701
|
Bickford PC, Kaneko Y, Grimmig B, Pappas C, Small B, Sanberg CD, Sanberg PR, Tan J, Douglas Shytle R. Nutraceutical intervention reverses the negative effects of blood from aged rats on stem cells. AGE (DORDRECHT, NETHERLANDS) 2015; 37:103. [PMID: 26410618 PMCID: PMC5005857 DOI: 10.1007/s11357-015-9840-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/15/2015] [Indexed: 06/05/2023]
Abstract
Aging is associated with a decline in function in many of the stem cell niches of the body. An emerging body of literature suggests that one of the reasons for this decline in function is due to cell non-autonomous influences on the niche from the body. For example, studies using the technique of parabiosis have demonstrated a negative influence of blood from aged mice on muscle satellite cells and neurogenesis in young mice. We examined if we could reverse this effect of aged serum on stem cell proliferation by treating aged rats with NT-020, a dietary supplement containing blueberry, green tea, vitamin D3, and carnosine that has been shown to increase neurogenesis in aged rats. Young and aged rats were administered either control NIH-31 diet or one supplemented with NT-020 for 28 days, and serum was collected upon euthanasia. The serum was used in cultures of both rat hippocampal neural progenitor cells (NPCs) and rat bone marrow-derived mesenchymal stem cells (MSCs). Serum from aged rats significantly reduced cell proliferation as measured by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and 5-bromo-2'-deoxyuridine (BrdU) assays in both NPCs and MSCs. Serum from aged rats treated with NT-020 was not different from serum from young rats. Therefore, NT-020 rescued the effect of serum from aged rats to reduce stem cell proliferation.
Collapse
Affiliation(s)
- Paula C Bickford
- Research Service, James A Haley Veterans Affairs Hospital|, Tampa, FL, 33620, USA.
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, MDC-78, USF Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33620, USA.
| | - Yuji Kaneko
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, MDC-78, USF Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33620, USA
| | - Bethany Grimmig
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, MDC-78, USF Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33620, USA
- Department of Molecular Pharmacology and Physiology, USF Morsani College of Medicine, Tampa, FL, 33620, USA
| | | | - Brent Small
- School of Aging Studies, USF, Tampa, FL, 33620, USA
| | | | - Paul R Sanberg
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, MDC-78, USF Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33620, USA
| | - Jun Tan
- Department of Psychiatry, USF Morsani College of Medicine, Tampa, FL, 33620, USA
| | - R Douglas Shytle
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, MDC-78, USF Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33620, USA
| |
Collapse
|
702
|
Xiang Y, Bu XL, Liu YH, Zhu C, Shen LL, Jiao SS, Zhu XY, Giunta B, Tan J, Song WH, Zhou HD, Zhou XF, Wang YJ. Physiological amyloid-beta clearance in the periphery and its therapeutic potential for Alzheimer's disease. Acta Neuropathol 2015; 130:487-99. [PMID: 26363791 PMCID: PMC4575389 DOI: 10.1007/s00401-015-1477-1] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 08/27/2015] [Accepted: 09/05/2015] [Indexed: 12/31/2022]
Abstract
Amyloid-beta (Aβ) plays a pivotal role in the pathogenesis of Alzheimer’s disease (AD). The physiological capacity of peripheral tissues and organs in clearing brain-derived Aβ and its therapeutic potential for AD remains largely unknown. Here, we measured blood Aβ levels in different locations of the circulation in humans and mice, and used a parabiosis model to investigate the effect of peripheral Aβ catabolism on AD pathogenesis. We found that blood Aβ levels in the inferior/posterior vena cava were lower than that in the superior vena cava in both humans and mice. In addition, injected 125I labeled Aβ40 was located mostly in the liver, kidney, gastrointestinal tract, and skin but very little in the brain; suggesting that Aβ derived from the brain can be cleared in the periphery. Parabiosis before and after Aβ deposition in the brain significantly reduced brain Aβ burden without alterations in the expression of amyloid precursor protein, Aβ generating and degrading enzymes, Aβ transport receptors, and AD-type pathologies including hyperphosphorylated tau, neuroinflammation, as well as neuronal degeneration and loss in the brains of parabiotic AD mice. Our study revealed that the peripheral system is potent in clearing brain Aβ and preventing AD pathogenesis. The present work suggests that peripheral Aβ clearance is a valid therapeutic approach for AD, and implies that deficits in the Aβ clearance in the periphery might also contribute to AD pathogenesis.
Collapse
Affiliation(s)
- Yang Xiang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing, China
| | - Xian-Le Bu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing, China
| | - Yu-Hui Liu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing, China
| | - Chi Zhu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing, China
| | - Lin-Lin Shen
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing, China
| | - Shu-Sheng Jiao
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing, China
| | - Xiao-Yan Zhu
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Brian Giunta
- Neuroimmunology Laboratory, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Jun Tan
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Wei-Hong Song
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC, Canada
| | - Hua-Dong Zhou
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing, China
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences and Sansom Institute, University of South Australia, Adelaide, SA, Australia
| | - Yan-Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing, China.
| |
Collapse
|
703
|
Zhang R, Chen HZ, Liu DP. The Four Layers of Aging. Cell Syst 2015; 1:180-6. [PMID: 27135911 DOI: 10.1016/j.cels.2015.09.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 07/06/2015] [Accepted: 09/04/2015] [Indexed: 02/06/2023]
Abstract
Instead of considering aging in terms of discrete hallmarks, we suggest that it operates in four layers, each at a different biological scale. Malfunctions within each layer-and connections between them-produce the aged phenotype and its associated susceptibility to disease.
Collapse
Affiliation(s)
- Ran Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, P.R. China
| | - Hou-Zao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, P.R. China.
| | - De-Pei Liu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, P.R. China.
| |
Collapse
|
704
|
Kell DB, Pretorius E. The simultaneous occurrence of both hypercoagulability and hypofibrinolysis in blood and serum during systemic inflammation, and the roles of iron and fibrin(ogen). Integr Biol (Camb) 2015; 7:24-52. [PMID: 25335120 DOI: 10.1039/c4ib00173g] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Although the two phenomena are usually studied separately, we summarise a considerable body of literature to the effect that a great many diseases involve (or are accompanied by) both an increased tendency for blood to clot (hypercoagulability) and the resistance of the clots so formed (hypofibrinolysis) to the typical, 'healthy' or physiological lysis. We concentrate here on the terminal stages of fibrin formation from fibrinogen, as catalysed by thrombin. Hypercoagulability goes hand in hand with inflammation, and is strongly influenced by the fibrinogen concentration (and vice versa); this can be mediated via interleukin-6. Poorly liganded iron is a significant feature of inflammatory diseases, and hypofibrinolysis may change as a result of changes in the structure and morphology of the clot, which may be mimicked in vitro, and may be caused in vivo, by the presence of unliganded iron interacting with fibrin(ogen) during clot formation. Many of these phenomena are probably caused by electrostatic changes in the iron-fibrinogen system, though hydroxyl radical (OH˙) formation can also contribute under both acute and (more especially) chronic conditions. Many substances are known to affect the nature of fibrin polymerised from fibrinogen, such that this might be seen as a kind of bellwether for human or plasma health. Overall, our analysis demonstrates the commonalities underpinning a variety of pathologies as seen in both hypercoagulability and hypofibrinolysis, and offers opportunities for both diagnostics and therapies.
Collapse
Affiliation(s)
- Douglas B Kell
- School of Chemistry and The Manchester Institute of Biotechnology, The University of Manchester, 131, Princess St, Manchester M1 7DN, Lancs, UK.
| | | |
Collapse
|
705
|
Müller L, Pawelec G. As we age: Does slippage of quality control in the immune system lead to collateral damage? Ageing Res Rev 2015; 23:116-23. [PMID: 25676139 DOI: 10.1016/j.arr.2015.01.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 01/16/2015] [Accepted: 01/26/2015] [Indexed: 12/22/2022]
Abstract
The vertebrate adaptive immune system is remarkable for its possession of a very broad range of antigen receptors imbuing the system with exquisite specificity, in addition to the phagocytic and inflammatory cells of the innate system shared with invertebrates. This system requires strict control both at the level of the generation the cells carrying these receptors and at the level of their activation and effector function mediation in order to avoid autoimmunity and mitigate immune pathology. Thus, quality control checkpoints are built into the system at multiple nodes in the response, relying on clonal selection and regulatory networks to maximize pathogen-directed effects and minimize collateral tissue damage. However, these checkpoints are compromised with age, resulting in poorer immune control manifesting as tissue-damaging autoimmune and inflammatory phenomena which can cause widespread systemic disease, paradoxically compounding the problems associated with increased susceptibility to infectious disease and possibly cancer in the elderly. Better understanding the reasons for slippage of immune control will pave the way for developing rational strategies for interventions to maintain appropriate immunity while reducing immunopathology.
Collapse
|
706
|
Mendelsohn AR, Larrick JW. Systemic factors mediate reversible age-associated brain dysfunction. Rejuvenation Res 2015; 17:525-8. [PMID: 25400086 DOI: 10.1089/rej.2014.1643] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Brain function declines in aging mammals. Recent work has identified dysregulation of key blood-borne factors whose altered expression during aging diminishes brain function in mice. Increased C-C motif chemokine 11 (CCL11) expression with aging is detrimental to brain function. On the other hand, plasma levels of the trophic factor growth/differentiation factor 11 (GDF11) decrease with aging. Restoration of youthful levels of GDF11 by injection partially restores brain function and neurogenesis by improving endothelial cell function and vasculature. Moreover, GDF11 has a rejuvenative effect on cardiac and skeletal muscle. Decreased type II interferon (IFN-II) and increased type I interferon (IFN-I) signaling during aging at the choroid plexus (CP), which constitutes the brain-cerebrospinal fluid barrier (B-CSF-B), negatively effects brain function. Blood from young mice contains factors that restore IFN-II levels. IFN-II is required for maintenance of the CP, and low IFN-II levels are associated with decreased cognitive abilities. IFN-I levels appear to drive increased CCL11 expression through the CSF. Blood from young animals does not restore IFN-I levels. However, injecting anti-interferon-α/β receptor (IFNAR) antibodies into the CSF inhibits downstream IFN-I gene and protein expression and decreases expression of CCL11, partially restoring neurogenesis and cognitive function. These results suggest that IFN-I plays a critical role in increasing CCL11 during aging of the brain. An emerging theme is that aging-associated loss of function in mammals may involve a set of defined, potentially reversible changes in many tissues and organs, including the brain, permitting development of potential rejuvenative therapies.
