot
- Groupe PiLeJe, 37 Quai de Grenelle, 75015 Paris Cedex 15, Naturopôle, Les Tiolans, 03800 Saint-Bonnet de Rochefort, France; (I.G.); (C.B.); (V.B.); (C.C.)
| | - César Cotte
- Groupe PiLeJe, 37 Quai de Grenelle, 75015 Paris Cedex 15, Naturopôle, Les Tiolans, 03800 Saint-Bonnet de Rochefort, France; (I.G.); (C.B.); (V.B.); (C.C.)
| | - Isabelle Ripoche
- Institut de Chimie de Clermont-Ferrand, Université Clermont Auvergne, CNRS, SIGMA Clermont, BP 10448, 63000 Clermont-Ferrand, France; (I.R.); (L.B.)
| | - Patrice Carde
- Département d’hématologie, Gustave Roussy Cancer Campus, université Paris Saclay, 94808 Villejuif, France;
| | - Lucile Berthomier
- Institut de Chimie de Clermont-Ferrand, Université Clermont Auvergne, CNRS, SIGMA Clermont, BP 10448, 63000 Clermont-Ferrand, France; (I.R.); (L.B.)
| | - Radhia M’Kacher
- Cell Environment, DNA damage R&D, 75020 Paris, France
- Correspondence: ; Tel.: +33-01-48-81-30-38
| |
Collapse
|
32
|
Ibañez S, Luebke JI, Chang W, Draguljić D, Weaver CM. Network Models Predict That Pyramidal Neuron Hyperexcitability and Synapse Loss in the dlPFC Lead to Age-Related Spatial Working Memory Impairment in Rhesus Monkeys. Front Comput Neurosci 2020; 13:89. [PMID: 32009920 PMCID: PMC6979278 DOI: 10.3389/fncom.2019.00089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/18/2019] [Indexed: 01/04/2023] Open
Abstract
Behavioral studies have shown spatial working memory impairment with aging in several animal species, including humans. Persistent activity of layer 3 pyramidal dorsolateral prefrontal cortex (dlPFC) neurons during delay periods of working memory tasks is important for encoding memory of the stimulus. In vitro studies have shown that these neurons undergo significant age-related structural and functional changes, but the extent to which these changes affect neural mechanisms underlying spatial working memory is not understood fully. Here, we confirm previous studies showing impairment on the Delayed Recognition Span Task in the spatial condition (DRSTsp), and increased in vitro action potential firing rates (hyperexcitability), across the adult life span of the rhesus monkey. We use a bump attractor model to predict how empirically observed changes in the aging dlPFC affect performance on the Delayed Response Task (DRT), and introduce a model of memory retention in the DRSTsp. Persistent activity-and, in turn, cognitive performance-in both models was affected much more by hyperexcitability of pyramidal neurons than by a loss of synapses. Our DRT simulations predict that additional changes to the network, such as increased firing of inhibitory interneurons, are needed to account for lower firing rates during the DRT with aging reported in vivo. Synaptic facilitation was an essential feature of the DRSTsp model, but it did not compensate fully for the effects of the other age-related changes on DRT performance. Modeling pyramidal neuron hyperexcitability and synapse loss simultaneously led to a partial recovery of function in both tasks, with the simulated level of DRSTsp impairment similar to that observed in aging monkeys. This modeling work integrates empirical data across multiple scales, from synapse counts to cognitive testing, to further our understanding of aging in non-human primates.
Collapse
Affiliation(s)
- Sara Ibañez
- Department of Mathematics, Franklin and Marshall College, Lancaster, PA, United States
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Jennifer I. Luebke
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Wayne Chang
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Danel Draguljić
- Department of Mathematics, Franklin and Marshall College, Lancaster, PA, United States
| | - Christina M. Weaver
- Department of Mathematics, Franklin and Marshall College, Lancaster, PA, United States
| |
Collapse
|
33
|
Kushwaha A, Thakur MK. Increase in hippocampal histone H3K9me3 is negatively correlated with memory in old male mice. Biogerontology 2019; 21:175-189. [DOI: 10.1007/s10522-019-09850-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/15/2019] [Indexed: 02/08/2023]
|
34
|
Gao T, Liu Y, Zhao Z, Luo Y, Wang L, Wang Y, Yin Y. L-655,708 Does not Prevent Isoflurane-induced Memory Deficits in Old Mice. Transl Neurosci 2019; 10:180-186. [PMID: 31410301 PMCID: PMC6689210 DOI: 10.1515/tnsci-2019-0032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 06/21/2019] [Indexed: 01/07/2023] Open
Abstract
Background General anesthesia and increasing age are two main risk factors for postoperative cognitive dysfunction (POCD). Effective agents for the prevention or treatment of POCD are urgently needed. L-655,708, an inverse agonist of α5 subunit-containing γ-aminobutyric acid subtype A (α5GABAA) receptors, can prevent anesthesia-induced memory deficits in young animals. However, there is a lack of evidence of its efficacy in old animals. Methodology Young (3- to 5-month-old) and old (18- to 20-month-old) mice were given an inhalation of 1.33% isoflurane for 1 hour and their associative memory was evaluated 24 hours after anesthesia using fear-conditioning tests (FCTs). To evaluate the effect of L-655,708, mice received intraperitoneal injections of L-655,708 (0.7 mg/kg) or vehicle 30 minutes before anesthesia. Results Old mice exhibited impaired memory and lower hippocampal α5GABAA levels than young mice under physiological conditions. Pre-injections of L-655,708 significantly alleviated isoflurane-induced memory decline in young mice, but not in old mice. Conclusions L-655,708 is not as effective for the prevention of POCD in old mice as it is in young mice. The use of inverse agonists of α5GABAA in preventing POCD in old patients should be carefully considered.
