101
|
Role of Microglia in Modulating Adult Neurogenesis in Health and Neurodegeneration. Int J Mol Sci 2020; 21:ijms21186875. [PMID: 32961703 PMCID: PMC7555074 DOI: 10.3390/ijms21186875] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023] Open
Abstract
Microglia are the resident immune cells of the brain, constituting the powerhouse of brain innate immunity. They originate from hematopoietic precursors that infiltrate the developing brain during different stages of embryogenesis, acquiring a phenotype characterized by the presence of dense ramifications. Microglial cells play key roles in maintaining brain homeostasis and regulating brain immune responses. They continuously scan and sense the brain environment to detect any occurring changes. Upon detection of a signal related to physiological or pathological processes, the cells are activated and transform to an amoeboid-like phenotype, mounting adequate responses that range from phagocytosis to secretion of inflammatory and trophic factors. The overwhelming evidence suggests that microglia are crucially implicated in influencing neuronal proliferation and differentiation, as well as synaptic connections, and thereby cognitive and behavioral functions. Here, we review the role of microglia in adult neurogenesis under physiological conditions, and how this role is affected in neurodegenerative diseases.
Collapse
|
102
|
Berger T, Lee H, Young AH, Aarsland D, Thuret S. Adult Hippocampal Neurogenesis in Major Depressive Disorder and Alzheimer's Disease. Trends Mol Med 2020; 26:803-818. [PMID: 32418723 DOI: 10.1016/j.molmed.2020.03.010] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/16/2020] [Accepted: 03/31/2020] [Indexed: 12/25/2022]
Abstract
Depression and dementia are major public health problems. Major depressive disorder (MDD) and Alzheimer's disease (AD) reciprocally elevate the risk for one another. No effective drug is available to treat AD and about one-third of depressive patients show treatment resistance. The biological connection between MDD and AD is still unclear. Uncovering this link might open novel ways of treatment and prevention to improve patient healthcare. Here, we discuss recent studies specifically on the role of human adult hippocampal neurogenesis (AHN) in MDD and AD. We compare diverse approaches to analyse the effect of MDD and AD on human AHN and analyse different studies implicating the role of human AHN as a potential converging mechanism in MDD and AD.
Collapse
Affiliation(s)
- Thomas Berger
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Hyunah Lee
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Allan H Young
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London and South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks Orchard Road, Beckenham, Kent, UK
| | - Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
103
|
Neuropeptides Modulate Local Astrocytes to Regulate Adult Hippocampal Neural Stem Cells. Neuron 2020; 108:349-366.e6. [PMID: 32877641 DOI: 10.1016/j.neuron.2020.07.039] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 06/12/2020] [Accepted: 07/29/2020] [Indexed: 12/20/2022]
Abstract
Neural stem cells (NSCs) in the dentate gyrus (DG) reside in a specialized local niche that supports their neurogenic proliferation to produce adult-born neurons throughout life. How local niche cells interact at the circuit level to ensure continuous neurogenesis from NSCs remains unknown. Here we report the role of endogenous neuropeptide cholecystokinin (CCK), released from dentate CCK interneurons, in regulating neurogenic niche cells and NSCs. Specifically, stimulating CCK release supports neurogenic proliferation of NSCs through a dominant astrocyte-mediated glutamatergic signaling cascade. In contrast, reducing dentate CCK induces reactive astrocytes, which correlates with decreased neurogenic proliferation of NSCs and upregulation of genes involved in immune processes. Our findings provide novel circuit-based information on how CCK acts on local astrocytes to regulate the key behavior of adult NSCs.
Collapse
|
104
|
Polysialylation and disease. Mol Aspects Med 2020; 79:100892. [PMID: 32863045 DOI: 10.1016/j.mam.2020.100892] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 12/31/2022]
Abstract
Polysialic acid (polySia, PSA) is a unique constituent of the glycocalyx on the surface of bacterial and vertebrate cells. In vertebrates, its biosynthesis is highly regulated, not only in quantity and quality, but also in time and location, which allows polySia to be involved in various important biological phenomena. Therefore, impairments in the expression and structure of polySia sometimes relate to diseases, such as schizophrenia, bipolar disorder, and cancer. Some bacteria express polySia as a tool for protecting themselves from the host immune system during invasion. PolySia is proven to be a biosafe material; polySia, as well as polySia-recognizing molecules, are key therapeutic agents. This review first comprehensive outlines the occurrence, features, biosynthesis, and functions of polySia and subsequently focuses on the related diseases.
Collapse
|
105
|
Morrone CD, Bazzigaluppi P, Beckett TL, Hill ME, Koletar MM, Stefanovic B, McLaurin J. Regional differences in Alzheimer's disease pathology confound behavioural rescue after amyloid-β attenuation. Brain 2020; 143:359-373. [PMID: 31782760 PMCID: PMC6935751 DOI: 10.1093/brain/awz371] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/16/2019] [Accepted: 10/01/2019] [Indexed: 12/31/2022] Open
Abstract
Failure of Alzheimer’s disease clinical trials to improve or stabilize cognition has led to the need for a better understanding of the driving forces behind cognitive decline in the presence of active disease processes. To dissect contributions of individual pathologies to cognitive function, we used the TgF344-AD rat model, which recapitulates the salient hallmarks of Alzheimer’s disease pathology observed in patient populations (amyloid, tau inclusions, frank neuronal loss, and cognitive deficits). scyllo-Inositol treatment attenuated amyloid-β peptide in disease-bearing TgF344-AD rats, which rescued pattern separation in the novel object recognition task and executive function in the reversal learning phase of the Barnes maze. Interestingly, neither activities of daily living in the burrowing task nor spatial memory in the Barnes maze were rescued by attenuating amyloid-β peptide. To understand the pathological correlates leading to behavioural rescue, we examined the neuropathology and in vivo electrophysiological signature of the hippocampus. Amyloid-β peptide attenuation reduced hippocampal tau pathology and rescued adult hippocampal neurogenesis and neuronal function, via improvements in cross-frequency coupling between theta and gamma bands. To investigate mechanisms underlying the persistence of spatial memory deficits, we next examined neuropathology in the entorhinal cortex, a region whose input to the hippocampus is required for spatial memory. Reduction of amyloid-β peptide in the entorhinal cortex had no effect on entorhinal tau pathology or entorhinal-hippocampal neuronal network dysfunction, as measured by an impairment in hippocampal response to entorhinal stimulation. Thus, rescue or not of cognitive function is dependent on regional differences of amyloid-β, tau and neuronal network dysfunction, demonstrating the importance of staging disease in patients prior to enrolment in clinical trials. These results further emphasize the need for combination therapeutic approaches across disease progression.