Collapse
Affiliation(s)
- Andrew R Mendelsohn
- Panorama Research Institute and Regenerative Sciences Institute , Sunnyvale, California
| | | |
Collapse
|
707
|
Rachul CM, Percec I, Caulfield T. The Fountain of Stem Cell-Based Youth? Online Portrayals of Anti-Aging Stem Cell Technologies. Aesthet Surg J 2015; 35:730-6. [PMID: 25922365 DOI: 10.1093/asj/sju111] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2014] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The hype surrounding stem cell science has created a market opportunity for the cosmetic industry. Cosmetic and anti-aging products and treatments that make claims regarding stem cell technology are increasingly popular, despite a lack of evidence for safety and efficacy of such products. OBJECTIVES This study explores how stem cell-based products and services are portrayed to the public through online sources, in order to gain insight into the key messages available to consumers. METHODS A content analysis of 100 web pages was conducted to examine the portrayals of stem cell-based cosmetic and anti-aging products and treatments. A qualitative discourse analysis of one web page further examined how language contributes to the portrayals of these products and treatments to public audiences. RESULTS The majority of web pages portrayed stem cell-based products as ready for public use. Very few web pages substantiated claims with scientific evidence, and even fewer mentioned any risks or limitations associated with stem cell science. The discourse analysis revealed that the framing and use of metaphor obscures the certainty of the efficacy of and length of time for stem cell-based anti-aging technology to be publicly available. CONCLUSIONS This study highlights the need to educate patients and the public on the current limits of stem cell applications in this context. In addition, generating scientific evidence for stem cell-based anti-aging and aesthetic applications is needed for optimizing benefits and minimizing adverse effects for the public. Having more evidence on efficacy and risks will help to protect patients who are eagerly seeking out these treatments.
Collapse
Affiliation(s)
- Christen M Rachul
- Ms Rachul is a PhD Candidate in Applied Linguistics and Language Studies, School of Linguistics and Language Studies, Carleton University, Ottawa, ON, Canada. Dr Percec is an Assistant Professor, Division of Plastic Surgery, Epigenetics Program, University of Pennsylvania, Philadelphia, PA. Professor Caulfield is the Canada Research Chair in Health Law and Policy; Trudeau Fellow and Professor, Faculty of Law and School of Public Health; and Research Director, Health Law Institute, University of Alberta, Edmonton, AB, Canada
| | - Ivona Percec
- Ms Rachul is a PhD Candidate in Applied Linguistics and Language Studies, School of Linguistics and Language Studies, Carleton University, Ottawa, ON, Canada. Dr Percec is an Assistant Professor, Division of Plastic Surgery, Epigenetics Program, University of Pennsylvania, Philadelphia, PA. Professor Caulfield is the Canada Research Chair in Health Law and Policy; Trudeau Fellow and Professor, Faculty of Law and School of Public Health; and Research Director, Health Law Institute, University of Alberta, Edmonton, AB, Canada
| | - Timothy Caulfield
- Ms Rachul is a PhD Candidate in Applied Linguistics and Language Studies, School of Linguistics and Language Studies, Carleton University, Ottawa, ON, Canada. Dr Percec is an Assistant Professor, Division of Plastic Surgery, Epigenetics Program, University of Pennsylvania, Philadelphia, PA. Professor Caulfield is the Canada Research Chair in Health Law and Policy; Trudeau Fellow and Professor, Faculty of Law and School of Public Health; and Research Director, Health Law Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
708
|
Abstract
The past two decades of research into the pathogenesis of Alzheimer disease (AD) have been driven largely by the amyloid hypothesis; the neuroinflammation that is associated with AD has been assumed to be merely a response to pathophysiological events. However, new data from preclinical and clinical studies have established that immune system-mediated actions in fact contribute to and drive AD pathogenesis. These insights have suggested both novel and well-defined potential therapeutic targets for AD, including microglia and several cytokines. In addition, as inflammation in AD primarily concerns the innate immune system - unlike in 'typical' neuroinflammatory diseases such as multiple sclerosis and encephalitides - the concept of neuroinflammation in AD may need refinement.
Collapse
|
709
|
Li X, Long J, He T, Belshaw R, Scott J. Integrated genomic approaches identify major pathways and upstream regulators in late onset Alzheimer's disease. Sci Rep 2015. [PMID: 26202100 PMCID: PMC4511863 DOI: 10.1038/srep12393] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Previous studies have evaluated gene expression in Alzheimer’s disease (AD) brains to identify mechanistic processes, but have been limited by the size of the datasets studied. Here we have implemented a novel meta-analysis approach to identify differentially expressed genes (DEGs) in published datasets comprising 450 late onset AD (LOAD) brains and 212 controls. We found 3124 DEGs, many of which were highly correlated with Braak stage and cerebral atrophy. Pathway Analysis revealed the most perturbed pathways to be (a) nitric oxide and reactive oxygen species in macrophages (NOROS), (b) NFkB and (c) mitochondrial dysfunction. NOROS was also up-regulated, and mitochondrial dysfunction down-regulated, in healthy ageing subjects. Upstream regulator analysis predicted the TLR4 ligands, STAT3 and NFKBIA, for activated pathways and RICTOR for mitochondrial genes. Protein-protein interaction network analysis emphasised the role of NFKB; identified a key interaction of CLU with complement; and linked TYROBP, TREM2 and DOK3 to modulation of LPS signalling through TLR4 and to phosphatidylinositol metabolism. We suggest that NEUROD6, ZCCHC17, PPEF1 and MANBAL are potentially implicated in LOAD, with predicted links to calcium signalling and protein mannosylation. Our study demonstrates a highly injurious combination of TLR4-mediated NFKB signalling, NOROS inflammatory pathway activation, and mitochondrial dysfunction in LOAD.
Collapse
Affiliation(s)
- Xinzhong Li
- Centre for Biostatistics, Bioinformatics and Biomarkers, Plymouth University, Plymouth UK
| | - Jintao Long
- Centre for Biostatistics, Bioinformatics and Biomarkers, Plymouth University, Plymouth UK
| | - Taigang He
- Institute of Cardiovascular and Cell Sciences, St. George University, London UK
| | - Robert Belshaw
- School of Biomedicine and Healthcare Sciences, Plymouth University, Plymouth UK
| | - James Scott
- National Heart and Lung Institute, Imperial College, London UK
| |
Collapse
|
710
|
Smith LK, He Y, Park JS, Bieri G, Snethlage CE, Lin K, Gontier G, Wabl R, Plambeck KE, Udeochu J, Wheatley EG, Bouchard J, Eggel A, Narasimha R, Grant JL, Luo J, Wyss-Coray T, Villeda SA. β2-microglobulin is a systemic pro-aging factor that impairs cognitive function and neurogenesis. Nat Med 2015; 21:932-7. [PMID: 26147761 PMCID: PMC4529371 DOI: 10.1038/nm.3898] [Citation(s) in RCA: 349] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 06/08/2015] [Indexed: 12/18/2022]
Abstract
Aging drives cognitive and regenerative impairments in the adult brain, increasing susceptibility to neurodegenerative disorders in healthy individuals. Experiments using heterochronic parabiosis, in which the circulatory systems of young and old animals are joined, indicate that circulating pro-aging factors in old blood drive aging phenotypes in the brain. Here we identify β2-microglobulin (B2M), a component of major histocompatibility complex class 1 (MHC I) molecules, as a circulating factor that negatively regulates cognitive and regenerative function in the adult hippocampus in an age-dependent manner. B2M is elevated in the blood of aging humans and mice, and it is increased within the hippocampus of aged mice and young heterochronic parabionts. Exogenous B2M injected systemically, or locally in the hippocampus, impairs hippocampal-dependent cognitive function and neurogenesis in young mice. The negative effects of B2M and heterochronic parabiosis are, in part, mitigated in the hippocampus of young transporter associated with antigen processing 1 (Tap1)-deficient mice with reduced cell surface expression of MHC I. The absence of endogenous B2M expression abrogates age-related cognitive decline and enhances neurogenesis in aged mice. Our data indicate that systemic B2M accumulation in aging blood promotes age-related cognitive dysfunction and impairs neurogenesis, in part via MHC I, suggesting that B2M may be targeted therapeutically in old age.