Collapse
Affiliation(s)
- Teng Gao
- Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Yue Liu
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Zifang Zhao
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yuan Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Lifang Wang
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Yiqing Yin
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, 100029, China
| |
Collapse
|
35
|
Shetty AK, Upadhya R, Madhu LN, Kodali M. Novel Insights on Systemic and Brain Aging, Stroke, Amyotrophic Lateral Sclerosis, and Alzheimer's Disease. Aging Dis 2019; 10:470-482. [PMID: 31011489 PMCID: PMC6457051 DOI: 10.14336/ad.2019.0330] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 03/30/2019] [Indexed: 12/11/2022] Open
Abstract
The mechanisms that underlie the pathophysiology of aging, amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD) and stroke are not fully understood and have been the focus of intense and constant investigation worldwide. Studies that provide insights on aging and age-related disease mechanisms are critical for advancing novel therapies that promote successful aging and prevent or cure multiple age-related diseases. The April 2019 issue of the journal, "Aging & Disease" published a series of articles that confer fresh insights on numerous age-related conditions and diseases. The age-related topics include the detrimental effect of overweight on energy metabolism and muscle integrity, senoinflammation as the cause of neuroinflammation, the link between systemic C-reactive protein and brain white matter loss, the role of miR-34a in promoting healthy heart and brain, the potential of sirtuin 3 for reducing cardiac and pulmonary fibrosis, and the promise of statin therapy for ameliorating asymptomatic intracranial atherosclerotic stenosis. Additional aging-related articles highlighted the involvement of miR-181b-5p and high mobility group box-1 in hypertension, Yes-associated protein in cataract formation, multiple miRs and long noncoding RNAs in coronary artery disease development, the role of higher meat consumption on sleep problems, and the link between glycated hemoglobin and depression. The topics related to ALS suggested that individuals with higher education and living in a rural environment have a higher risk for developing ALS, and collagen XIX alpha 1 is a prognostic biomarker of ALS. The topics discussed on AD implied that extracellular amyloid β42 is likely the cause of intraneuronal neurofibrillary tangle accumulation in familial AD and traditional oriental concoctions may be useful for slowing down the progression of AD. The article on stroke suggested that inhibition of the complement system is likely helpful in promoting brain repair after ischemic stroke. The significance of the above findings for understanding the pathogenesis in aging, ALS, AD, and stroke, slowing down the progression of aging, ALS and AD, and promoting brain repair after stroke are discussed.
Collapse
Affiliation(s)
- Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas, USA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas, USA
| | - Leelavathi N. Madhu
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas, USA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas, USA
| |
Collapse
|
36
|
Shetty AK, Kodali M, Upadhya R, Madhu LN. Emerging Anti-Aging Strategies - Scientific Basis and Efficacy. Aging Dis 2018; 9:1165-1184. [PMID: 30574426 PMCID: PMC6284760 DOI: 10.14336/ad.2018.1026] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022] Open
Abstract
The prevalence of age-related diseases is in an upward trend due to increased life expectancy in humans. Age-related conditions are among the leading causes of morbidity and death worldwide currently. Therefore, there is an urgent need to find apt interventions that slow down aging and reduce or postpone the incidence of debilitating age-related diseases. This review discusses the efficacy of emerging anti-aging approaches for maintaining better health in old age. There are many anti-aging strategies in development, which include procedures such as augmentation of autophagy, elimination of senescent cells, transfusion of plasma from young blood, intermittent fasting, enhancement of adult neurogenesis, physical exercise, antioxidant intake, and stem cell therapy. Multiple pre-clinical studies suggest that administration of autophagy enhancers, senolytic drugs, plasma from young blood, drugs that enhance neurogenesis and BDNF are promising approaches to sustain normal health during aging and also to postpone age-related neurodegenerative diseases such as Alzheimer's disease. Stem cell therapy has also shown promise for improving regeneration and function of the aged or Alzheimer's disease brain. Several of these approaches are awaiting critical appraisal in clinical trials to determine their long-term efficacy and possible adverse effects. On the other hand, procedures such as intermittent fasting, physical exercise, intake of antioxidants such as resveratrol and curcumin have shown considerable promise for improving function in aging, some of which are ready for large-scale clinical trials, as they are non-invasive, and seem to have minimal side effects. In summary, several approaches are at the forefront of becoming mainstream therapies for combating aging and postponing age-related diseases in the coming years.