Collapse
Affiliation(s)
- Christopher D Morrone
- Sunnybrook Research Institute, Biological Sciences, 2075 Bayview Ave, Toronto, ON, Canada.,University of Toronto, Faculty of Medicine, Department of Laboratory Medicine and Pathobiology, 1 King's College Cir, Toronto, ON, Canada
| | - Paolo Bazzigaluppi
- Sunnybrook Research Institute, Physical Sciences, 2075 Bayview Ave, Toronto, ON, Canada
| | - Tina L Beckett
- Sunnybrook Research Institute, Biological Sciences, 2075 Bayview Ave, Toronto, ON, Canada
| | - Mary E Hill
- Sunnybrook Research Institute, Biological Sciences, 2075 Bayview Ave, Toronto, ON, Canada
| | - Margaret M Koletar
- Sunnybrook Research Institute, Physical Sciences, 2075 Bayview Ave, Toronto, ON, Canada
| | - Bojana Stefanovic
- Sunnybrook Research Institute, Physical Sciences, 2075 Bayview Ave, Toronto, ON, Canada.,University of Toronto, Faculty of Medicine, Department of Medical Biophysics, 101 College St Suite 15-701, Toronto, ON, Canada
| | - JoAnne McLaurin
- Sunnybrook Research Institute, Biological Sciences, 2075 Bayview Ave, Toronto, ON, Canada.,University of Toronto, Faculty of Medicine, Department of Laboratory Medicine and Pathobiology, 1 King's College Cir, Toronto, ON, Canada
| |
Collapse
|
106
|
Vilor-Tejedor N, Operto G, Evans TE, Falcon C, Crous-Bou M, Minguillón C, Cacciaglia R, Milà-Alomà M, Grau-Rivera O, Suárez-Calvet M, Garrido-Martín D, Morán S, Esteller M, Adams HH, Molinuevo JL, Guigó R, Gispert JD. Effect of BDNF Val66Met on hippocampal subfields volumes and compensatory interaction with APOE-ε4 in middle-age cognitively unimpaired individuals from the ALFA study. Brain Struct Funct 2020; 225:2331-2345. [PMID: 32804326 PMCID: PMC7544723 DOI: 10.1007/s00429-020-02125-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/30/2020] [Indexed: 11/08/2022]
Abstract
Background Current evidence supports the involvement of brain-derived neurotrophic factor (BDNF) Val66Met polymorphism, and the ε4 allele of APOE gene in hippocampal-dependent functions. Previous studies on the association of Val66Met with whole hippocampal volume included patients of a variety of disorders. However, it remains to be elucidated whether there is an impact of BDNF Val66Met polymorphism on the volumes of the hippocampal subfield volumes (HSv) in cognitively unimpaired (CU) individuals, and the interactive effect with the APOE-ε4 status. Methods BDNF Val66Met and APOE genotypes were determined in a sample of 430 CU late/middle-aged participants from the ALFA study (ALzheimer and FAmilies). Participants underwent a brain 3D-T1-weighted MRI scan, and volumes of the HSv were determined using Freesurfer (v6.0). The effects of the BDNF Val66Met genotype on the HSv were assessed using general linear models corrected by age, gender, education, number of APOE-ε4 alleles and total intracranial volume. We also investigated whether the association between APOE-ε4 allele and HSv were modified by BDNF Val66Met genotypes. Results BDNF Val66Met carriers showed larger bilateral volumes of the subiculum subfield. In addition, HSv reductions associated with APOE-ε4 allele were significantly moderated by BDNF Val66Met status. BDNF Met carriers who were also APOE-ε4 homozygous showed patterns of higher HSv than BDNF Val carriers. Conclusion To our knowledge, the present study is the first to show that carrying the BDNF Val66Met polymorphisms partially compensates the decreased on HSv associated with APOE-ε4 in middle-age cognitively unimpaired individuals. Electronic supplementary material The online version of this article (10.1007/s00429-020-02125-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Natalia Vilor-Tejedor
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, C. Doctor Aiguader 88, Edif. PRBB, 08003, Barcelona, Spain. .,Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain. .,Erasmus MC University Medical Center Rotterdam, Department of Clinical Genetics, Rotterdam, The Netherlands. .,Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| | - Grégory Operto
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Tavia E Evans
- Erasmus MC University Medical Center Rotterdam, Department of Clinical Genetics, Rotterdam, The Netherlands
| | - Carles Falcon
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Marta Crous-Bou
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO), Hospitalet de Llobregat, Barcelona, Spain
| | - Carolina Minguillón
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Raffaele Cacciaglia
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Marta Milà-Alomà
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Oriol Grau-Rivera
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Servei de Neurologia, Hospital del Mar, Barcelona, Spain
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Servei de Neurologia, Hospital del Mar, Barcelona, Spain
| | - Diego Garrido-Martín
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, C. Doctor Aiguader 88, Edif. PRBB, 08003, Barcelona, Spain
| | - Sebastián Morán
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Manel Esteller
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Biomedical Research Institute (IDIBELL), Barcelona, Spain.,Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.,Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain
| | - Hieab H Adams
- Erasmus MC University Medical Center Rotterdam, Department of Clinical Genetics, Rotterdam, The Netherlands.,Erasmus MC University Medical Center Rotterdam, Department of Epidemiology, Rotterdam, The Netherlands.,Erasmus MC University Medical Center Rotterdam, Department of Radiology, Rotterdam, The Netherlands
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Roderic Guigó
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, C. Doctor Aiguader 88, Edif. PRBB, 08003, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain. .,Universitat Pompeu Fabra (UPF), Barcelona, Spain. .,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain.
| | | |
Collapse
|
107
|
Seki T. Understanding the Real State of Human Adult Hippocampal Neurogenesis From Studies of Rodents and Non-human Primates. Front Neurosci 2020; 14:839. [PMID: 32848586 PMCID: PMC7432251 DOI: 10.3389/fnins.2020.00839] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/20/2020] [Indexed: 12/11/2022] Open
Abstract
The concept of adult hippocampal neurogenesis (AHN) has been widely accepted, and a large number of studies have been performed in rodents using modern experimental techniques, which have clarified the nature and developmental processes of adult neural stem/progenitor cells, the functions of AHN, such as memory and learning, and its association with neural diseases. However, a fundamental problem is that it remains unclear as to what extent AHN actually occurs in humans. The answer to this is indispensable when physiological and pathological functions of human AHN are deduced from studies of rodent AHN, but there are controversial data on the extent of human AHN. In this review, studies on AHN performed in rodents and humans will be briefly reviewed, followed by a discussion of the studies in non-human primates. Then, how data of rodent and non-human primate AHN should be applied for understanding human AHN will be discussed.