Collapse
Affiliation(s)
- Lucas K Smith
- 1] Department of Anatomy, University of California San Francisco, San Francisco, California, USA. [2] The Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, San Francisco, California, USA. [3] Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, California, USA
| | - Yingbo He
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Jeong-Soo Park
- 1] Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA. [2] Department of Biochemistry, Dankook University College of Medicine, Cheonan, Korea
| | - Gregor Bieri
- 1] Department of Anatomy, University of California San Francisco, San Francisco, California, USA. [2] The Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, San Francisco, California, USA. [3] Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA. [4] Neuroscience Graduate Program, Stanford University School of Medicine, Stanford, California, USA
| | - Cedric E Snethlage
- 1] Department of Anatomy, University of California San Francisco, San Francisco, California, USA. [2] The Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, San Francisco, California, USA
| | - Karin Lin
- 1] Department of Anatomy, University of California San Francisco, San Francisco, California, USA. [2] The Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, San Francisco, California, USA. [3] Neuroscience Graduate Program, University of California San Francisco, San Francisco, California, USA
| | - Geraldine Gontier
- 1] Department of Anatomy, University of California San Francisco, San Francisco, California, USA. [2] The Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, San Francisco, California, USA
| | - Rafael Wabl
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Kristopher E Plambeck
- 1] Department of Anatomy, University of California San Francisco, San Francisco, California, USA. [2] The Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, San Francisco, California, USA
| | - Joe Udeochu
- 1] Department of Anatomy, University of California San Francisco, San Francisco, California, USA. [2] The Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, San Francisco, California, USA. [3] Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, California, USA
| | - Elizabeth G Wheatley
- 1] Department of Anatomy, University of California San Francisco, San Francisco, California, USA. [2] The Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, San Francisco, California, USA. [3] Developmental and Stem Cell Biology Graduate Program, University of California San Francisco, San Francisco, California, USA
| | - Jill Bouchard
- 1] Department of Anatomy, University of California San Francisco, San Francisco, California, USA. [2] The Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, San Francisco, California, USA
| | - Alexander Eggel
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland
| | - Ramya Narasimha
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Jacqueline L Grant
- 1] Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA. [2] Neuroscience Graduate Program, Stanford University School of Medicine, Stanford, California, USA
| | - Jian Luo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Tony Wyss-Coray
- 1] Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA. [2] Neuroscience Graduate Program, Stanford University School of Medicine, Stanford, California, USA. [3] Center for Tissue Regeneration, Repair and Restoration, Veterans' Affairs (VA) Palo Alto Health Care System, Palo Alto, California, USA
| | - Saul A Villeda
- 1] Department of Anatomy, University of California San Francisco, San Francisco, California, USA. [2] The Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, San Francisco, California, USA. [3] Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, California, USA. [4] Neuroscience Graduate Program, University of California San Francisco, San Francisco, California, USA. [5] Developmental and Stem Cell Biology Graduate Program, University of California San Francisco, San Francisco, California, USA. [6] California Institute for Quantitative Biosciences (QB3), San Francisco, California, USA
| |
Collapse
|
711
|
Thieme R, Kurz S, Kolb M, Debebe T, Holtze S, Morhart M, Huse K, Szafranski K, Platzer M, Hildebrandt TB, Birkenmeier G. Analysis of Alpha-2 Macroglobulin from the Long-Lived and Cancer-Resistant Naked Mole-Rat and Human Plasma. PLoS One 2015; 10:e0130470. [PMID: 26103567 PMCID: PMC4477878 DOI: 10.1371/journal.pone.0130470] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/20/2015] [Indexed: 01/18/2023] Open
Abstract
Background The naked mole-rat (NMR) is a long-lived and cancer resistant species. Identification of potential anti-cancer and age related mechanisms is of great interest and makes this species eminent to investigate anti-cancer strategies and understand aging mechanisms. Since it is known that the NMR expresses higher liver mRNA-levels of alpha 2-macroglobulin than mice, nothing is known about its structure, functionality or expression level in the NMR compared to the human A2M. Results Here we show a comprehensive analysis of NMR- and human plasma-A2M, showing a different prediction in glycosylation of NMR-A2M, which results in a higher molecular weight compared to human A2M. Additionally, we found a higher concentration of A2M (8.3±0.44 mg/mL vs. and 4.4±0.20 mg/mL) and a lower total plasma protein content (38.7±1.79 mg/mL vs. 61.7±3.20 mg/mL) in NMR compared to human. NMR-A2M can be transformed by methylamine and trypsin resulting in a conformational change similar to human A2M. NMR-A2M is detectable by a polyclonal antibody against human A2M. Determination of tryptic and anti-tryptic activity of NMR and human plasma revealed a higher anti-tryptic activity of the NMR plasma. On the other hand, less proteolytic activity was found in NMR plasma compared to human plasma. Conclusion We found transformed NMR-A2M binding to its specific receptor LRP1. We could demonstrate lower protein expression of LRP1 in the NMR liver tissue compared to human but higher expression of A2M. This was accompanied by a higher EpCAM protein expression as central adhesion molecule in cancer progression. NMR-plasma was capable to increase the adhesion in human fibroblast in vitro most probably by increasing CD29 protein expression. This is the first report, demonstrating similarities as well as distinct differences between A2M in NMR and human plasma. This might be directly linked to the intriguing phenotype of the NMR and suggests that A2M might probably play an important role in anti-cancer and the anti-aging mechanisms in the NMR.
Collapse
Affiliation(s)
- René Thieme
- Institute of Biochemistry, University of Leipzig, Leipzig, Germany
| | - Susanne Kurz
- Institute of Biochemistry, University of Leipzig, Leipzig, Germany
| | - Marlen Kolb
- Institute of Biochemistry, University of Leipzig, Leipzig, Germany
| | - Tewodros Debebe
- Institute of Biochemistry, University of Leipzig, Leipzig, Germany
| | - Susanne Holtze
- Department of Reproduction Management, Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | - Michaela Morhart
- Department of Reproduction Management, Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | - Klaus Huse
- Fritz Lipmann Institute–Leibniz Institute for Age Research, Jena, Germany
| | - Karol Szafranski
- Fritz Lipmann Institute–Leibniz Institute for Age Research, Jena, Germany
| | - Matthias Platzer
- Fritz Lipmann Institute–Leibniz Institute for Age Research, Jena, Germany
| | - Thomas B. Hildebrandt
- Department of Reproduction Management, Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | - Gerd Birkenmeier
- Institute of Biochemistry, University of Leipzig, Leipzig, Germany
- * E-mail:
| |
Collapse
|
712
|
Abstract
In the past 15 years, the field of physiology has been radically challenged by landmark studies using novel tools of genetic engineering. Particular to our interest, the reciprocal interactions between the skeleton and the nervous system were shown to be major ones. The demonstration that brain, via multiple pathways, is a powerful regulator of bone growth, has shed light on an important central regulation of skeletal homeostasis. More recently, it was shown that bone might return the favor to the brain through the secretion of a bone-derived hormone, osteocalcin. The skeleton influences development and cognitive functions of the central nervous system at different stages throughout life suggesting an intimate dialogue between bone and brain.
Collapse
Affiliation(s)
- Alexandre Chamouni
- Centre de Médecine Moléculaire, Institut Necker-Enfants Malades (INEM), 75014, Paris, France
| | | | | |
Collapse
|
713
|
Barreiro E, Sznajder JI, Nader GA, Budinger GRS. Muscle dysfunction in patients with lung diseases: a growing epidemic. Am J Respir Crit Care Med 2015; 191:616-9. [PMID: 25767924 DOI: 10.1164/rccm.201412-2189oe] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Esther Barreiro
- 1 Respiratory Medicine Department-Lung Cancer Research Group, Institute of Medical Research of Hospital del Mar (IMIM)-Hospital del Mar, Parc de Salut Mar, Barcelona Biomedical Research Park, Barcelona, Spain
| | | | | | | |
Collapse
|
714
|
Sankaran VG, Weiss MJ. Anemia: progress in molecular mechanisms and therapies. Nat Med 2015; 21:221-30. [PMID: 25742458 DOI: 10.1038/nm.3814] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 02/04/2015] [Indexed: 12/12/2022]
Abstract
Anemia is a major source of morbidity and mortality worldwide. Here we review recent insights into how red blood cells (RBCs) are produced, the pathogenic mechanisms underlying various forms of anemia, and novel therapies derived from these findings. It is likely that these new insights, mainly arising from basic scientific studies, will contribute immensely to both the understanding of frequently debilitating forms of anemia and the ability to treat affected patients. Major worldwide diseases that are likely to benefit from new advances include the hemoglobinopathies (β-thalassemia and sickle cell disease); rare genetic disorders of RBC production; and anemias associated with chronic kidney disease, inflammation, and cancer. Promising new approaches to treatment include drugs that target recently defined pathways in RBC production, iron metabolism, and fetal globin-family gene expression, as well as gene therapies that use improved viral vectors and newly developed genome editing technologies.
Collapse
Affiliation(s)
- Vijay G Sankaran
- 1] Division of Hematology and Oncology, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA. [2] Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA. [3] Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
715
|
Baron D, Giral M, Brouard S. Reconsidering the detection of tolerance to individualize immunosuppression minimization and to improve long-term kidney graft outcomes. Transpl Int 2015; 28:938-59. [DOI: 10.1111/tri.12578] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/03/2015] [Accepted: 04/02/2015] [Indexed: 01/03/2023]
Affiliation(s)
- Daniel Baron
- INSERM; UMR 1064; Nantes France
- CHU de Nantes; ITUN; Nantes France
- Faculté de Médecine; Université de Nantes; Nantes France
| | - Magali Giral
- INSERM; UMR 1064; Nantes France
- CHU de Nantes; ITUN; Nantes France
- Faculté de Médecine; Université de Nantes; Nantes France
| | - Sophie Brouard
- INSERM; UMR 1064; Nantes France
- CHU de Nantes; ITUN; Nantes France
- Faculté de Médecine; Université de Nantes; Nantes France
| |
Collapse
|
716
|
Thannickal VJ, Murthy M, Balch WE, Chandel NS, Meiners S, Eickelberg O, Selman M, Pardo A, White ES, Levy BD, Busse PJ, Tuder RM, Antony VB, Sznajder JI, Budinger GRS. Blue journal conference. Aging and susceptibility to lung disease. Am J Respir Crit Care Med 2015; 191:261-9. [PMID: 25590812 DOI: 10.1164/rccm.201410-1876pp] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The aging of the population in the United States and throughout the developed world has increased morbidity and mortality attributable to lung disease, while the morbidity and mortality from other prevalent diseases has declined or remained stable. Recognizing the importance of aging in the development of lung disease, the American Thoracic Society (ATS) highlighted this topic as a core theme for the 2014 annual meeting. The relationship between aging and lung disease was discussed in several oral symposiums and poster sessions at the annual ATS meeting. In this article, we used the input gathered at the conference to develop a broad framework and perspective to stimulate basic, clinical, and translational research to understand how the aging process contributes to the onset and/or progression of lung diseases. A consistent theme that emerged from the conference was the need to apply novel, systems-based approaches to integrate a growing body of genomic, epigenomic, transcriptomic, and proteomic data and elucidate the relationship between biologic hallmarks of aging, altered lung function, and increased susceptibility to lung diseases in the older population. The challenge remains to causally link the molecular and cellular changes of aging with age-related changes in lung physiology and disease susceptibility. The purpose of this review is to stimulate further research to identify new strategies to prevent or treat age-related lung disease.