Collapse
Affiliation(s)
- Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, Texas 76504, USA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, Texas 76504, USA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, Texas 76504, USA
| | - Leelavathi N. Madhu
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
| |
Collapse
|
37
|
Shah-Basak PP, Kielar A, Deschamps T, Verhoeff NP, Jokel R, Meltzer J. Spontaneous oscillatory markers of cognitive status in two forms of dementia. Hum Brain Mapp 2018; 40:1594-1607. [PMID: 30421472 DOI: 10.1002/hbm.24470] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/09/2018] [Accepted: 11/01/2018] [Indexed: 01/23/2023] Open
Abstract
Abnormal oscillatory brain activity in dementia may indicate incipient neuronal/synaptic dysfunction, rather than frank structural atrophy. Leveraging a potential link between the degree of abnormal oscillatory activity and cognitive symptom severity, one could localize brain regions in a diseased but pre-atrophic state, which may be more amenable to interventions. In the current study, we evaluated the relationships among cognitive deficits, regional volumetric changes, and resting-state magnetoencephalography abnormalities in patients with mild cognitive impairment (MCI; N = 10; age: 75.9 ± 7.3) or primary progressive aphasia (PPA; N = 12; 69.7 ± 8.0), and compared them to normal aging [young (N = 18; 24.6 ± 3.5), older controls (N = 24; 67.2 ± 9.7]. Whole-brain source-level resting-state estimates of relative oscillatory power in the delta (1-4 Hz), theta (4-7 Hz), alpha (8-12 Hz), and beta (15-30 Hz) bands were combined with gray matter volumes and cognitive scores to examine between-group differences and brain-behavior correlations. Language and executive function (EF) abilities were impaired in patients with PPA, while episodic memory was impaired in MCI. Widespread oscillatory speeding and volumetric shrinkage was associated with normal aging, whereas the trajectory in PPA indicated widespread oscillatory slowing with additional volumetric reductions. Increases in delta and decreases in alpha power uniquely predicted group membership to PPA. Beyond volumetric reductions, more delta predicted poorer memory. In patients with MCI, no consistent group difference among oscillatory measures was found. The contributions of delta/alpha power on memory abilities were larger than volumetric differences. Spontaneous oscillatory abnormalities in association with cognitive symptom severity can serve as a marker of neuronal dysfunction in dementia, providing targets for promising treatments.
Collapse
Affiliation(s)
- Priyanka P Shah-Basak
- Rotman Research Institute, Baycrest Health Sciences Centre, Toronto, Ontario, Canada.,Canadian Partnership for Stroke Recovery, Ottawa, Ontario, Canada
| | - Aneta Kielar
- Rotman Research Institute, Baycrest Health Sciences Centre, Toronto, Ontario, Canada.,Canadian Partnership for Stroke Recovery, Ottawa, Ontario, Canada.,Department of Speech, Language, and Hearing Sciences, University of Arizona, Tucson, Arizona
| | - Tiffany Deschamps
- Rotman Research Institute, Baycrest Health Sciences Centre, Toronto, Ontario, Canada
| | - Nicolaas Paul Verhoeff
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Department of Psychiatry, Baycrest Health Sciences, North York, Ontario, Canada
| | - Regina Jokel
- Rotman Research Institute, Baycrest Health Sciences Centre, Toronto, Ontario, Canada.,Department of Psychiatry, Baycrest Health Sciences, North York, Ontario, Canada.,Department of Speech-Language Pathology, University of Toronto, Toronto, Ontario, Canada
| | - Jed Meltzer
- Rotman Research Institute, Baycrest Health Sciences Centre, Toronto, Ontario, Canada.,Canadian Partnership for Stroke Recovery, Ottawa, Ontario, Canada.,Department of Speech-Language Pathology, University of Toronto, Toronto, Ontario, Canada.,Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
38
|
Farzaei MH, Bahramsoltani R, Abbasabadi Z, Braidy N, Nabavi SM. Role of green tea catechins in prevention of age-related cognitive decline: Pharmacological targets and clinical perspective. J Cell Physiol 2018; 234:2447-2459. [PMID: 30187490 DOI: 10.1002/jcp.27289] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 08/30/2018] [Indexed: 12/19/2022]
Abstract
Over the past decade, a wide range of scientific investigations have been performed to reveal neuropathological aspects of cognitive disorders; however, only limited therapeutic approaches currently exist. The failures of conventional therapeutic options as well as the predicted dramatic rise in the prevalence of cognitive decline in the coming future show the necessity for novel therapeutic agents. Recently, a wide range of research has focused on pharmacological activities of green tea catechins worldwide. Current investigations have clarified mechanistic effects of the catechins in inflammatory cascades, oxidative damages, different cellular transcription as well as transduction pathway in various body systems. It has been demonstrated that green tea polyphenols prevent age-related neurodegeneration through improvement of endogenous antioxidant defense mechanisms, modulation of neural growth factors, attenuation of neuroinflammatory pathway, and regulation of apoptosis. The catechins exhibited beneficial effects in cellular and animal models of neurodegenerative diseases including Alzheimer's disease, MS, and Parkinson's disease. The present review discusses the current pharmacological targets, which can be involved in the treatment of cognitive decline and addresses the action of catechin derivatives elicited from green tea on the multiple neural targets.