Collapse
Affiliation(s)
- Tatsunori Seki
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
108
|
Jiang M, Vanan S, Tu HT, Zhang W, Zhang ZW, Chia SY, Jang SE, Zeng XX, Yu WP, Xu J, Guo KH, Zeng L. Amyloid precursor protein intracellular domain-dependent regulation of FOXO3a inhibits adult hippocampal neurogenesis. Neurobiol Aging 2020; 95:250-263. [PMID: 32866886 DOI: 10.1016/j.neurobiolaging.2020.07.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 07/14/2020] [Accepted: 07/29/2020] [Indexed: 10/23/2022]
Abstract
The amyloid precursor protein (APP) intracellular domain (AICD) is a metabolic by-product of APP produced through sequential proteolytic cleavage by α-, β-, and γ-secretases. The interaction between AICD and Fe65 has been reported to impair adult neurogenesis in vivo. However, the exact role of AICD in mediating neural stem cell fate remains unclear. To identify the role of AICD in neuronal proliferation and differentiation, as well as to clarify the molecular mechanisms underlying the role of AICD in neurogenesis, we first generated a mouse model expressing the Rosa26-based AICD transgene. AICD overexpression did not alter the spatiotemporal expression pattern of full-length APP or accumulation of its metabolites. In addition, AICD decreased the newly generated neural progenitor cell (NPC) pool, inhibited the proliferation and differentiation efficiency of NPCs, and increased cell death both in vitro and in vivo. Given that abnormal neurogenesis is often associated with depression-like behavior in adult mice, we conducted a forced swim test and tail suspension test with AICD mice and found a depression-like behavioral phenotype in AICD transgenic mice. Moreover, AICD stimulated FOXO3a transcriptional activation, which in turn negatively regulated AICD. In addition, functional loss of FOXO3a in NPCs derived from the hippocampal dentate gyrus of adult AICD transgenic mice rescued neurogenesis defects. AICD also increased the mRNA expression of FOXO3a target genes related to neurogenesis and cell death. These results suggest that FOXO3a is the functional target of AICD in neurogenesis regulation. Our study reveals the role of AICD in mediating neural stem cell fate to maintain homeostasis during brain development via interaction with FOXO3a.
Collapse
Affiliation(s)
- Mei Jiang
- Department of Neurobiology and Anatomy, Sun Yat-Sen University Zhongshan School of Medicine, Guangzhou, PR China; Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Sarivin Vanan
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Hai-Tao Tu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Wei Zhang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Zhi-Wei Zhang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Sook-Yoong Chia
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Se Eun Jang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Xiao-Xia Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Wei-Ping Yu
- Animal Gene Editing Laboratory, Biological resource Centre, A∗STAR, Singapore; Institute of Molecular and Cell Biology, A∗STAR, Proteos, Singapore
| | - Jie Xu
- Department of Neurobiology and Anatomy, Sun Yat-Sen University Zhongshan School of Medicine, Guangzhou, PR China.
| | - Kai-Hua Guo
- Department of Neurobiology and Anatomy, Sun Yat-Sen University Zhongshan School of Medicine, Guangzhou, PR China.
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore; Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore; Lee Kong Chian School of Medicine, Nanyang Technology University, Novena Campus, Singapore.
| |
Collapse
|
109
|
Calió ML, Henriques E, Siena A, Bertoncini CRA, Gil-Mohapel J, Rosenstock TR. Mitochondrial Dysfunction, Neurogenesis, and Epigenetics: Putative Implications for Amyotrophic Lateral Sclerosis Neurodegeneration and Treatment. Front Neurosci 2020; 14:679. [PMID: 32760239 PMCID: PMC7373761 DOI: 10.3389/fnins.2020.00679] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive and devastating multifactorial neurodegenerative disorder. Although the pathogenesis of ALS is still not completely understood, numerous studies suggest that mitochondrial deregulation may be implicated in its onset and progression. Interestingly, mitochondrial deregulation has also been associated with changes in neural stem cells (NSC) proliferation, differentiation, and migration. In this review, we highlight the importance of mitochondrial function for neurogenesis, and how both processes are correlated and may contribute to the pathogenesis of ALS; we have focused primarily on preclinical data from animal models of ALS, since to date no studies have evaluated this link using human samples. As there is currently no cure and no effective therapy to counteract ALS, we have also discussed how improving neurogenic function by epigenetic modulation could benefit ALS. In support of this hypothesis, changes in histone deacetylation can alter mitochondrial function, which in turn might ameliorate cellular proliferation as well as neuronal differentiation and migration. We propose that modulation of epigenetics, mitochondrial function, and neurogenesis might provide new hope for ALS patients, and studies exploring these new territories are warranted in the near future.
Collapse
Affiliation(s)
| | - Elisandra Henriques
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Science, São Paulo, Brazil
| | - Amanda Siena
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Science, São Paulo, Brazil
| | - Clélia Rejane Antonio Bertoncini
- CEDEME, Center of Development of Experimental Models for Medicine and Biology, Federal University of São Paulo, São Paulo, Brazil
| | - Joana Gil-Mohapel
- Division of Medical Sciences, Faculty of Medicine, University of Victoria and Island Medical Program, University of British Columbia, Victoria, BC, Canada
| | - Tatiana Rosado Rosenstock
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Science, São Paulo, Brazil
| |
Collapse
|
110
|
Kandasamy M, Anusuyadevi M, Aigner KM, Unger MS, Kniewallner KM, de Sousa DMB, Altendorfer B, Mrowetz H, Bogdahn U, Aigner L. TGF-β Signaling: A Therapeutic Target to Reinstate Regenerative Plasticity in Vascular Dementia? Aging Dis 2020; 11:828-850. [PMID: 32765949 PMCID: PMC7390515 DOI: 10.14336/ad.2020.0222] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/22/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular dementia (VaD) is the second leading form of memory loss after Alzheimer's disease (AD). Currently, there is no cure available. The etiology, pathophysiology and clinical manifestations of VaD are extremely heterogeneous, but the impaired cerebral blood flow (CBF) represents a common denominator of VaD. The latter might be the result of atherosclerosis, amyloid angiopathy, microbleeding and micro-strokes, together causing blood-brain barrier (BBB) dysfunction and vessel leakage, collectively originating from the consequence of hypertension, one of the main risk factors for VaD. At the histopathological level, VaD displays abnormal vascular remodeling, endothelial cell death, string vessel formation, pericyte responses, fibrosis, astrogliosis, sclerosis, microglia activation, neuroinflammation, demyelination, white matter lesions, deprivation of synapses and neuronal loss. The transforming growth factor (TGF) β has been identified as one of the key molecular factors involved in the aforementioned various pathological aspects. Thus, targeting TGF-β signaling in the brain might be a promising therapeutic strategy to mitigate vascular pathology and improve cognitive functions in patients with VaD. This review revisits the recent understanding of the role of TGF-β in VaD and associated pathological hallmarks. It further explores the potential to modulate certain aspects of VaD pathology by targeting TGF-β signaling.
Collapse
Affiliation(s)
- Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India.
- Faculty Recharge Programme, University Grants Commission (UGC-FRP), New Delhi, India.
| | - Muthuswamy Anusuyadevi
- Molecular Gerontology Group, Department of Biochemistry, School of Life Sciences, Bharathidhasan University, Tiruchirappalli, Tamil Nadu, India.
| | - Kiera M Aigner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Michael S Unger
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Kathrin M Kniewallner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Diana M Bessa de Sousa
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Barbara Altendorfer
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Heike Mrowetz
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Ulrich Bogdahn
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
- Velvio GmbH, Regensburg, Germany.