Collapse
|
717
|
Jawaid A, Kremer EA, Piatek A, Schulz PE. Improvement of age-related memory impairment with infusion of young plasma: a role for the peripheral amyloid sink? J Am Geriatr Soc 2015; 63:419-20. [PMID: 25688632 DOI: 10.1111/jgs.13262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Ali Jawaid
- Brain Research Institute, University of Zurich/Swiss Federal Institute of Technology, Zurich, Switzerland
| | | | | | | |
Collapse
|
718
|
Pani G. Neuroprotective effects of dietary restriction: Evidence and mechanisms. Semin Cell Dev Biol 2015; 40:106-14. [DOI: 10.1016/j.semcdb.2015.03.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 03/03/2015] [Accepted: 03/04/2015] [Indexed: 12/30/2022]
|
719
|
Intravenous infusion of monocytes isolated from 2-week-old mice enhances clearance of Beta-amyloid plaques in an Alzheimer mouse model. PLoS One 2015; 10:e0121930. [PMID: 25830951 PMCID: PMC4382317 DOI: 10.1371/journal.pone.0121930] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 02/13/2015] [Indexed: 01/15/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by the deposition of β-amyloid (Aβ) senile plaques and tau-associated neurofibrillary tangles. Other disease features include neuroinflammation and cholinergic neurodegeneration, indicating their possible importance in disease propagation. Recent studies have shown that monocytic cells can migrate into the AD brain toward Aβ plaques and reduce plaque burden. The purpose of this study was to evaluate whether the administration of intravenous infusions of 'young' CD11b-positive (+) monocytes into an AD mouse model can enhance Aβ plaque clearance and attenuate cognitive deficits. Peripheral monocytes were isolated from two-week-old wildtype mice using the Pluriselect CD11b+ isolation method and characterized by FACS analysis for surface marker expression and effective phagocytosis of 1 μm fluorescent microspheres, FITC-Dextran or FITC-Aβ1-42. The isolated monocytes were infused via the tail vein into a transgenic AD mouse model, which expresses the Swedish, Dutch/Iowa APP mutations (APPSwDI). The infusions began when animals reached 5 months of age, when little plaque deposition is apparent and were repeated again at 6 and 7 months of age. At 8 months of age, brains were analyzed for Aβ+ plaques, inflammatory processes and microglial (Iba1) activation. Our data show that infusions of two-week-old CD11b+ monocytes into adult APPSwDI mice results in a transient improvement of memory function, a reduction (30%) in Aβ plaque load and significantly in small (<20 μm) and large (>40 μm) plaques. In addition, we observe a reduction in Iba1+ cells, as well as no marked elevations in cytokine levels or other indicators of inflammation. Taken together, our findings indicate that young CD11b+ monocytes may serve as therapeutic candidates for improved Aβ clearance in AD.
Collapse
|
720
|
Burns TC, Li MD, Mehta S, Awad AJ, Morgan AA. Mouse models rarely mimic the transcriptome of human neurodegenerative diseases: A systematic bioinformatics-based critique of preclinical models. Eur J Pharmacol 2015; 759:101-17. [PMID: 25814260 DOI: 10.1016/j.ejphar.2015.03.021] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 03/12/2015] [Accepted: 03/12/2015] [Indexed: 12/12/2022]
Abstract
Translational research for neurodegenerative disease depends intimately upon animal models. Unfortunately, promising therapies developed using mouse models mostly fail in clinical trials, highlighting uncertainty about how well mouse models mimic human neurodegenerative disease at the molecular level. We compared the transcriptional signature of neurodegeneration in mouse models of Alzheimer׳s disease (AD), Parkinson׳s disease (PD), Huntington׳s disease (HD) and amyotrophic lateral sclerosis (ALS) to human disease. In contrast to aging, which demonstrated a conserved transcriptome between humans and mice, only 3 of 19 animal models showed significant enrichment for gene sets comprising the most dysregulated up- and down-regulated human genes. Spearman׳s correlation analysis revealed even healthy human aging to be more closely related to human neurodegeneration than any mouse model of AD, PD, ALS or HD. Remarkably, mouse models frequently upregulated stress response genes that were consistently downregulated in human diseases. Among potential alternate models of neurodegeneration, mouse prion disease outperformed all other disease-specific models. Even among the best available animal models, conserved differences between mouse and human transcriptomes were found across multiple animal model versus human disease comparisons, surprisingly, even including aging. Relative to mouse models, mouse disease signatures demonstrated consistent trends toward preserved mitochondrial function protein catabolism, DNA repair responses, and chromatin maintenance. These findings suggest a more complex and multifactorial pathophysiology in human neurodegeneration than is captured through standard animal models, and suggest that even among conserved physiological processes such as aging, mice are less prone to exhibit neurodegeneration-like changes. This work may help explain the poor track record of mouse-based translational therapies for neurodegeneration and provides a path forward to critically evaluate and improve animal models of human disease.
Collapse
Affiliation(s)
- Terry C Burns
- Department of Neurosurgery, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA.
| | - Matthew D Li
- Department of Neurosurgery, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Swapnil Mehta
- Department of Neurosurgery, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Ahmed J Awad
- Department of Neurosurgery, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Alexander A Morgan
- Department of Neurosurgery, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA
| |
Collapse
|
721
|
Burns TC, Verfaillie CM. From mice to mind: Strategies and progress in translating neuroregeneration. Eur J Pharmacol 2015; 759:90-100. [PMID: 25814255 DOI: 10.1016/j.ejphar.2015.03.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 02/18/2015] [Accepted: 03/12/2015] [Indexed: 12/20/2022]
Abstract
Decisions about what experimental therapies are advanced to clinical trials are based almost exclusively on findings in preclinical animal studies. Over the past 30 years, animal models have forecast the success of hundreds of neuroprotective pharmacological therapies for stroke, Alzheimer׳s disease, spinal cord injury, traumatic brain injury and amyotrophic lateral sclerosis. Yet almost without exception, all have failed. Rapid advances in stem cell technologies have raised new hopes that these neurological diseases may one day be treatable. Still, how can neuroregenerative therapies be translated into clinical realities if available animal models are such poor surrogates of human disease? To address this question we discuss human and rodent neurogenesis, evaluate mechanisms of action for cellular therapies and describe progress in translating neuroregeneration to date. We conclude that not only are appropriate animal models critical to the development of safe and effective therapies, but that the multiple mechanisms of stem cell-mediated therapies may be particularly well suited to the mechanistically diverse nature of central nervous system diseases in mice and man.
Collapse
Affiliation(s)
- Terry C Burns
- Department of Neurosurgery and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, USA.
| | | |
Collapse
|
722
|
Huang TT, Leu D, Zou Y. Oxidative stress and redox regulation on hippocampal-dependent cognitive functions. Arch Biochem Biophys 2015; 576:2-7. [PMID: 25797440 DOI: 10.1016/j.abb.2015.03.014] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/12/2015] [Accepted: 03/13/2015] [Indexed: 12/17/2022]
Abstract
Hippocampal-dependent cognitive functions rely on production of new neurons and maintenance of dendritic structures to provide the synaptic plasticity needed for learning and formation of new memories. Hippocampal formation is exquisitely sensitive to patho-physiological changes, and reduced antioxidant capacity and exposure to low dose irradiation can significantly impede hippocampal-dependent functions of learning and memory by reducing the production of new neurons and alter dendritic structures in the hippocampus. Although the mechanism leading to impaired cognitive functions is complex, persistent oxidative stress likely plays an important role in the SOD-deficient and radiation-exposed hippocampal environment. Aging is associated with increased production of pro-oxidants and accumulation of oxidative end products. Similar to the hippocampal defects observed in SOD-deficient mice and mice exposed to low dose irradiation, reduced capacity in learning and memory, diminishing hippocampal neurogenesis, and altered dendritic network are universal in the aging brains. Given the similarities in cellular and structural changes in the aged, SOD-deficient, and radiation-exposed hippocampal environment and the corresponding changes in cognitive decline, understanding the shared underlying mechanism will provide more flexible and efficient use of SOD deficiency or irradiation to model age-related changes in cognitive functions and identify potential therapeutic or intervention methods.
Collapse
Affiliation(s)
- Ting-Ting Huang
- Geriatric Research, Education, and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA; Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| | - David Leu
- Geriatric Research, Education, and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA; Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Yani Zou
- Geriatric Research, Education, and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA; Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
723
|
Lynch DB, Jeffery IB, O'Toole PW. The role of the microbiota in ageing: current state and perspectives. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2015; 7:131-8. [PMID: 25777986 PMCID: PMC4406138 DOI: 10.1002/wsbm.1293] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 01/19/2015] [Accepted: 01/22/2015] [Indexed: 12/19/2022]
Abstract
Since the application of high-throughput technologies to investigate complex microbial communities, alterations in the human gut microbiota have been associated with an increasing number of diseases and conditions. This field of research has developed into an area of intense study which is quite different to the microbial investigations that have preceded it in terms of both the broadness of the area of research and the complexity of the analyses. In this review, we discuss gut microbiota changes observed in ageing in the context of the physiological changes that accompany senescence, examine what correlations can be established or inferred, and we discuss what key questions remain to be answered in the field. © 2015 The Authors. WIREs Systems Biology and Medicine published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Denise B Lynch
- School of Microbiology, University College Cork, Ireland; Alimentary Pharmabiotic Centre, University College Cork, Ireland
| | | | | |
Collapse
|
724
|
Cognitive frailty, a novel target for the prevention of elderly dependency. Ageing Res Rev 2015; 20:1-10. [PMID: 25555677 DOI: 10.1016/j.arr.2014.12.004] [Citation(s) in RCA: 208] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 12/12/2014] [Accepted: 12/16/2014] [Indexed: 01/10/2023]
Abstract
Frailty is a complex and heterogeneous clinical syndrome. Cognitive frailty has been considered as a subtype of frailty. In this study, we refine the definition of cognitive frailty based on existing reports about frailty and the latest progress in cognition research. We obtain evidence from the literature regarding the role of pre-physical frailty in pathological aging. We propose that cognitive impairment of cognitive frailty results from physical or pre-physical frailty and comprises two subtypes: the reversible and the potentially reversible. Reversible cognitive impairment is indicated by subjective cognitive decline (SCD) and/or positive fluid and imaging biomarkers of amyloid-β accumulation and neurodegeneration. Potentially reversible cognitive impairment is MCI (CDR=0.5). Based on the severity of cognitive impairment, it is possible to determine the primary and secondary preventative measures for cognitive frailty. We further determine whether SCD is a component of pre-clinical AD or the early stage of other neurodegenerative diseases, which is required for guiding personal clinical intervention.