Collapse
Affiliation(s)
- Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Roodabeh Bahramsoltani
- Department of Pharmacy in Persian Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Abbasabadi
- Phyto Pharmacology Interest Group (PPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
39
|
Guo Y, Li H, Ke X, Deng M, Wu Z, Cai Y, Afewerky HK, Zhang X, Pei L, Lu Y. Degradation of Caytaxin Causes Learning and Memory Deficits via Activation of DAPK1 in Aging. Mol Neurobiol 2018; 56:3368-3379. [PMID: 30120735 DOI: 10.1007/s12035-018-1312-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/08/2018] [Indexed: 12/11/2022]
Abstract
Loss of memory is an inevitable clinic sign in aging, but its underlying mechanisms remain unclear. Here we show that death-associated protein kinase (DAPK1) is involved in the decays of learning and memory in aging via degradation of Caytaxin, a brain-specific member of BNIP-2. DAPK1 becomes activated in the hippocampus of mice during aging. Activation of DAPK1 is closely associated with degradation of Caytaxin protein. Silencing Caytaxin by the expression of small interfering RNA (siRNA) that targets specifically to Caytaxin in the hippocampus of adult mice impairs the learning and memory. Genetic inactivation of DAPK1 by deletion of DAPK1 kinase domain prevents the degradation of Caytaxin and protects against learning and memory declines. Thus, activation of DAPK1 impairs learning and memory by degrading Caytaxin during aging.
Collapse
Affiliation(s)
- Yu Guo
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hao Li
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao Ke
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Manfei Deng
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhuoze Wu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - You Cai
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Henok Kessete Afewerky
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Pathology and Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaoan Zhang
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lei Pei
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Department of Neurobiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.
| | - Youming Lu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China. .,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
40
|
Tobar E, Abedrapo MA, Godoy JA, Llanos JL, Díaz MJ, Azolas R, Bocic GR, Escobar JA, Cornejo RA, Romero CM. Impact of hypotension and global hypoperfusion in postoperative delirium: a pilot study in older adults undergoing open colon surgery. BRAZILIAN JOURNAL OF ANESTHESIOLOGY (ENGLISH EDITION) 2018. [PMID: 29287672 PMCID: PMC9391697 DOI: 10.1016/j.bjane.2017.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Background Post-operative delirium is a serious complication in patients undergoing major abdominal surgery. It remains unclear whether peri-operative hemodynamic and perfusion variables affect the risk for postoperative delirium. The objective of this pilot study was to evaluate the association between perfusion and hemodynamics peri-operative with the appearance of post-operative delirium. Methods Prospective cohort study of adults 60 years or older undergoing elective open colon surgery. Multimodal hemodynamic and perfusion variables were monitored, including central venous oxygenation (ScvO2), lactate levels, and non-invasive cerebral oxygenation (rSO2), according to a standard anesthesia protocol. Fisher's exact test or Student's t-test were used to compare patients who developed post-operative delirium with those who did not (p < 0.05). Results We studied 28 patients, age 73 ± 7 years, 60.7% female. Two patients developed post-operative delirium (7.1%). These two patients had fewer years of education than those without delirium (p = 0.031). None of the peri-operative blood pressure variables were associated with incidence of post-operative delirium. In terms of perfusion parameters, postoperative ScvO2 was lower in the delirium than the non-delirium group, without reaching statistical significance (65 ± 10% vs. 74 ± 5%; p = 0.08), but the delta-ScvO2 (the difference between means post-operative and intra-operative) was associated with post-operative delirium (p = 0.043). Post-operative lactate and rSO2 variables were not associated with delirium. Conclusions Our pilot study suggests an association between delta ScvO2 and post-operative delirium, and a tendency to lower post-operative ScvO2 in patients who developed delirium. Further studies are necessary to elucidate this association.
Collapse
Affiliation(s)
- Eduardo Tobar
- Hospital Clínico Universidad de Chile, Departamento de Medicina Interna Norte, Unidad de Pacientes Críticos, Santiago, Chile.