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
111
|
Labusch M, Mancini L, Morizet D, Bally-Cuif L. Conserved and Divergent Features of Adult Neurogenesis in Zebrafish. Front Cell Dev Biol 2020; 8:525. [PMID: 32695781 PMCID: PMC7338623 DOI: 10.3389/fcell.2020.00525] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/03/2020] [Indexed: 12/14/2022] Open
Abstract
Adult neurogenesis, i.e., the generation of neurons from neural stem cells (NSCs) in the adult brain, contributes to brain plasticity in all vertebrates. It varies, however, greatly in extent, location and physiological characteristics between species. During the last decade, the teleost zebrafish (D. rerio) was increasingly used to study the molecular and cellular properties of adult NSCs, in particular as a prominent NSC population was discovered at the ventricular surface of the dorsal telencephalon (pallium), in territories homologous to the adult neurogenic niches of rodents. This model, for its specific features (large NSC population, amenability to intravital imaging, high regenerative capacity) allowed rapid progress in the characterization of basic adult NSC features. We review here these findings, with specific comparisons with the situation in rodents. We specifically discuss the cellular nature of NSCs (astroglial or neuroepithelial cells), their heterogeneities and their neurogenic lineages, and the mechanisms controlling NSC quiescence and fate choices, which all impact the neurogenic output. We further discuss the regulation of NSC activity in response to physiological triggers and non-physiological conditions such as regenerative contexts.
Collapse
Affiliation(s)
- Miriam Labusch
- Zebrafish Neurogenetics Unit, Institut Pasteur, UMR 3738, CNRS, Team Supported by the Ligue Nationale Contre le Cancer, Paris, France.,Sorbonne Université, Collège Doctoral, Paris, France
| | - Laure Mancini
- Zebrafish Neurogenetics Unit, Institut Pasteur, UMR 3738, CNRS, Team Supported by the Ligue Nationale Contre le Cancer, Paris, France.,Sorbonne Université, Collège Doctoral, Paris, France
| | - David Morizet
- Zebrafish Neurogenetics Unit, Institut Pasteur, UMR 3738, CNRS, Team Supported by the Ligue Nationale Contre le Cancer, Paris, France.,Sorbonne Université, Collège Doctoral, Paris, France
| | - Laure Bally-Cuif
- Zebrafish Neurogenetics Unit, Institut Pasteur, UMR 3738, CNRS, Team Supported by the Ligue Nationale Contre le Cancer, Paris, France
| |
Collapse
|
112
|
Soluble SORLA Enhances Neurite Outgrowth and Regeneration through Activation of the EGF Receptor/ERK Signaling Axis. J Neurosci 2020; 40:5908-5921. [PMID: 32601248 DOI: 10.1523/jneurosci.0723-20.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 01/01/2023] Open
Abstract
SORLA is a transmembrane trafficking protein associated with Alzheimer's disease risk. Although SORLA is abundantly expressed in neurons, physiological roles for SORLA remain unclear. Here, we show that cultured transgenic neurons overexpressing SORLA feature longer neurites, and accelerated neurite regeneration with wounding. Enhanced release of a soluble form of SORLA (sSORLA) is observed in transgenic mouse neurons overexpressing human SORLA, while purified sSORLA promotes neurite extension and regeneration. Phosphoproteomic analyses demonstrate enrichment of phosphoproteins related to the epidermal growth factor (EGFR)/ERK pathway in SORLA transgenic mouse hippocampus from both genders. sSORLA coprecipitates with EGFR in vitro, and sSORLA treatment increases EGFR Y1173 phosphorylation, which is involved in ERK activation in cultured neurons. Furthermore, sSORLA triggers ERK activation, whereas pharmacological EGFR or ERK inhibition reverses sSORLA-dependent enhancement of neurite outgrowth. In search for downstream ERK effectors activated by sSORLA, we identified upregulation of Fos expression in hippocampus from male mice overexpressing SORLA by RNAseq analysis. We also found that Fos is upregulated and translocates to the nucleus in an ERK-dependent manner in neurons treated with sSORLA. Together, these results demonstrate that sSORLA is an EGFR-interacting protein that activates EGFR/ERK/Fos signaling to enhance neurite outgrowth and regeneration.SIGNIFICANCE STATEMENT SORLA is a transmembrane trafficking protein previously known to reduce the levels of amyloid-β, which is critical in the pathogenesis of Alzheimer's disease. In addition, SORLA mutations are a risk factor for Alzheimer's disease. Interestingly, the SORLA ectodomain is cleaved into a soluble form, sSORLA, which has been shown to regulate cytoskeletal signaling pathways and cell motility in cells outside the nervous system. We show here that sSORLA binds and activates the EGF receptor to induce downstream signaling through the ERK serine/threonine kinase and the Fos transcription factor, thereby enhancing neurite outgrowth. These findings reveal a novel role for sSORLA in promoting neurite regeneration through the EGF receptor/ERK/Fos pathway, thereby demonstrating a potential neuroprotective mechanism involving SORLA.
Collapse
|
113
|
Synaptic Loss, ER Stress and Neuro-Inflammation Emerge Late in the Lateral Temporal Cortex and Associate with Progressive Tau Pathology in Alzheimer's Disease. Mol Neurobiol 2020; 57:3258-3272. [PMID: 32514860 PMCID: PMC7340653 DOI: 10.1007/s12035-020-01950-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/22/2020] [Indexed: 11/17/2022]
Abstract
The complex multifactorial nature of AD pathogenesis has been highlighted by evidence implicating additional neurodegenerative mechanisms, beyond that of amyloid-β (Aβ) and tau. To provide insight into cause and effect, we here investigated the temporal profile and associations of pathological changes in synaptic, endoplasmic reticulum (ER) stress and neuro-inflammatory markers. Quantifications were established via immunoblot and immunohistochemistry protocols in post-mortem lateral temporal cortex (n = 46). All measures were assessed according to diagnosis (non-AD vs. AD), neuropathological severity (low (Braak ≤ 2) vs. moderate (3–4) vs. severe (≥ 5)) and individual Braak stage, and were correlated with Aβ and tau pathology and cognitive scores. Postsynaptic PSD-95, but not presynaptic synaptophysin, was decreased in AD cases and demonstrated a progressive decline across disease severity and Braak stage, yet not with cognitive scores. Of all investigated ER stress markers, only phospho-protein kinase RNA-like ER kinase (p-PERK) correlated with Braak stage and was increased in diagnosed AD cases. A similar relationship was observed for the astrocytic glial fibrillary acidic protein (GFAP); however, the associated aquaporin 4 and microglial Iba1 remained unchanged. Pathological alterations in these markers preferentially correlated with measures of tau over those related to Aβ. Notably, GFAP also correlated strongly with Aβ markers and with all assessments of cognition. Lateral temporal cortex-associated synaptic, ER stress and neuro-inflammatory pathologies are here determined as late occurrences in AD progression, largely associated with tau pathology. Moreover, GFAP emerged as the most robust indicator of disease progression, tau/Aβ pathology, and cognitive impairment.