Collapse
|
725
|
Khong DM, Dudakov JA, Hammett MV, Jurblum MI, Khong SML, Goldberg GL, Ueno T, Spyroglou L, Young LF, van den Brink MRM, Boyd RL, Chidgey AP. Enhanced hematopoietic stem cell function mediates immune regeneration following sex steroid blockade. Stem Cell Reports 2015; 4:445-58. [PMID: 25733018 PMCID: PMC4375937 DOI: 10.1016/j.stemcr.2015.01.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 01/24/2015] [Accepted: 01/26/2015] [Indexed: 12/12/2022] Open
Abstract
Mechanisms underlying age-related defects within lymphoid-lineages remain poorly understood. We previously reported that sex steroid ablation (SSA) induced lymphoid rejuvenation and enhanced recovery from hematopoietic stem cell (HSC) transplantation (HSCT). We herein show that, mechanistically, SSA induces hematopoietic and lymphoid recovery by functionally enhancing both HSC self-renewal and propensity for lymphoid differentiation through intrinsic molecular changes. Our transcriptome analysis revealed further hematopoietic support through rejuvenation of the bone marrow (BM) microenvironment, with upregulation of key hematopoietic factors and master regulatory factors associated with aging such as Foxo1. These studies provide important cellular and molecular insights into understanding how SSA-induced regeneration of the hematopoietic compartment can underpin recovery of the immune system following damaging cytoablative treatments. These findings support a short-term strategy for clinical use of SSA to enhance the production of lymphoid cells and HSC engraftment, leading to improved outcomes in adult patients undergoing HSCT and immune depletion in general. Sex steroid ablation (SSA) increases number of hematopoietic stem cells (HSCs) SSA enhances reconstitution potential and self-renewal of HSCs SSA reverses the age-associated decline in Foxo1 expression by hematopoietic niche There is an increase in niche expression of hematopoiesis-associated factors after SSA
Collapse
Affiliation(s)
- Danika M Khong
- Stem Cells and Immune Regeneration Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - Jarrod A Dudakov
- Stem Cells and Immune Regeneration Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| | - Maree V Hammett
- Stem Cells and Immune Regeneration Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - Marc I Jurblum
- Stem Cells and Immune Regeneration Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - Sacha M L Khong
- Stem Cells and Immune Regeneration Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - Gabrielle L Goldberg
- Stem Cells and Immune Regeneration Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - Tomoo Ueno
- Stem Cells and Immune Regeneration Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - Lisa Spyroglou
- Stem Cells and Immune Regeneration Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - Lauren F Young
- Stem Cells and Immune Regeneration Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | | | - Richard L Boyd
- Stem Cells and Immune Regeneration Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - Ann P Chidgey
- Stem Cells and Immune Regeneration Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
726
|
Bukovsky A. Novel methods of treating ovarian infertility in older and POF women, testicular infertility, and other human functional diseases. Reprod Biol Endocrinol 2015; 13:10. [PMID: 25889983 PMCID: PMC4414002 DOI: 10.1186/s12958-015-0001-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/28/2015] [Indexed: 12/11/2022] Open
Abstract
In vitro maturation (IVM) and in vitro fertilization (IVF) technologies are facing with growing demands of older women to conceive. Although ovarian stem cells (OSCs) of older women are capable of producing in vitro fresh oocyte-like cells (OLCs), such cells cannot respond to IVM and IVF due to the lack of granulosa cells required for their maturation. Follicular renewal is also dependent on support of circulating blood mononuclear cells. They induce intermediary stages of meiosis (metaphase I chromosomal duplication and crossover, anaphase, telophase, and cytokinesis) in newly emerging ovarian germ cells, as for the first time demonstrated here, induce formation of granulosa cells, and stimulate follicular growth and development. A pretreatment of OSC culture with mononuclear cells collected from blood of a young healthy fertile woman may cause differentiation of bipotential OSCs into both developing germ and granulosa cells. A small blood volume replacement may enable treatment of ovarian infertility in vivo. The transferred mononuclear cells may temporarily rejuvenate virtually all tissues, including improvement of the function of endocrine tissues. Formation of new follicles and their development may be sufficient for IVM and IVF. The novel proposed in vitro approaches may be used as a second possibility. Infertility of human males affects almost a half of the infertility cases worldwide. Small blood volume replacement from young healthy fertile men may also be easy approach for the improvement of sperm quality in older or other affected men. In addition, body rejuvenation by small blood volume replacement from young healthy individuals of the same sex could represent a decline of in vitro methodology in favor of in vivo treatment for human functional diseases. Here we propose for the first time that blood mononuclear cells are essential for rejuvenation of those tissues, where immune system components participate in an appropriate division and differentiation of tissue stem cells. If needed, small blood volume replacement from distinct young healthy individuals could be utilized in six month intervals for repair of young altered or aged reproductive and other tissue functions. Systemic and local use of honey bee propolis tincture is an alternative option for functional rejuvenation of some tissues.
Collapse
Affiliation(s)
- Antonin Bukovsky
- The Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| |
Collapse
|
727
|
Xu Y, Cao W, Zhou M, Li C, Luo Y, Wang H, Zhao R, Jiang S, Yang J, Liu Y, Wang X, Li X, Xiong W, Ma J, Peng S, Zeng Z, Li X, Tan M, Li G. Inactivation of BRD7 results in impaired cognitive behavior and reduced synaptic plasticity of the medial prefrontal cortex. Behav Brain Res 2015; 286:1-10. [PMID: 25721744 DOI: 10.1016/j.bbr.2015.02.031] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 02/10/2015] [Accepted: 02/16/2015] [Indexed: 10/24/2022]
Abstract
BRD7 is a bromodomain-containing protein (BCP), and recent evidence implicates the role of BCPs in the initiation and development of neurodevelopmental disorders. However, few studies have investigated the biological functions of BRD7 in the central nervous system. In our study, BRD7 was found to be widely expressed in various regions of the mouse brain, including the medial prefrontal cortex (mPFC), caudate putamen (CPu), hippocampus (Hip), midbrain (Mb), cerebellum (Cb), and mainly co-localized with neuron but not with glia. Using a BRD7 knockout mouse model and a battery of behavioral tests, we report that disruption of BRD7 results in impaired cognitive behavior leaving the emotional behavior unaffected. Moreover, a series of proteins involved in synaptic plasticity were decreased in the medial prefrontal cortex and there was a concomitant decrease in neuronal spine density and dendritic branching in the medial prefrontal cortex. However, no significant difference was found in the hippocampus compared to the wild-type mice. Thus, BRD7 might play a critical role in the regulation of synaptic plasticity and affect cognitive behavior.
Collapse
Affiliation(s)
- Yang Xu
- Cancer Research Institute, Central South University, Xiangya Road 110 Changsha, Hunan Province 410078, People's Republic of China
| | - Wenyu Cao
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Tongzipo Road 172, Changsha, Hunan Province 410013, People's Republic of China
| | - Ming Zhou
- Cancer Research Institute, Central South University, Xiangya Road 110 Changsha, Hunan Province 410078, People's Republic of China.
| | - Changqi Li
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Tongzipo Road 172, Changsha, Hunan Province 410013, People's Republic of China
| | - Yanwei Luo
- Cancer Research Institute, Central South University, Xiangya Road 110 Changsha, Hunan Province 410078, People's Republic of China
| | - Heran Wang
- Cancer Research Institute, Central South University, Xiangya Road 110 Changsha, Hunan Province 410078, People's Republic of China
| | - Ran Zhao
- Cancer Research Institute, Central South University, Xiangya Road 110 Changsha, Hunan Province 410078, People's Republic of China
| | - Shihe Jiang
- Cancer Research Institute, Central South University, Xiangya Road 110 Changsha, Hunan Province 410078, People's Republic of China
| | - Jing Yang
- Cancer Research Institute, Central South University, Xiangya Road 110 Changsha, Hunan Province 410078, People's Republic of China
| | - Yukun Liu
- Cancer Research Institute, Central South University, Xiangya Road 110 Changsha, Hunan Province 410078, People's Republic of China
| | - Xinye Wang
- Cancer Research Institute, Central South University, Xiangya Road 110 Changsha, Hunan Province 410078, People's Republic of China
| | - Xiayu Li
- The Third Xiang-Ya Hospital, Central South University, Tongzipo Road 237, Changsha, Hunan Province 410013, People's Republic of China
| | - Wei Xiong
- Cancer Research Institute, Central South University, Xiangya Road 110 Changsha, Hunan Province 410078, People's Republic of China
| | - Jian Ma
- Cancer Research Institute, Central South University, Xiangya Road 110 Changsha, Hunan Province 410078, People's Republic of China
| | - Shuping Peng
- Cancer Research Institute, Central South University, Xiangya Road 110 Changsha, Hunan Province 410078, People's Republic of China
| | - Zhaoyang Zeng
- Cancer Research Institute, Central South University, Xiangya Road 110 Changsha, Hunan Province 410078, People's Republic of China
| | - Xiaoling Li
- Cancer Research Institute, Central South University, Xiangya Road 110 Changsha, Hunan Province 410078, People's Republic of China
| | - Ming Tan
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Guiyuan Li
- Cancer Research Institute, Central South University, Xiangya Road 110 Changsha, Hunan Province 410078, People's Republic of China.
| |
Collapse
|
728
|
|
729
|
The effects of testosterone and insulin-like growth factor 1 on motor system form and function. Exp Gerontol 2015; 64:81-6. [PMID: 25681641 DOI: 10.1016/j.exger.2015.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 01/31/2015] [Accepted: 02/10/2015] [Indexed: 12/25/2022]
Abstract
In this perspective article, we review the effects of selected anabolic hormones on the motoric system and speculate on the role these hormones may have on influencing muscle and physical function via their impact on the nervous system. Both muscle strength and anabolic hormone levels decline around middle age into old age over a similar time period, and several animal and human studies indicate that exogenously increasing anabolic hormones (e.g., testosterone and insulin-like growth factor-1 (IGF-1)) in aged subjects is positively associated with improved muscle strength. While most studies in humans have focused on the effects of anabolic hormones on muscle growth, few have considered the impact these hormones have on the motoric system. However, data from animals demonstrate that administering either testosterone or IGF-1 to cells of the central and peripheral motor system can increase cell excitability, attenuate atrophic changes, and improve regenerative capacity of motor neurons. While these studies do not directly indicate that changes in anabolic hormones contribute to reduced human performance in the elderly (e.g., muscle weakness and physical limitations), they do suggest that additional research is warranted along these lines.