| | - Mario A Abedrapo
- Hospital Clínico Universidad de Chile, Departamento de Cirugía Norte, Equipo de Coloproctología, Santiago, Chile
| | - Jaime A Godoy
- Hospital Clínico Universidad de Chile, Departamento de Anestesiología e Reanimación, Santiago, Chile
| | - Jose L Llanos
- Hospital Clínico Universidad de Chile, Departamento de Cirugía Norte, Equipo de Coloproctología, Santiago, Chile
| | - Mauricio J Díaz
- Hospital Clínico Universidad de Chile, Departamento de Cirugía Norte, Equipo de Coloproctología, Santiago, Chile
| | - Rodrigo Azolas
- Hospital Clínico Universidad de Chile, Departamento de Cirugía Norte, Equipo de Coloproctología, Santiago, Chile
| | - Gunther R Bocic
- Hospital Clínico Universidad de Chile, Departamento de Cirugía Norte, Equipo de Coloproctología, Santiago, Chile
| | - Jaime A Escobar
- Hospital Clínico Universidad de Chile, Departamento de Anestesiología e Reanimación, Santiago, Chile
| | - Rodrigo A Cornejo
- Hospital Clínico Universidad de Chile, Departamento de Medicina Interna Norte, Unidad de Pacientes Críticos, Santiago, Chile
| | - Carlos M Romero
- Hospital Clínico Universidad de Chile, Departamento de Medicina Interna Norte, Unidad de Pacientes Críticos, Santiago, Chile
| |
Collapse
|
41
|
[Impact of hypotension and global hypoperfusion in postoperative delirium: a pilot study in older adults undergoing open colon surgery]. Rev Bras Anestesiol 2017; 68:135-141. [PMID: 29287672 DOI: 10.1016/j.bjan.2017.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 06/05/2017] [Accepted: 10/04/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Post-operative delirium is a serious complication in patients undergoing major abdominal surgery. It remains unclear whether peri-operative hemodynamic and perfusion variables affect the risk for postoperative delirium. The objective of this pilot study was to evaluate the association between perfusion and hemodynamics peri-operative with the appearance of post-operative delirium. METHODS Prospective cohort study of adults 60 years or older undergoing elective open colon surgery. Multimodal hemodynamic and perfusion variables were monitored, including central venous oxygenation (ScvO2), lactate levels, and non-invasive cerebral oxygenation (rSO2), according to a standard anesthesia protocol. Fisher's exact test or Student's t-test were used to compare patients who developed post-operative delirium with those who did not (p<0.05). RESULTS We studied 28 patients, age 73±7 years, 60.7% female. Two patients developed post-operative delirium (7.1%). These two patients had fewer years of education than those without delirium (p=0.031). None of the peri-operative blood pressure variables were associated with incidence of post-operative delirium. In terms of perfusion parameters, postoperative ScvO2 was lower in the delirium than the non-delirium group, without reaching statistical significance (65±10% vs. 74±5%; p=0.08), but the delta-ScvO2 (the difference between means post-operative and intra-operative) was associated with post-operative delirium (p=0.043). Post-operative lactate and rSO2 variables were not associated with delirium. CONCLUSIONS Our pilot study suggests an association between delta ScvO2 and post-operative delirium, and a tendency to lower post-operative ScvO2 in patients who developed delirium. Further studies are necessary to elucidate this association.
Collapse
|
42
|
Wang N, Ji S, Zhang H, Mei S, Qiao L, Jin X. Herba Cistanches: Anti-aging. Aging Dis 2017; 8:740-759. [PMID: 29344414 PMCID: PMC5758349 DOI: 10.14336/ad.2017.0720] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 07/20/2017] [Indexed: 12/11/2022] Open
Abstract
The Cistanche species ("Rou Cong Rong" in Chinese) is an endangered wild species growing in arid or semi-arid areas. The dried fleshy stem of Cistanches has been used as a tonic in China for many years. Modern pharmacological studies have since demonstrated that Herba Cistanches possesses broad medicinal functions, especially for use in anti-senescence, anti-oxidation, neuroprotection, anti-inflammation, hepatoprotection, immunomodulation, anti-neoplastic, anti-osteoporosis and the promotion of bone formation. This review summarizes the up-to-date and comprehensive information on Herba Cistanches covering the aspects of the botany, traditional uses, phytochemistry and pharmacology, to lay ground for fully elucidating the potential mechanisms of Herba Cistanches' anti-aging effect and promote its clinical application as an anti-aging herbal medicine.
Collapse
Affiliation(s)
- Ningqun Wang
- Department of Traditional Chinese Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Shaozhen Ji
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Hao Zhang
- Department of Radiology, Dongfang Hospital of Beijing University of Chinese Medicine, Beijing 100078, China
| | - Shanshan Mei
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Lumin Qiao
- Department of Emergency, Traditional Chinese Medicine Hospital of Yinchuan, Ningxia Hui Nationality Autonomous Region 750001, China.
| | - Xianglan Jin
- Department of Neurology, Dongfang Hospital of Beijing University of Chinese Medicine, Beijing 100078, China.
| |
Collapse
|
43
|
Xu Z, Feng W, Shen Q, Yu N, Yu K, Wang S, Chen Z, Shioda S, Guo Y. Rhizoma Coptidis and Berberine as a Natural Drug to Combat Aging and Aging-Related Diseases via Anti-Oxidation and AMPK Activation. Aging Dis 2017; 8:760-777. [PMID: 29344415 PMCID: PMC5758350 DOI: 10.14336/ad.2016.0620] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 06/20/2017] [Indexed: 12/25/2022] Open
Abstract
Aging is the greatest risk factor for human diseases, as it results in cellular growth arrest, impaired tissue function and metabolism, ultimately impacting life span. Two different mechanisms are thought to be primary causes of aging. One is cumulative DNA damage induced by a perpetuating cycle of oxidative stress; the other is nutrient-sensing adenosine monophosphate-activated protein kinase (AMPK) and rapamycin (mTOR)/ ribosomal protein S6 (rpS6) pathways. As the main bioactive component of natural Chinese medicine rhizoma coptidis (RC), berberine has recently been reported to expand life span in Drosophila melanogaster, and attenuate premature cellular senescence. Most components of RC including berberine, coptisine, palmatine, and jatrorrhizine have been found to have beneficial effects on hyperlipidemia, hyperglycemia and hypertension aging-related diseases. The mechanism of these effects involves multiple cellular kinase and signaling pathways, including anti-oxidation, activation of AMPK signaling and its downstream targets, including mTOR/rpS6, Sirtuin1/ forkhead box transcription factor O3 (FOXO3), nuclear factor erythroid-2 related factor-2 (Nrf2), nicotinamide adenine dinucleotide (NAD+) and nuclear factor-κB (NF-κB) pathways. Most of these mechanisms converge on AMPK regulation on mitochondrial oxidative stress. Therefore, such evidence supports the possibility that rhizoma coptidis, in particular berberine, is a promising anti-aging natural product, and has pharmaceutical potential in combating aging-related diseases via anti-oxidation and AMPK cellular kinase activation.