Collapse
|
114
|
Traumatic brain injury and hippocampal neurogenesis: Functional implications. Exp Neurol 2020; 331:113372. [PMID: 32504636 DOI: 10.1016/j.expneurol.2020.113372] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 05/23/2020] [Accepted: 05/30/2020] [Indexed: 12/15/2022]
Abstract
In the adult brain, self-renewing radial-glia like (RGL) progenitor cells have been shown to reside in the subventricular zone and the subgranular zone of the hippocampus. A large body of evidence shows that experiences such as learning, enriched environment and stress can alter proliferation and differentiation of RGL progenitor cells. The progenitor cells present in the subgranular zone of the hippocampus divide to give rise to newborn neurons that migrate to the dentate gyrus where they differentiate into adult granule neurons. These newborn neurons have been found to have a unique role in certain types of hippocampus-dependent learning and memory, including goal-directed behaviors that require pattern separation. Experimental traumatic brain injury (TBI) in rodents has been shown to alter hippocampal neurogenesis, including triggering the acute loss of newborn neurons, as well as progenitor cell hyper-proliferation. In this review, we discuss the role of hippocampal neurogenesis in learning and memory. Furthermore, we review evidence for the molecular mechanisms that contribute to newborn neuron loss, as well as increased progenitor cell proliferation after TBI. Finally, we discuss strategies aimed at enhancing neurogenesis after TBI and their possible therapeutic benefits.
Collapse
|
115
|
Núñez C, Callén A, Lombardini F, Compta Y, Stephan-Otto C. Different Cortical Gyrification Patterns in Alzheimer's Disease and Impact on Memory Performance. Ann Neurol 2020; 88:67-80. [PMID: 32277502 DOI: 10.1002/ana.25741] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The study of cortical gyrification in Alzheimer's disease (AD) could help to further understanding of the changes undergone in the brain during neurodegeneration. Here, we aimed to study brain gyrification differences between healthy controls (HC), mild cognitive impairment (MCI) patients, and AD patients, and explore how cerebral gyrification patterns were associated with memory and other cognitive functions. METHODS We applied surface-based morphometry techniques in 2 large, independent cross-sectional samples, obtained from the Alzheimer's Disease Neuroimaging Initiative project. Both samples, encompassing a total of 1,270 participants, were analyzed independently. RESULTS Unexpectedly, we found that AD patients presented a more gyrificated entorhinal cortex than HC. Conversely, the insular cortex of AD patients was hypogyrificated. A decrease in the gyrification of the insular cortex was also found in older HC participants as compared with younger HC, which argues against the specificity of this finding in AD. However, an increased degree of folding of the insular cortex was specifically associated with better memory function and semantic fluency, only in AD patients. Overall, MCI patients presented an intermediate gyrification pattern. All these findings were consistently observed in the two samples. INTERPRETATION The marked atrophy of the medial temporal lobe observed in AD patients may explain the increased folding of the entorhinal cortex. We additionally speculate regarding alternative mechanisms that may also alter its folding. The association between increased gyrification of the insular cortex and memory function, specifically observed in AD, could be suggestive of compensatory mechanisms to overcome the loss of memory function. ANN NEUROL 2020 ANN NEUROL 2020;88:67-80.
Collapse
Affiliation(s)
- Christian Núñez
- Parc Sanitari Sant Joan de Déu, Sant Boi de Llobregat, Barcelona, Spain.,Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Antonio Callén
- Parc Sanitari Sant Joan de Déu, Sant Boi de Llobregat, Barcelona, Spain.,Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Federica Lombardini
- Parc Sanitari Sant Joan de Déu, Sant Boi de Llobregat, Barcelona, Spain.,Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Yaroslau Compta
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona & Maria de Maeztu Excellence Center Institute of Neuroscience, University of Barcelona, Barcelona, Spain.,Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Spain
| | - Christian Stephan-Otto
- Parc Sanitari Sant Joan de Déu, Sant Boi de Llobregat, Barcelona, Spain.,Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | | |
Collapse
|
116
|
Tai W, Xu XM, Zhang CL. Regeneration Through in vivo Cell Fate Reprogramming for Neural Repair. Front Cell Neurosci 2020; 14:107. [PMID: 32390804 PMCID: PMC7193690 DOI: 10.3389/fncel.2020.00107] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/07/2020] [Indexed: 12/30/2022] Open
Abstract
The adult mammalian central nervous system (CNS) has very limited regenerative capacity upon neural injuries or under degenerative conditions. In recent years, however, significant progress has been made on in vivo cell fate reprogramming for neural regeneration. Resident glial cells can be reprogrammed into neuronal progenitors and mature neurons in the CNS of adult mammals. In this review article, we briefly summarize the current knowledge on innate adult neurogenesis under pathological conditions and then focus on induced neurogenesis through cell fate reprogramming. We discuss how the reprogramming process can be regulated and raise critical issues requiring careful considerations to move the field forward. With emerging evidence, we envision that fate reprogramming-based regenerative medicine will have a great potential for treating neurological conditions such as brain injury, spinal cord injury (SCI), Alzheimer’s disease (AD), Parkinson’s disease (PD), and retinopathy.
Collapse
Affiliation(s)
- Wenjiao Tai
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indianapolis, IN, United States.,Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
117
|
Kim S, Kim N, Lee J, Kim S, Hong J, Son S, Do Heo W. Dynamic Fas signaling network regulates neural stem cell proliferation and memory enhancement. SCIENCE ADVANCES 2020; 6:eaaz9691. [PMID: 32494656 PMCID: PMC7176421 DOI: 10.1126/sciadv.aaz9691] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/31/2020] [Indexed: 06/11/2023]
Abstract
Activation of Fas (CD95) is observed in various neurological disorders and can lead to both apoptosis and prosurvival outputs, yet how Fas signaling operates dynamically in the hippocampus is poorly understood. The optogenetic dissection of a signaling network can yield molecular-level explanations for cellular responses or fates, including the signaling dysfunctions seen in numerous diseases. Here, we developed an optogenetically activatable Fas that works in a physiologically plausible manner. Fas activation in immature neurons of the dentate gyrus triggered mammalian target of rapamycin (mTOR) activation and subsequent brain-derived neurotrophic factor secretion. Phosphorylation of extracellular signal-regulated kinase (Erk) in neural stem cells was induced under prolonged Fas activation. Repetitive activation of this signaling network yielded proliferation of neural stem cells and a transient increase in spatial working memory in mice. Our results demonstrate a novel Fas signaling network in the dentate gyrus and illuminate its consequences for adult neurogenesis and memory enhancement.
Collapse
Affiliation(s)
- Seokhwi Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Nury Kim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Jinsu Lee
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea
| | - Sungsoo Kim
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea
| | - Jongryul Hong
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea
| | - Seungkyu Son
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea
| | - Won Do Heo
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea
- KAIST Institute for the BioCentury, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
118
|
Sahab-Negah S, Hajali V, Moradi HR, Gorji A. The Impact of Estradiol on Neurogenesis and Cognitive Functions in Alzheimer's Disease. Cell Mol Neurobiol 2020; 40:283-299. [PMID: 31502112 DOI: 10.1007/s10571-019-00733-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/31/2019] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is described as cognitive and memory impairments with a sex-related epidemiological profile, affecting two times more women than men. There is emerging evidence that alternations in the hippocampal neurogenesis occur at the early stage of AD. Therapies that may effectively slow, stop, or regenerate the dying neurons in AD are being extensively investigated in the last few decades, but none has yet been found to be effective. The regulation of endogenous neurogenesis is one of the main therapeutic targets for AD. Mounting evidence indicates that the neurosteroid estradiol (17β-estradiol) plays a supporting role in neurogenesis, neuronal activity, and synaptic plasticity of AD. This effect may provide preventive and/or therapeutic approaches for AD. In this article, we discuss the molecular mechanism of potential estradiol modulatory action on endogenous neurogenesis, synaptic plasticity, and cognitive function in AD.