Collapse
|
730
|
Katsimpardi L, Rubin LL. Young systemic factors as a medicine for age-related neurodegenerative diseases. NEUROGENESIS 2015; 2:e1004971. [PMID: 27502604 PMCID: PMC4973601 DOI: 10.1080/23262133.2015.1004971] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 12/14/2014] [Accepted: 01/03/2015] [Indexed: 01/19/2023]
Abstract
It is widely known that neurogenesis, brain function and cognition decline with aging. Increasing evidence suggests that cerebrovascular dysfunction is a major cause of cognitive impairment in the elderly but is also involved in age-related neurodegenerative diseases. Finding ways and molecules that reverse this process and ameliorate age- and disease-related cognitive impairment by targeting vascular and neurogenic deterioration would be of great therapeutic value. In Katsimpardi et al. we reported that young blood has a dual beneficial effect in the aged brain by restoring age-related decline in neurogenesis as well as inducing a striking remodeling of the aged vasculature and restoring blood flow to youthful levels. Additionally, we identified a youthful systemic factor, GDF11 that recapitulates these beneficial effects of young blood. We believe that the identification of young systemic factors that can rejuvenate the aged brain opens new roads to therapeutic intervention for neurodegenerative diseases by targeting both neural stem cells and neurogenesis as well as at the vasculature.
Collapse
Affiliation(s)
- Lida Katsimpardi
- Department of Stem Cell and Regenerative Biology; Harvard University and Harvard Stem Cell Institute ; Cambridge, MA USA
| | - Lee L Rubin
- Department of Stem Cell and Regenerative Biology; Harvard University and Harvard Stem Cell Institute ; Cambridge, MA USA
| |
Collapse
|
731
|
A common gene signature across multiple studies relate biomarkers and functional regulation in tolerance to renal allograft. Kidney Int 2015; 87:984-95. [PMID: 25629549 PMCID: PMC4424816 DOI: 10.1038/ki.2014.395] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 10/02/2014] [Accepted: 10/23/2014] [Indexed: 01/21/2023]
Abstract
Patients tolerant to a kidney graft display a specific blood cell transcriptional pattern but results from five different studies were inconsistent, raising the question of relevance for future clinical application. To resolve this, we sought to identify a common gene signature, specific functional and cellular components, and discriminating biomarkers for tolerance following kidney transplantation. A meta-analysis of studies identified a robust gene signature involving proliferation of B and CD4 T cells, and inhibition of CD14 monocyte related functions among 96 tolerant samples. This signature was further supported through a cross-validation approach, yielding 92.5% accuracy independent of the study of origin. Experimental validation, performed on new tolerant samples and using a selection of the top-20 biomarkers, returned 91.7% of good classification. Beyond the confirmation of B-cell involvement, our data also indicated participation of other cell subsets in tolerance. Thus, the use of the top 20 biomarkers, mostly centered on B cells, may provide a common and standardized tool towards personalized medicine for the monitoring of tolerant or low-risk patients among kidney allotransplant recipients. These data point to a global preservation of genes favoring the maintenance of a homeostatic and ‘healthy' environment in tolerant patients and may contribute to a better understanding of tolerance maintenance mechanisms.
Collapse
|
732
|
Ding DC, Chang YH, Shyu WC, Lin SZ. Human umbilical cord mesenchymal stem cells: a new era for stem cell therapy. Cell Transplant 2015; 24:339-47. [PMID: 25622293 DOI: 10.3727/096368915x686841] [Citation(s) in RCA: 353] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The human umbilical cord is a promising source of mesenchymal stem cells (HUCMSCs). Unlike bone marrow stem cells, HUCMSCs have a painless collection procedure and faster self-renewal properties. Different derivation protocols may provide different amounts and populations of stem cells. Stem cell populations have also been reported in other compartments of the umbilical cord, such as the cord lining, perivascular tissue, and Wharton's jelly. HUCMSCs are noncontroversial sources compared to embryonic stem cells. They can differentiate into the three germ layers that promote tissue repair and modulate immune responses and anticancer properties. Thus, they are attractive autologous or allogenic agents for the treatment of malignant and nonmalignant solid and soft cancers. HUCMCs also can be the feeder layer for embryonic stem cells or other pluripotent stem cells. Regarding their therapeutic value, storage banking system and protocols should be established immediately. This review critically evaluates their therapeutic value, challenges, and future directions for their clinical applications.
Collapse
Affiliation(s)
- Dah-Ching Ding
- Department of Obstetrics and Gynecology, Buddhist Tzu Chi General Hospital, Tzu Chi University, Hualien, Taiwan
| | | | | | | |
Collapse
|
733
|
Abstract
Blood vessels are critical to deliver oxygen and nutrients to all of the tissues and organs throughout the body. The blood vessels that vascularize the central nervous system (CNS) possess unique properties, termed the blood-brain barrier, which allow these vessels to tightly regulate the movement of ions, molecules, and cells between the blood and the brain. This precise control of CNS homeostasis allows for proper neuronal function and also protects the neural tissue from toxins and pathogens, and alterations of these barrier properties are an important component of pathology and progression of different neurological diseases. The physiological barrier is coordinated by a series of physical, transport, and metabolic properties possessed by the endothelial cells (ECs) that form the walls of the blood vessels, and these properties are regulated by interactions with different vascular, immune, and neural cells. Understanding how these different cell populations interact to regulate the barrier properties is essential for understanding how the brain functions during health and disease.
Collapse
Affiliation(s)
- Richard Daneman
- Departments of Neuroscience and Pharmacology, University of California, San Diego, San Diego, California 92093
| | - Alexandre Prat
- Department of Neuroscience, Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| |
Collapse
|
734
|
The systemic milieu as a mediator of dietary influence on stem cell function during ageing. Ageing Res Rev 2015; 19:53-64. [PMID: 25481406 DOI: 10.1016/j.arr.2014.11.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 11/20/2014] [Accepted: 11/26/2014] [Indexed: 01/09/2023]
Abstract
The regenerative decline of organisms during ageing is linked to the reduced proliferative activity, impaired function and exhaustion of tissue-specific stem and progenitor cells. Studies using heterochronic parabiosis, involving the surgical attachment of young and old organisms so that they share a common vascular system, have revealed that the systemic environment has a profound effect on stem cell function. In particular, specific youthful rejuvenating circulatory factors reverse age-related declines in stem cell function, whereas the old milieu contains inhibitory factors that impede stem cell function in young animals. Similarly, the effects of certain dietary interventions, namely calorie restriction, also induce a more youthful cellular and molecular phenotype in ageing stem cells throughout the body. Further to this, there are key molecular pathways involved in translating the availability of nutrients into altered stem cell function, including signalling in the insulin and insulin-like growth factor and mechanistic target of rapamycin (mTOR) pathways. In this review, we discuss the potential role of dietary interventions to promote a more rejuvenating systemic milieu in order to enhance stem cell function and promote healthy ageing.
Collapse
|
735
|
|
736
|
Costa V, Lugert S, Jagasia R. Role of adult hippocampal neurogenesis in cognition in physiology and disease: pharmacological targets and biomarkers. Handb Exp Pharmacol 2015; 228:99-155. [PMID: 25977081 DOI: 10.1007/978-3-319-16522-6_4] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Adult hippocampal neurogenesis is a remarkable form of brain structural plasticity by which new functional neurons are generated from adult neural stem cells/precursors. Although the precise role of this process remains elusive, adult hippocampal neurogenesis is important for learning and memory and it is affected in disease conditions associated with cognitive impairment, depression, and anxiety. Immature neurons in the adult brain exhibit an enhanced structural and synaptic plasticity during their maturation representing a unique population of neurons to mediate specific hippocampal function. Compelling preclinical evidence suggests that hippocampal neurogenesis is modulated by a broad range of physiological stimuli which are relevant in cognitive and emotional states. Moreover, multiple pharmacological interventions targeting cognition modulate adult hippocampal neurogenesis. In addition, recent genetic approaches have shown that promoting neurogenesis can positively modulate cognition associated with both physiology and disease. Thus the discovery of signaling pathways that enhance adult neurogenesis may lead to therapeutic strategies for improving memory loss due to aging or disease. This chapter endeavors to review the literature in the field, with particular focus on (1) the role of hippocampal neurogenesis in cognition in physiology and disease; (2) extrinsic and intrinsic signals that modulate hippocampal neurogenesis with a focus on pharmacological targets; and (3) efforts toward novel strategies pharmacologically targeting neurogenesis and identification of biomarkers of human neurogenesis.