Collapse
Affiliation(s)
- Zhifang Xu
- 1Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.,2Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Wei Feng
- 3South Branch of Guang'anmen Hospital, China Academy of Chinese Medical Science, Beijing 102618, China
| | - Qian Shen
- 4Dongfang hospital, Beijing University of Chinese Medicine, Beijing 100078, China
| | - Nannan Yu
- 1Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Kun Yu
- 1Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Shenjun Wang
- 1Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.,2Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Zhigang Chen
- 4Dongfang hospital, Beijing University of Chinese Medicine, Beijing 100078, China
| | - Seiji Shioda
- 5Peptide Drug Innovation, Global Research Center for Innovative Life Science, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Shinagawa, Tokyo 142-8501, Japan
| | - Yi Guo
- 1Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.,2Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| |
Collapse
|
44
|
Singh P, Srivas S, Thakur MK. Epigenetic Regulation of Memory-Therapeutic Potential for Disorders. Curr Neuropharmacol 2017; 15:1208-1221. [PMID: 28393704 PMCID: PMC5725549 DOI: 10.2174/1570159x15666170404144522] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/03/2017] [Accepted: 03/25/2017] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Memory is a vital function which declines in different physiological and pathological conditions such as aging and neurodegenerative diseases. Research in the past has reported that memory formation and consolidation require the precise expression of synaptic plasticity genes. However, little is known about the regulation of these genes. Epigenetic modification is now a well established mechanism that regulates synaptic plasticity genes and neuronal functions including memory. Therefore, we have reviewed the epigenetic regulation of memory and its therapeutic potential for memory dysfunction during aging and neurological disorders. METHOD Research reports and online contents relevant to epigenetic regulation of memory during physiological and pathological conditions have been compiled and discussed. RESULTS Epigenetic modifications include mainly DNA methylation and hydroxymethylation, histone acetylation and methylation which involve chromatin modifying enzymes. These epigenetic marks change during memory formation and impairment due to dementia, aging and neurodegeneration. As the epigenetic modifications are reversible, they can be modulated by enzyme inhibitors leading to the recovery of memory. CONCLUSION Epigenetic modifications could be exploited as a potential therapeutic target to recover memory disorders during aging and pathological conditions.
Collapse
Affiliation(s)
- Padmanabh Singh
- Biochemistry and Molecular Biology Laboratory, Brain Research Centre, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221 005, India
| | - Sweta Srivas
- Biochemistry and Molecular Biology Laboratory, Brain Research Centre, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221 005, India
| | - M K Thakur
- Biochemistry and Molecular Biology Laboratory, Brain Research Centre, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221 005, India
| |
Collapse
|
45
|
Increased GSNOR Expression during Aging Impairs Cognitive Function and Decreases S-Nitrosation of CaMKIIα. J Neurosci 2017; 37:9741-9758. [PMID: 28883020 DOI: 10.1523/jneurosci.0681-17.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/27/2017] [Accepted: 08/03/2017] [Indexed: 11/21/2022] Open
Abstract
As the population ages, an increasing number of people suffer from age-related cognitive impairment. However, the mechanisms underlying this process remain unclear. Here, we found that S-nitrosoglutathione reductase (GSNOR), the key enzyme that metabolizes intracellular nitric oxide (NO) and regulates S-nitrosation, was significantly increased in the hippocampus of both aging humans and mice. Transgenic mice overexpressing GSNOR exclusively in neurons showed cognitive impairment in behavioral tests, including the Morris water maze, fear conditioning, and the Y-maze test. We also found that GSNOR transgenic mice have LTP defects and lower dendrite spine density, whereas GSNOR knock-out mice rescued the age-related cognitive impairment. Analysis of S-nitrosation showed significantly decreased hippocampal CaMKIIα S-nitrosation in naturally aged mice and GSNOR transgenic mice. Consistent with the change in CaMKIIα S-nitrosation, the accumulation of CaMKIIα in the hippocampal synaptosomal fraction, as well as its downstream signaling targets p(S831)-GLUR1, was also significantly decreased. All these effects could be rescued in the GSNOR knock-out mice. We further verified that the S-nitrosation of CaMKIIα was responsible for the CaMKIIα synaptosomal accumulation by mutating CaMKIIα S-nitrosated sites (C280/C289). Upregulation of the NO signaling pathway rescued the cognitive impairment in GSNOR transgenic mice. In summary, our research demonstrates that GSNOR impairs cognitive function in aging and it could serve as a new potential target for the treatment of age-related cognitive impairment. In contrast to the free radical theory of aging, NO signaling deficiency may be the main mediator of age-related cognitive impairment.SIGNIFICANCE STATEMENT This study indicated that S-nitrosoglutathione reductase (GSNOR), a key protein S-nitrosation metabolic enzyme, is a new potential target in age-related cognitive impairment; and in contrast to the free radical theory of aging, NO signaling deficiency may be the main cause of this process. In addition, increased GSNOR expression during aging decreases S-nitrosation of CaMKIIα and reduces CaMKIIα synaptosomal accumulation. To our knowledge, it is for the first time to show the cellular function regulation of CaMKIIα by GSNOR-dependent S-nitrosation as a new post-translational modification after its phosphorylation was explored. These findings elucidate a novel mechanism of age-related cognitive impairment and may provide a new potential target and strategy for slowing down this process.