Collapse
Affiliation(s)
- Sajad Sahab-Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Vahid Hajali
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Moradi
- Histology and Embryology Group, Basic Science Department, Faculty of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ali Gorji
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neurosurgery and Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany.
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Domagkstr. 11, Münster, Germany.
| |
Collapse
|
119
|
Cranston AL, Wysocka A, Steczkowska M, Zadrożny M, Palasz E, Harrington CR, Theuring F, Wischik CM, Riedel G, Niewiadomska G. Cholinergic and inflammatory phenotypes in transgenic tau mouse models of Alzheimer's disease and frontotemporal lobar degeneration. Brain Commun 2020; 2:fcaa033. [PMID: 32954291 PMCID: PMC7425524 DOI: 10.1093/braincomms/fcaa033] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 01/23/2023] Open
Abstract
An early and sizeable loss of basal forebrain cholinergic neurons is a well-characterized feature associated with measurable deficits in spatial learning and cognitive impairment in patients with Alzheimer’s disease. In addition, pro-inflammatory glial cells such as astrocytes and microglia may play a key role in the neurodegenerative cascade of Alzheimer’s disease and tauopathies. We recently presented two mouse models: Line 1, expressing the truncated tau fragment identified as the core of the Alzheimer’s paired helical filament, and Line 66, expressing full-length human tau carrying a double mutation (P301S and G335D). Line 1 mice have a pathology that is akin to Alzheimer’s, whilst Line 66 resembles frontotemporal lobar degeneration. However, their cholinergic and inflammatory phenotypes remain elusive. We performed histological evaluation of choline acetyltransferase, acetylcholinesterase, p75 neurotrophin receptor, microglial ionized calcium binding adaptor molecule 1 and astrocytic glial fibrillary acidic protein in the basal forebrain, hippocampus and cortex of these models. A significant lowering of choline acetyltransferase-positive neurons and p75-positive neurons in the basal forebrain of Line 1 at 3, 6 and 9 months was observed in two independent studies, alongside a significant decrease in acetylcholinesterase staining in the cortex and hippocampus. The reductions in choline acetyltransferase positivity varied between 30% and 50% at an age when Line 1 mice show spatial learning impairments. Furthermore, an increase in microglial ionized calcium binding adaptor molecule 1 staining was observed in the basal forebrain, hippocampus and entorhinal cortex of Line 1 at 6 months. Line 66 mice displayed an intact cholinergic basal forebrain, and no difference in p75-positive neurons at 3 or 9 months. In addition, Line 66 exhibited significant microglial ionized calcium binding adaptor molecule 1 increase in the basal forebrain and hippocampus, suggesting a prominent neuroinflammatory profile. Increased concentrations of microglial interleukin-1β and astrocytic complement 3 were also seen in the hippocampus of both Line 1 and Line 66. The cholinergic deficit in Line 1 mice confirms the Alzheimer’s disease-like phenotype in Line 1 mice, whilst Line 66 revealed no measurable change in total cholinergic expression, a phenotypic trait of frontotemporal lobar degeneration. These two transgenic lines are therefore suitable for discriminating mechanistic underpinnings between the Alzheimer’s and frontotemporal lobar degeneration-like phenotypes of these mice.
Collapse
Affiliation(s)
- Anna L Cranston
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Adrianna Wysocka
- Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw 02-093, Poland
| | | | | | - Ewelina Palasz
- Mossakowski Medical Research Centre, Warsaw 02-106, Poland
| | - Charles R Harrington
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.,TauRx Therapeutics Ltd, Foresterhill, Aberdeen AB25 2ZP, UK
| | - Franz Theuring
- Institute of Pharmacology, Charite-Universitätsmedizin Berlin, Berlin, Germany
| | - Claude M Wischik
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.,TauRx Therapeutics Ltd, Aberdeen AB24 5RP, UK
| | - Gernot Riedel
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Grazyna Niewiadomska
- Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw 02-093, Poland
| |
Collapse
|
120
|
Hierro-Bujalance C, Del Marco A, José Ramos-Rodríguez J, Infante-Garcia C, Bella Gomez-Santos S, Herrera M, Garcia-Alloza M. Cell proliferation and neurogenesis alterations in Alzheimer's disease and diabetes mellitus mixed murine models. J Neurochem 2020; 154:673-692. [PMID: 32068886 DOI: 10.1111/jnc.14987] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 02/07/2020] [Accepted: 02/07/2020] [Indexed: 12/22/2022]
Abstract
The classic neuropathological features of Alzheimer's disease (AD) are accompanied by other complications, including alterations in adult cell proliferation and neurogenesis. Moreover recent studies have shown that traditional markers of the neurogenic process, such as doublecortin (DCX), may also be expressed in CD8+ T cells and ionized calcium-binding adaptor molecule 1 (Iba1+ ) microglia, in the close proximity to senile plaques, increasing the complexity of the condition. Altered glucose tolerance, observed in metabolic alteratioins, may accelerate the neurodegenerative process and interfere with normal adult cell proliferation and neurogenesis. To further explore the role of metabolic disease in AD, we analyzed cell proliferation and neurogenesis using 5'-bromo-2'-deoxyuridine and DCX immunohistochemistry in three different mouse models of AD and metabolic alterations: APP/PS1xdb/db mice, APP/PS1 mice on a long-term high-fat diet, and APP/PS1 mice treated with streptozotozin. As reported previously, an overall reduction in cell proliferation and neurogenesis was observed after streptozotocin administration. In contrast, an increase in cell proliferation and neurogenesis was detected in neurogenic niches in 14- and 26-week-old APP/PS1xdb/db mice, accompanied by a slight increase in cortical cell proliferation. While a similar trend was observed in animals on a high-fat diet, differences were not statistically significant. We observed very few DCX+ /CD8+ cells and no DCX+ /Iba1+ cells were observed in the close proximity to senile plaques in any of the groups. Interestingly, metabolic parameters such as body weight and glucose and insulin levels were identified as reliable predictors of cell proliferation and neurogenesis in APP/PS1xdb/db mice. Furthermore, metabolic parameters were also associated with altered Aβ levels in the cortex and hippocampus of APP/PS1xdb/db mice. Altogether, our data suggest that metabolic disease may also interfere with central complications in AD.