Collapse
Affiliation(s)
- Veronica Costa
- Roche Pharmaceutical Research and Early Development, Neuroscience Ophthalmology and Rare Diseases (NORD), Roche Innovation Center Basel, 124 Grenzacherstrasse, 4070, Basel, Switzerland
| | | | | |
Collapse
|
737
|
Wang F, Liu H, Shen X, Ao H, Moore N, Gao L, Chen L, Hu H, Ma H, Yang Z, Zhai C, Qin J, Zhou G, Peng Y, Feng X, Li R, Liang C. The combined treatment of amyloid-β1-42-stimulated bone marrow–derived dendritic cells plus splenocytes from young mice prevents the development of Alzheimer's disease in APPswe/PSENldE9 mice. Neurobiol Aging 2015; 36:111-22. [DOI: 10.1016/j.neurobiolaging.2014.06.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 06/23/2014] [Accepted: 06/28/2014] [Indexed: 10/25/2022]
|
738
|
Serre-Miranda C, Roque S, Santos NC, Portugal-Nunes C, Costa P, Palha JA, Sousa N, Correia-Neves M. Effector memory CD4(+) T cells are associated with cognitive performance in a senior population. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2014; 2:e54. [PMID: 25566544 PMCID: PMC4277304 DOI: 10.1212/nxi.0000000000000054] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 11/18/2014] [Indexed: 01/24/2023]
Abstract
Objective: Immunosenescence and cognitive decline are common markers of the aging process. Taking into consideration the heterogeneity observed in aging processes and the recently described link between lymphocytes and cognition, we herein explored the possibility of an association between alterations in lymphocytic populations and cognitive performance. Methods: In a cohort of cognitively healthy adults (n = 114), previously characterized by diverse neurocognitive/psychological performance patterns, detailed peripheral blood immunophenotyping of both the innate and adaptive immune systems was performed by flow cytometry. Results: Better cognitive performance was associated with lower numbers of effector memory CD4+ T cells and higher numbers of naive CD8+ T cells and B cells. Furthermore, effector memory CD4+ T cells were found to be predictors of general and executive function and memory, even when factors known to influence cognitive performance in older individuals (e.g., age, sex, education, and mood) were taken into account. Conclusions: This is the first study in humans associating specific phenotypes of the immune system with distinct cognitive performance in healthy aging.
Collapse
Affiliation(s)
- Cláudia Serre-Miranda
- Life and Health Sciences Research Institute (ICVS) (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), School of Health Sciences, University of Minho, Braga; ICVS/3B's - PT Government Associate Laboratory (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), Braga/Guimarães; and Clinical Academic Center-Braga (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S.), Braga, Portugal
| | - Susana Roque
- Life and Health Sciences Research Institute (ICVS) (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), School of Health Sciences, University of Minho, Braga; ICVS/3B's - PT Government Associate Laboratory (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), Braga/Guimarães; and Clinical Academic Center-Braga (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S.), Braga, Portugal
| | - Nadine Correia Santos
- Life and Health Sciences Research Institute (ICVS) (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), School of Health Sciences, University of Minho, Braga; ICVS/3B's - PT Government Associate Laboratory (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), Braga/Guimarães; and Clinical Academic Center-Braga (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S.), Braga, Portugal
| | - Carlos Portugal-Nunes
- Life and Health Sciences Research Institute (ICVS) (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), School of Health Sciences, University of Minho, Braga; ICVS/3B's - PT Government Associate Laboratory (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), Braga/Guimarães; and Clinical Academic Center-Braga (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S.), Braga, Portugal
| | - Patrício Costa
- Life and Health Sciences Research Institute (ICVS) (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), School of Health Sciences, University of Minho, Braga; ICVS/3B's - PT Government Associate Laboratory (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), Braga/Guimarães; and Clinical Academic Center-Braga (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S.), Braga, Portugal
| | - Joana Almeida Palha
- Life and Health Sciences Research Institute (ICVS) (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), School of Health Sciences, University of Minho, Braga; ICVS/3B's - PT Government Associate Laboratory (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), Braga/Guimarães; and Clinical Academic Center-Braga (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S.), Braga, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS) (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), School of Health Sciences, University of Minho, Braga; ICVS/3B's - PT Government Associate Laboratory (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), Braga/Guimarães; and Clinical Academic Center-Braga (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S.), Braga, Portugal
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute (ICVS) (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), School of Health Sciences, University of Minho, Braga; ICVS/3B's - PT Government Associate Laboratory (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S., M.C.-N.), Braga/Guimarães; and Clinical Academic Center-Braga (C.S.-M., S.R., N.C.S., C.P.-N., P.C., J.A.P., N.S.), Braga, Portugal
| |
Collapse
|
739
|
Johansson JU, Woodling NS, Wang Q, Panchal M, Liang X, Trueba-Saiz A, Brown HD, Mhatre SD, Loui T, Andreasson KI. Prostaglandin signaling suppresses beneficial microglial function in Alzheimer's disease models. J Clin Invest 2014; 125:350-64. [PMID: 25485684 DOI: 10.1172/jci77487] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 10/30/2014] [Indexed: 12/25/2022] Open
Abstract
Microglia, the innate immune cells of the CNS, perform critical inflammatory and noninflammatory functions that maintain normal neural function. For example, microglia clear misfolded proteins, elaborate trophic factors, and regulate and terminate toxic inflammation. In Alzheimer's disease (AD), however, beneficial microglial functions become impaired, accelerating synaptic and neuronal loss. Better understanding of the molecular mechanisms that contribute to microglial dysfunction is an important objective for identifying potential strategies to delay progression to AD. The inflammatory cyclooxygenase/prostaglandin E2 (COX/PGE2) pathway has been implicated in preclinical AD development, both in human epidemiology studies and in transgenic rodent models of AD. Here, we evaluated murine models that recapitulate microglial responses to Aβ peptides and determined that microglia-specific deletion of the gene encoding the PGE2 receptor EP2 restores microglial chemotaxis and Aβ clearance, suppresses toxic inflammation, increases cytoprotective insulin-like growth factor 1 (IGF1) signaling, and prevents synaptic injury and memory deficits. Our findings indicate that EP2 signaling suppresses beneficial microglia functions that falter during AD development and suggest that inhibition of the COX/PGE2/EP2 immune pathway has potential as a strategy to restore healthy microglial function and prevent progression to AD.
Collapse
|
740
|
Bouchard J, Villeda SA. Aging and brain rejuvenation as systemic events. J Neurochem 2014; 132:5-19. [PMID: 25327899 PMCID: PMC4301186 DOI: 10.1111/jnc.12969] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 09/25/2014] [Indexed: 12/13/2022]
Abstract
The effects of aging were traditionally thought to be immutable, particularly evident in the loss of plasticity and cognitive abilities occurring in the aged central nervous system (CNS). However, it is becoming increasingly apparent that extrinsic systemic manipulations such as exercise, caloric restriction, and changing blood composition by heterochronic parabiosis or young plasma administration can partially counteract this age-related loss of plasticity in the aged brain. In this review, we discuss the process of aging and rejuvenation as systemic events. We summarize genetic studies that demonstrate a surprising level of malleability in organismal lifespan, and highlight the potential for systemic manipulations to functionally reverse the effects of aging in the CNS. Based on mounting evidence, we propose that rejuvenating effects of systemic manipulations are mediated, in part, by blood-borne ‘pro-youthful’ factors. Thus, systemic manipulations promoting a younger blood composition provide effective strategies to rejuvenate the aged brain. As a consequence, we can now consider reactivating latent plasticity dormant in the aged CNS as a means to rejuvenate regenerative, synaptic, and cognitive functions late in life, with potential implications even for extending lifespan.
Collapse
Affiliation(s)
- Jill Bouchard
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA; The Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, San Francisco, California, USA
| | | |
Collapse
|
741
|
Notable advances 2014. Nat Med 2014; 20:1368-9. [PMID: 25473912 DOI: 10.1038/nm1214-1368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
742
|
[Dracula was right: The elixir of youth could be the young blood]. ACTA ACUST UNITED AC 2014; 90:453. [PMID: 25443204 DOI: 10.1016/j.oftal.2014.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 05/28/2014] [Indexed: 11/22/2022]
|
743
|
Wejbrandt A. Defining aging in cyborgs: a bio-techno-social definition of aging. J Aging Stud 2014; 31:104-9. [PMID: 25456627 DOI: 10.1016/j.jaging.2014.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 09/10/2014] [Accepted: 09/10/2014] [Indexed: 11/17/2022]
Abstract
Initially the aim of this article was to discuss and define aging at the intersection point between biology and sociology. However, recent biomedical and technological advances are changing the discourse on aging, and against this background the author of this article argues that current definitions of aging should be improved. The author emphasizes that there is a need to update current definitions of aging, or to formulate new multidisciplinary ones. The author suggests that (besides biology, psychology and sociology) the technological discipline should be included in the integrative gerontology model. Finally, in this article a new definition of aging is put forward. According to the author of this article, human bio-techno-social aging is characterized by: (a) a time-bound process of change including, (b) both reversible and irreversible biological processes, (c) social processes forming an irreversible chain of events, and (d) an increasing use of technological artifacts whose purpose is to support or replace damaged biological functions; and/or an increasing use of technological artifacts whose purpose is to facilitate or enable interaction.
Collapse
Affiliation(s)
- Anita Wejbrandt
- Department of Sociology, Uppsala University, Box 624, S-751 26 Uppsala, Sweden.
| |
Collapse
|
744
|
Cerpa W, Ramos-Fernández E, Inestrosa NC. Modulation of the NMDA Receptor Through Secreted Soluble Factors. Mol Neurobiol 2014; 53:299-309. [PMID: 25429903 DOI: 10.1007/s12035-014-9009-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 11/14/2014] [Indexed: 12/11/2022]
Abstract
Synaptic activity is a critical determinant in the formation and development of excitatory synapses in the central nervous system (CNS). The excitatory current is produced and regulated by several ionotropic receptors, including those that respond to glutamate. These channels are in turn regulated through several secreted factors that function as synaptic organizers. Specifically, Wnt, brain-derived neurotrophic factor (BDNF), fibroblast growth factor (FGF), and transforming growth factor (TGF) particularly regulate the N-methyl-D-aspartate receptor (NMDAR) glutamatergic channel. These factors likely regulate early embryonic development and directly control key proteins in the function of important glutamatergic channels. Here, we review the secreted molecules that participate in synaptic organization and discuss the cell signaling behind of this fine regulation. Additionally, we discuss how these factors are dysregulated in some neuropathologies associated with glutamatergic synaptic transmission in the CNS.