Collapse
|
46
|
Li X, Ren C, Li S, Han R, Gao J, Huang Q, Jin K, Luo Y, Ji X. Limb Remote Ischemic Conditioning Promotes Myelination by Upregulating PTEN/Akt/mTOR Signaling Activities after Chronic Cerebral Hypoperfusion. Aging Dis 2017; 8:392-401. [PMID: 28840054 PMCID: PMC5524802 DOI: 10.14336/ad.2016.1227] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 12/27/2016] [Indexed: 12/31/2022] Open
Abstract
Limb Remote ischemic conditioning (LRIC) has been proved to be a promising neuroprotective method in white matter lesions after ischemia; however, its mechanism underlying protection after chronic cerebral hypoperfusion remains largely unknown. Here, we investigated whether LRIC promoted myelin growth by activating PI3K/Akt/mTOR signal pathway in a rat chronic hypoperfusion model. Thirty adult male Sprague Dawley underwent permanent double carotid artery (2VO), and limb remote ischemic conditioning was applied for 3 days after the 2VO surgery. Cognitive function, oligodendrocyte counts, myelin density, apoptosis and proliferation activity, as well as PTEN/Akt/mTOR signaling activity were determined 4 weeks after treatment. We found that LRIC significantly inhibited oligodendrocytes apoptosis (p<0.05), promoted myelination (p<0.01) in the corpus callosum and improved spatial learning impairment (p<0.05) at 4 weeks after chronic cerebral hypoperfusion. Oligodendrocytes proliferation, along with demyelination, in corpus callosum were not obviously affected by LRIC (p>0.05). Western blot analysis indicated that LRIC upregulated PTEN/Akt/mTOR signaling activities in corpus callosum (p<0.05). Our results suggest that LRIC exerts neuroprotective effect on white matter injuries through activating PTEN/Akt/mTOR signaling pathway after chronic cerebral hypoperfusion.
Collapse
Affiliation(s)
- Xiaohua Li
- 1Institute of Hypoxia Medicine, Xuanwu hospital, Capital Medical University, Beijing 100053, China
| | - Changhong Ren
- 1Institute of Hypoxia Medicine, Xuanwu hospital, Capital Medical University, Beijing 100053, China.,6Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Sijie Li
- 5Emergency department, Xuanwu hospital, Capital Medical University, Beijing 100053, China
| | - Rongrong Han
- 1Institute of Hypoxia Medicine, Xuanwu hospital, Capital Medical University, Beijing 100053, China
| | - Jinhuan Gao
- 1Institute of Hypoxia Medicine, Xuanwu hospital, Capital Medical University, Beijing 100053, China
| | - Qingjian Huang
- 1Institute of Hypoxia Medicine, Xuanwu hospital, Capital Medical University, Beijing 100053, China
| | - Kunlin Jin
- 4Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center at Fort Worth, Texas 76107, USA
| | - Yinghao Luo
- 2Department of China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xunming Ji
- 1Institute of Hypoxia Medicine, Xuanwu hospital, Capital Medical University, Beijing 100053, China.,3Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, Beijing, China.,6Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| |
Collapse
|
47
|
Metastability in Senescence. Trends Cogn Sci 2017; 21:509-521. [DOI: 10.1016/j.tics.2017.04.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 04/05/2017] [Accepted: 04/13/2017] [Indexed: 02/01/2023]
|
48
|
de Assis GG, de Almondes KM. Exercise-dependent BDNF as a Modulatory Factor for the Executive Processing of Individuals in Course of Cognitive Decline. A Systematic Review. Front Psychol 2017; 8:584. [PMID: 28469588 PMCID: PMC5395613 DOI: 10.3389/fpsyg.2017.00584] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 03/29/2017] [Indexed: 01/02/2023] Open
Abstract
Background: Aging naturally triggers a decline in cognition as result of deterioration in cerebral circuits, thus the executive functions (EFs) suffer changes that progress from mild to severe states of impairment. Exercise instead, works as a strategy for cognitive enhancement by modulating neuronal plasticity through the regulation of BDNF. However, whether the exercise-dependent BDNF may improve higher complexity processes such as the EFs is still in a studying process. Results: Current data on exercise-dependent BDNF changes for aging individuals in a course of cognitive impairment was summarized to investigate whether the exercise regulation of BDNF is effective to pronounce long term changes on executive controls. While the exercise-dependent regulation of BDNF is currently undeniable, the role of exercise dependent BDNF as a tool for the improvement of EFs in individuals with dementia is still less clear and seldom discussed. The summary of findings indicate a limited number of studies addressing exercise in order to discuss parameters related to either BDNF or executive functioning in such population conditions (n = 215), further narrowing to a total of 5 studies presenting analysis of both parameters. Nonetheless, positive outcomes from BDNF and EF variables were displayed by all the populations exposed to exercise across studies. Aerobic exercise was shown to be a major source for the enhancement of the BDNF-dependent executive functioning, when compared to cognitive stimulation. Moreover, the effect of exercise-dependent BDNF on domains of executive functioning appears to occur in a dose-dependent manner for the aging individuals, independently of cognitive condition.