Collapse
Affiliation(s)
- Carmen Hierro-Bujalance
- Division of Physiology. School of Medicine, Edificio Andrés Segovia. C/Dr. Marañón 3, 3er piso, (11002) Cadiz. Universidad de Cadiz, Cadiz, Spain.,Instituto de Investigación Biomédica e Innovación en Ciencias Biomédicas de la Provincia de Cadiz (INiBICA), Cadiz, Spain
| | - Angel Del Marco
- Division of Physiology. School of Medicine, Edificio Andrés Segovia. C/Dr. Marañón 3, 3er piso, (11002) Cadiz. Universidad de Cadiz, Cadiz, Spain.,Instituto de Investigación Biomédica e Innovación en Ciencias Biomédicas de la Provincia de Cadiz (INiBICA), Cadiz, Spain
| | - Juan José Ramos-Rodríguez
- Division of Physiology. School of Medicine, Edificio Andrés Segovia. C/Dr. Marañón 3, 3er piso, (11002) Cadiz. Universidad de Cadiz, Cadiz, Spain
| | - Carmen Infante-Garcia
- Division of Physiology. School of Medicine, Edificio Andrés Segovia. C/Dr. Marañón 3, 3er piso, (11002) Cadiz. Universidad de Cadiz, Cadiz, Spain.,Instituto de Investigación Biomédica e Innovación en Ciencias Biomédicas de la Provincia de Cadiz (INiBICA), Cadiz, Spain
| | - Sara Bella Gomez-Santos
- Division of Physiology. School of Medicine, Edificio Andrés Segovia. C/Dr. Marañón 3, 3er piso, (11002) Cadiz. Universidad de Cadiz, Cadiz, Spain
| | - Marta Herrera
- Division of Physiology. School of Medicine, Edificio Andrés Segovia. C/Dr. Marañón 3, 3er piso, (11002) Cadiz. Universidad de Cadiz, Cadiz, Spain.,Instituto de Investigación Biomédica e Innovación en Ciencias Biomédicas de la Provincia de Cadiz (INiBICA), Cadiz, Spain
| | - Monica Garcia-Alloza
- Division of Physiology. School of Medicine, Edificio Andrés Segovia. C/Dr. Marañón 3, 3er piso, (11002) Cadiz. Universidad de Cadiz, Cadiz, Spain.,Instituto de Investigación Biomédica e Innovación en Ciencias Biomédicas de la Provincia de Cadiz (INiBICA), Cadiz, Spain
| |
Collapse
|
121
|
Neuroprotective Role of Dietary Supplementation with Omega-3 Fatty Acids in the Presence of Basal Forebrain Cholinergic Neurons Degeneration in Aged Mice. Int J Mol Sci 2020; 21:ijms21051741. [PMID: 32143275 PMCID: PMC7084583 DOI: 10.3390/ijms21051741] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 02/24/2020] [Accepted: 03/01/2020] [Indexed: 01/05/2023] Open
Abstract
As major components of neuronal membranes, omega-3 polyunsaturated fatty acids (n-3 PUFA) exhibit a wide range of regulatory functions. Recent human and animal studies indicate that n-3 PUFA may exert beneficial effects on aging processes. Here we analyzed the neuroprotective influence of n-3 PUFA supplementation on behavioral deficits, hippocampal neurogenesis, volume loss, and astrogliosis in aged mice that underwent a selective depletion of basal forebrain cholinergic neurons. Such a lesion represents a valid model to mimic a key component of the cognitive deficits associated with dementia. Aged mice were supplemented with n-3 PUFA or olive oil (as isocaloric control) for 8 weeks and then cholinergically depleted with mu-p75-saporin immunotoxin. Two weeks after lesioning, mice were behaviorally tested to assess anxious, motivational, social, mnesic, and depressive-like behaviors. Subsequently, morphological and biochemical analyses were performed. In lesioned aged mice the n-3 PUFA pre-treatment preserved explorative skills and associative retention memory, enhanced neurogenesis in the dentate gyrus, and reduced volume and VAChT levels loss as well as astrogliosis in hippocampus. The present findings demonstrating that n-3 PUFA supplementation before cholinergic depletion can counteract behavioral deficits and hippocampal neurodegeneration in aged mice advance a low-cost, non-invasive preventive tool to enhance life quality during aging.
Collapse
|
122
|
Lucassen PJ, Fitzsimons CP, Salta E, Maletic-Savatic M. Adult neurogenesis, human after all (again): Classic, optimized, and future approaches. Behav Brain Res 2020; 381:112458. [DOI: 10.1016/j.bbr.2019.112458] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 11/29/2019] [Accepted: 12/28/2019] [Indexed: 02/08/2023]
|
123
|
Multiple inflammatory profiles of microglia and altered neuroimages in APP/PS1 transgenic AD mice. Brain Res Bull 2020; 156:86-104. [DOI: 10.1016/j.brainresbull.2020.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/14/2019] [Accepted: 01/03/2020] [Indexed: 12/11/2022]
|
124
|
Hour FQ, Moghadam AJ, Shakeri-Zadeh A, Bakhtiyari M, Shabani R, Mehdizadeh M. Magnetic targeted delivery of the SPIONs-labeled mesenchymal stem cells derived from human Wharton's jelly in Alzheimer's rat models. J Control Release 2020; 321:430-441. [PMID: 32097673 DOI: 10.1016/j.jconrel.2020.02.035] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/16/2020] [Accepted: 02/21/2020] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) as a progressive neurodegenerative disorder is one of the leading causes of death globally. Among all treatment approaches, mesenchymal stem cells (MSCs)-based therapy is a promising modality for neurological disorders including the AD. This study aimed to magnetically deliver human Wharton's jelly-derived MSCs (WJ-MSCs) toward the hippocampal area within the AD rat's brain and determine the effects of them in cognitive improvement. Rats were randomly divided into five groups as follow: vehicle-treated control, AD model (injection of 8 μg/kg of amyloid β 1-42), IV-NTC (treated with IV-injected Non-Targeted Cells), IV-TC (treated with IV-injected Targeted Cells), and ICV-NTC (treated with Intracerebroventricular-injected Non-Targeted Cells). WJ-MSCs were labeled with dextran-coated superparamagnetic iron oxide nanoparticles (dex-SPIONs, 50 μg/ml), by bio-mimicry method. SPIONs-labeled MSCs were highly prussian blue positive with an intracellular iron concentration of 2.9 ± 0.08 pg/cell, which were successfully targeted into the hippocampus of AD rats by a halbach magnet array as magnetic targeted cell delivery (MTCD) technique. Presence of SPIONs-labeled cells in hippocampal area was proved by magnetic resonance imaging (MRI) in which signal intensity was reduced by increasing the number of these cells. Behavioral examinations showed that WJ-MSCs caused memory and cognitive improvement. Also, histological assessments showed functional improvement of hippocampal cells by expression of choline acetyltransferase (ChAT) and acetylcholinesterase (AChE). Overall, this study indicates MTCD approach as an alternative in MSC-based regenerative medicine because it approximately has the same results as invasive directly ICV-injection method has.