Collapse
Affiliation(s)
- Waldo Cerpa
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Eva Ramos-Fernández
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centre for Healthy Brain Ageing, School of Psychiatry, UNSW, Faculty of Medicine, University of New South Wales, Sydney, Australia.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
745
|
Abstract
Pancreatic islets secrete hormones that play a key role in regulating blood glucose levels (glycemia). Age-dependent impairment of islet function and concomitant dysregulation of glycemia are major health threats in aged populations. However, the major causes of the age-dependent decline of islet function are still disputed. Here we demonstrate that aging of pancreatic islets in mice and humans is notably associated with inflammation and fibrosis of islet blood vessels but does not affect glucose sensing and the insulin secretory capacity of islet beta cells. Accordingly, when transplanted into the anterior chamber of the eye of young mice with diabetes, islets from old mice are revascularized with healthy blood vessels, show strong islet cell proliferation, and fully restore control of glycemia. Our results indicate that beta cell function does not decline with age and suggest that islet function is threatened by an age-dependent impairment of islet vascular function. Strategies to mitigate age-dependent dysregulation in glycemia should therefore target systemic and/or local inflammation and fibrosis of the aged islet vasculature.
Collapse
|
746
|
Torres M, Rojas M, Campillo N, Cardenes N, Montserrat JM, Navajas D, Farré R. Parabiotic model for differentiating local and systemic effects of continuous and intermittent hypoxia. J Appl Physiol (1985) 2014; 118:42-7. [PMID: 25377885 DOI: 10.1152/japplphysiol.00858.2014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hypoxia can be damaging either because cells are directly sensitive to low oxygen pressure in their local microenvironment and/or because they are exposed to circulating factors systemically secreted in response to hypoxia. The conventional hypoxia model, breathing hypoxic air, does not allow one to distinguish between these local and systemic effects. Here we propose and validate a model for differentially applying local and systemic hypoxic challenges in an animal. We used parabiosis, two mice sharing circulation by surgical union through the skin, and tested the hypothesis that when one of the parabionts breathes room air and the other one is subjected to hypoxic air, both mice share systemic circulation but remain normoxic and hypoxic, respectively. We tested two common hypoxic paradigms in 10 parabiotic pairs: continuous hypoxia (10% O2) mimicking chronic lung diseases, and intermittent hypoxia (40 s, 21% O2; 20 s, 5% O2) simulating sleep apnea. Arterial oxygen saturation and oxygen partial pressure at muscle tissue were measured in both parabionts. Effective cross-circulation was assessed by intraperitoneally injecting a dye in one of the parabionts and measuring blood dye concentration in both animals after 2 h. The results confirmed the hypothesis that tissues of the parabiont under room air were perfused with normally oxygenated blood and, at the same time, were exposed to all of the systemic mediators secreted by the other parabiont actually subjected to hypoxia. In conclusion, combination of parabiosis and hypoxic/normoxic air breathing is a novel approach to investigate the effects of local and systemic hypoxia in respiratory diseases.
Collapse
Affiliation(s)
- Marta Torres
- CIBER de Enfermedades Respiratorias, Bunyola, Spain; Sleep Laboratory, Hospital Clinic, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Mauricio Rojas
- Dorothy P. & Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Noelia Campillo
- CIBER de Enfermedades Respiratorias, Bunyola, Spain; Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Nayra Cardenes
- Dorothy P. & Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Josep M Montserrat
- CIBER de Enfermedades Respiratorias, Bunyola, Spain; Sleep Laboratory, Hospital Clinic, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Institut Investigacions Biomediques August Pi Sunyer, Barcelona, Spain; and
| | - Daniel Navajas
- CIBER de Enfermedades Respiratorias, Bunyola, Spain; Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Institut de Bioenginyeria de Catalunya, Barcelona, Spain
| | - Ramon Farré
- CIBER de Enfermedades Respiratorias, Bunyola, Spain; Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Institut Investigacions Biomediques August Pi Sunyer, Barcelona, Spain; and
| |
Collapse
|
747
|
Shytikov D, Balva O, Debonneuil E, Glukhovskiy P, Pishel I. Aged mice repeatedly injected with plasma from young mice: a survival study. Biores Open Access 2014; 3:226-32. [PMID: 25371859 PMCID: PMC4215333 DOI: 10.1089/biores.2014.0043] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
It was reported using various biological models that the administration of blood factors from young animals to old animals could rejuvenate certain functions. To assess the anti-aging effect of young blood we tested the influence of repeated injections of plasma from young mice on the lifespan of aged mice. One group of 36 CBA/Ca female mice aged 10-12 months was treated by repeated injections of plasma from 2- to 4-month-old females (averaging 75-150 μL per injection, once intravenously and once intraperitoneally per week for 16 months). Their lifespan was compared to a control group that received saline injections. The median lifespan of mice from the control group was 27 months versus 26.4 months in plasma-treated group; the repeated injections of young plasma did not significantly impact either median or maximal lifespan.
Collapse
Affiliation(s)
- Dmytro Shytikov
- D.F. Chebotarev State Institute of Gerontology NAMS , Lab Pathophysiology and Immunology, Kyiv, Ukraine
| | - Olexiy Balva
- D.F. Chebotarev State Institute of Gerontology NAMS , Lab Pathophysiology and Immunology, Kyiv, Ukraine
| | | | - Pavel Glukhovskiy
- National University , Department of Mathematics and Natural Sciences, Los Angeles, California
| | - Iryna Pishel
- D.F. Chebotarev State Institute of Gerontology NAMS , Lab Pathophysiology and Immunology, Kyiv, Ukraine
| |
Collapse
|
748
|
Campos PB, Paulsen BS, Rehen SK. Accelerating neuronal aging in in vitro model brain disorders: a focus on reactive oxygen species. Front Aging Neurosci 2014; 6:292. [PMID: 25386139 PMCID: PMC4209886 DOI: 10.3389/fnagi.2014.00292] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/06/2014] [Indexed: 12/20/2022] Open
Abstract
In this review, we discuss insights gained through the use of stem cell preparations regarding the modeling of neurological diseases, the need for aging neurons derived from pluripotent stem cells to further advance the study of late-onset adult neurological diseases, and the extent to which mechanisms linked to the mismanagement of reactive oxygen species (ROS). The context of these issues can be revealed using the three disease states of Parkinson’s (PD), Alzheimer’s (AD), and schizophrenia, as considerable insights have been gained into these conditions through the use of stem cells in terms of disease etiologies and the role of oxidative stress. The latter subject is a primary area of interest of our group. After discussing the molecular models of accelerated aging, we highlight the role of ROS for the three diseases explored here. Importantly, we do not seek to provide an extensive account of all genetic mutations for each of the three disorders discussed in this review, but we aim instead to provide a conceptual framework that could maximize the gains from merging the approaches of stem cell microsystems and the study of oxidative stress in disease in order to optimize therapeutics and determine new molecular targets against oxidative stress that spare stem cell proliferation and development.
Collapse
Affiliation(s)
- Priscila Britto Campos
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Bruna S Paulsen
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Stevens K Rehen
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro Rio de Janeiro, Brazil ; D'Or Institute for Research and Education (IDOR) Rio de Janeiro, Brazil
| |
Collapse
|
749
|
Enhancing dentate gyrus function with dietary flavanols improves cognition in older adults. Nat Neurosci 2014; 17:1798-803. [PMID: 25344629 DOI: 10.1038/nn.3850] [Citation(s) in RCA: 235] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 10/01/2014] [Indexed: 02/08/2023]
Abstract
The dentate gyrus (DG) is a region in the hippocampal formation whose function declines in association with human aging and is therefore considered to be a possible source of age-related memory decline. Causal evidence is needed, however, to show that DG-associated memory decline in otherwise healthy elders can be improved by interventions that enhance DG function. We addressed this issue by first using a high-resolution variant of functional magnetic resonance imaging (fMRI) to map the precise site of age-related DG dysfunction and to develop a cognitive task whose function localized to this anatomical site. Then, in a controlled randomized trial, we applied these tools to study healthy 50-69-year-old subjects who consumed either a high or low cocoa flavanol-containing diet for 3 months. A high-flavanol intervention was found to enhance DG function, as measured by fMRI and by cognitive testing. Our findings establish that DG dysfunction is a driver of age-related cognitive decline and suggest non-pharmacological means for its amelioration.
Collapse
|
750
|
Al-Amin MM, Hasan SMN, Alam T, Hasan AT, Hossain I, Didar RR, Alam MA, Rahman MM. Tadalafil enhances working memory, and reduces hippocampal oxidative stress in both young and aged mice. Eur J Pharmacol 2014; 745:84-90. [PMID: 25446565 DOI: 10.1016/j.ejphar.2014.10.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 10/13/2014] [Accepted: 10/14/2014] [Indexed: 10/24/2022]
Abstract
Tadalafil, a type-5 phosphodiesterase enzyme inhibitor with long half-life used to treat erectile dysfunction. Recently it has been reported that tadalafil improves cognitive function. Here, we aimed to investigate the age dependent effects of tadalafil on memory, locomotor, behavior, and oxidative stress in the hippocampus. Tadalafil was orally administered everyday (5 mg/kg) to young (2 months) and old (16 months) healthy mice for 4 weeks. Control mice from each group received equal volume of 0.9% normal saline for the same duration. Memory and locomotor activity were tested using radial arm maze and open field test respectively. The level of malondialdehyde (MDA), nitric oxide (NO), and advanced protein oxidation product (APOP) was analyzed and catalase activity was determined from the isolated hippocampus. Treatment with tadalafil in aged mice improves working memory than the corresponding tadalafil treated young mice in radial arm maze test. Tadalafil treated mice traveled less distance in the center and the mean speed of tadalafil treated aged mice was significantly lower than the tadalafil treated young mice in open field test. Tadalafil treatment elicited a decrease of MDA level in the hippocampus of aged mice than that of young mice. APOP level was decreased only in aged mice treated with tadalafil. Treatment with tadalafil decreased NO and increased catalase activity in both young and aged mice. On the basis of previous and our findings, we conclude that tadalafil treatment reduces oxidative stress while increased cGMP level in the hippocampus might be responsible for memory enhancement.
Collapse
Affiliation(s)
- Md Mamun Al-Amin
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka 1229, Bangladesh.
| | - S M Nageeb Hasan
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Tanzir Alam
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Ahmed Tasdid Hasan
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Imran Hossain
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Rohini Rowshan Didar
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Md Ashraful Alam
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Md Mahbubur Rahman
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka 1229, Bangladesh
| |
Collapse
|