Collapse
Affiliation(s)
- Gilmara G. de Assis
- Department of Psychology and Postgraduate Program in Psychobiology, Federal University of Rio Grande do NorteNatal, Brazil
| | - Katie Moraes de Almondes
- Department of Psychology and Postgraduate Program in Psychobiology, Federal University of Rio Grande do NorteNatal, Brazil
| |
Collapse
|
49
|
Singh P, Thakur MK. Histone Deacetylase 2 Inhibition Attenuates Downregulation of Hippocampal Plasticity Gene Expression during Aging. Mol Neurobiol 2017; 55:2432-2442. [PMID: 28364391 DOI: 10.1007/s12035-017-0490-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/14/2017] [Indexed: 12/31/2022]
Abstract
The brain undergoes several anatomical, biochemical, and molecular changes during aging, which subsequently result in downregulation of synaptic plasticity genes and decline of memory. However, the regulation of these genes during aging is not clearly understood. Previously, we reported that the expression of histone deacetylase (HDAC)2 was upregulated in the hippocampus of old mice and negatively correlated with the decline in recognition memory. As HDAC2 regulates key synaptic plasticity neuronal immediate early genes (IEGs), we have examined their expression and epigenetic regulation. We noted that the expression of neuronal IEGs decreased both at mRNA and protein level in the hippocampus of old mice. To explore the underlying regulation, we analyzed the binding of HDAC2 and level of histone acetylation at the promoter of neuronal IEGs. While the binding of HDAC2 was higher, H3K9 and H3K14 acetylation level was lower at the promoter of these genes in old as compared to young and adult mice. Further, we inhibited HDAC2 non-specifically by sodium butyrate and specifically by antisense oligonucleotide to recover epigenetic modification, expression of neuronal IEGs, and memory in old mice. Inhibition of HDAC2 increased histone H3K9 and H3K14 acetylation level at the promoter of neuronal IEGs, their expression, and recognition memory in old mice as compared to control. Thus, inhibition of HDAC2 can be used as a therapeutic target to recover decline in memory due to aging and associated neurological disorders.
Collapse
Affiliation(s)
- Padmanabh Singh
- Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221 005, India
| | - M K Thakur
- Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221 005, India.
| |
Collapse
|
50
|
Lin CH, Lin E, Lane HY. Genetic Biomarkers on Age-Related Cognitive Decline. Front Psychiatry 2017; 8:247. [PMID: 29209239 PMCID: PMC5702307 DOI: 10.3389/fpsyt.2017.00247] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/07/2017] [Indexed: 12/29/2022] Open
Abstract
With ever-increasing elder populations, age-related cognitive decline, which is characterized as a gradual decline in cognitive capacity in the aging process, has turned out to be a mammoth public health concern. Since genetic information has become increasingly important to explore the biological mechanisms of cognitive decline, the search for genetic biomarkers of cognitive aging has received much attention. There is growing evidence that single-nucleotide polymorphisms (SNPs) within the ADAMTS9, BDNF, CASS4, COMT, CR1, DNMT3A, DTNBP1, REST, SRR, TOMM40, circadian clock, and Alzheimer's diseases-associated genes may contribute to susceptibility to cognitive aging. In this review, we first illustrated evidence of the genetic contribution to disease susceptibility to age-related cognitive decline in recent studies ranging from approaches of candidate genes to genome-wide association studies. We then surveyed a variety of association studies regarding age-related cognitive decline with consideration of gene-gene and gene-environment interactions. Finally, we highlighted their limitations and future directions. In light of advances in precision medicine and multi-omics technologies, future research in genomic medicine promises to lead to innovative ideas that are relevant to disease prevention and novel drugs for cognitive aging.
Collapse
Affiliation(s)
- Chieh-Hsin Lin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Center for General Education, Cheng Shiu University, Kaohsiung, Taiwan
| | - Eugene Lin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Department of Electrical Engineering, University of Washington, Seattle, WA, United States.,TickleFish Systems Corporation, Seattle, WA, United States
| | - Hsien-Yuan Lane
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Department of Psychiatry, Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|