Collapse
Affiliation(s)
- Farshid Qiyami Hour
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Johari Moghadam
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Shakeri-Zadeh
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Bakhtiyari
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ronak Shabani
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Mehdi Mehdizadeh
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
125
|
Ahmad SAI, Anam MB, Istiaq A, Ito N, Ohta K. Tsukushi is essential for proper maintenance and terminal differentiation of mouse hippocampal neural stem cells. Dev Growth Differ 2020; 62:108-117. [DOI: 10.1111/dgd.12649] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/19/2019] [Accepted: 12/23/2019] [Indexed: 11/30/2022]
Affiliation(s)
- Shah Adil Ishtiyaq Ahmad
- Department of Developmental Neurobiology Graduate School of Life Sciences Kumamoto University Kumamoto Japan
- Stem Cell‐Based Tissue Regeneration Research and Education Unit Kumamoto University Kumamoto Japan
- Department of Biotechnology and Genetic Engineering Mawlana Bhashani Science and Technology University Tangail Bangladesh
| | - Mohammad Badrul Anam
- Department of Developmental Neurobiology Graduate School of Life Sciences Kumamoto University Kumamoto Japan
- Stem Cell‐Based Tissue Regeneration Research and Education Unit Kumamoto University Kumamoto Japan
- HIGO Program Kumamoto University Kumamoto Japan
| | - Arif Istiaq
- Department of Developmental Neurobiology Graduate School of Life Sciences Kumamoto University Kumamoto Japan
- Stem Cell‐Based Tissue Regeneration Research and Education Unit Kumamoto University Kumamoto Japan
- HIGO Program Kumamoto University Kumamoto Japan
| | - Naofumi Ito
- Department of Developmental Neurobiology Graduate School of Life Sciences Kumamoto University Kumamoto Japan
- Stem Cell‐Based Tissue Regeneration Research and Education Unit Kumamoto University Kumamoto Japan
| | - Kunimasa Ohta
- Department of Developmental Neurobiology Graduate School of Life Sciences Kumamoto University Kumamoto Japan
- Stem Cell‐Based Tissue Regeneration Research and Education Unit Kumamoto University Kumamoto Japan
- HIGO Program Kumamoto University Kumamoto Japan
- AMED Core Research for Evolutional Science and Technology (AMED‐CREST) Japan Agency for Medical Research and Development (AMED) Tokyo Japan
| |
Collapse
|
126
|
Neuroinflammation and Neurogenesis in Alzheimer's Disease and Potential Therapeutic Approaches. Int J Mol Sci 2020; 21:ijms21030701. [PMID: 31973106 PMCID: PMC7037892 DOI: 10.3390/ijms21030701] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/17/2020] [Accepted: 01/19/2020] [Indexed: 12/17/2022] Open
Abstract
In adult brain, new neurons are generated throughout adulthood in the subventricular zone and the dentate gyrus; this process is commonly known as adult neurogenesis. The regulation or modulation of adult neurogenesis includes various intrinsic pathways (signal transduction pathway and epigenetic or genetic modulation pathways) or extrinsic pathways (metabolic growth factor modulation, vascular, and immune system pathways). Altered neurogenesis has been identified in Alzheimer's disease (AD), in both human AD brains and AD rodent models. The exact mechanism of the dysregulation of adult neurogenesis in AD has not been completely elucidated. However, neuroinflammation has been demonstrated to alter adult neurogenesis. The presence of various inflammatory components, such as immune cells, cytokines, or chemokines, plays a role in regulating the survival, proliferation, and maturation of neural stem cells. Neuroinflammation has also been considered as a hallmark neuropathological feature of AD. In this review, we summarize current, state-of-the art perspectives on adult neurogenesis, neuroinflammation, and the relationship between these two phenomena in AD. Furthermore, we discuss the potential therapeutic approaches, focusing on the anti-inflammatory and proneurogenic interventions that have been reported in this field.
Collapse
|
127
|
Chen SY, Lin MC, Tsai JS, He PL, Luo WT, Chiu IM, Herschman HR, Li HJ. Exosomal 2',3'-CNP from mesenchymal stem cells promotes hippocampus CA1 neurogenesis/neuritogenesis and contributes to rescue of cognition/learning deficiencies of damaged brain. Stem Cells Transl Med 2020; 9:499-517. [PMID: 31943851 PMCID: PMC7103625 DOI: 10.1002/sctm.19-0174] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 11/25/2019] [Indexed: 12/28/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been used in clinical studies to treat neurological diseases and damage. However, implanted MSCs do not achieve their regenerative effects by differentiating into and replacing neural cells. Instead, MSC secretome components mediate the regenerative effects of MSCs. MSC-derived extracellular vesicles (EVs)/exosomes carry cargo responsible for rescuing brain damage. We previously showed that EP4 antagonist-induced MSC EVs/exosomes have enhanced regenerative potential to rescue hippocampal damage, compared with EVs/exosomes from untreated MSCs. Here we show that EP4 antagonist-induced MSC EVs/exosomes promote neurosphere formation in vitro and increase neurogenesis and neuritogenesis in damaged hippocampi; basal MSC EVs/exosomes do not contribute to these regenerative effects. 2',3'-Cyclic nucleotide 3'-phosphodiesterase (CNP) levels in EP4 antagonist-induced MSC EVs/exosomes are 20-fold higher than CNP levels in basal MSC EVs/exosomes. Decreasing elevated exosomal CNP levels in EP4 antagonist-induced MSC EVs/exosomes reduced the efficacy of these EVs/exosomes in promoting β3-tubulin polymerization and in converting toxic 2',3'-cAMP into neuroprotective adenosine. CNP-depleted EP4 antagonist-induced MSC EVs/exosomes lost the ability to promote neurogenesis and neuritogenesis in damaged hippocampi. Systemic administration of EV/exosomes from EP4 -antagonist derived MSC EVs/exosomes repaired cognition, learning, and memory deficiencies in mice caused by hippocampal damage. In contrast, CNP-depleted EP4 antagonist-induced MSC EVs/exosomes failed to repair this damage. Exosomal CNP contributes to the ability of EP4 antagonist-elicited MSC EVs/exosomes to promote neurogenesis and neuritogenesis in damaged hippocampi and recovery of cognition, memory, and learning. This experimental approach should be generally applicable to identifying the role of EV/exosomal components in eliciting a variety of biological responses.
Collapse
Affiliation(s)
- Shih-Yin Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Meng-Chieh Lin
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Jia-Shiuan Tsai
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Pei-Lin He
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Wen-Ting Luo
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Ing-Ming Chiu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Harvey R Herschman
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California.,Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, California.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California
| | - Hua-Jung Li
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
128
|
Cicvaric A, Sachernegg HM, Stojanovic T, Symmank D, Smani T, Moeslinger T, Uhrin P, Monje FJ. Podoplanin Gene Disruption in Mice Promotes in vivo Neural Progenitor Cells Proliferation, Selectively Impairs Dentate Gyrus Synaptic Depression and Induces Anxiety-Like Behaviors. Front Cell Neurosci 2020; 13:561. [PMID: 32009902 PMCID: PMC6974453 DOI: 10.3389/fncel.2019.00561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/05/2019] [Indexed: 12/20/2022] Open
Abstract
Podoplanin (Pdpn), a brain-tumor-related glycoprotein identified in humans and animals, is endogenously expressed in several organs critical for life support such as kidney, lung, heart and brain. In the brain, Pdpn has been identified in proliferative nestin-positive adult neural progenitor cells and in neurons of the neurogenic hippocampal dentate gyrus (DG), a structure associated to anxiety, critical for learning and memory functions and severely damaged in people with Alzheimer's Disease (AD). The in vivo role of Pdpn in adult neurogenesis and anxiety-like behavior remained however unexplored. Using mice with disrupted Pdpn gene as a model organism and applying combined behavioral, molecular biological and electrophysiological assays, we here show that the absence of Pdpn selectively impairs long-term synaptic depression in the neurogenic DG without affecting the CA3-Schaffer's collateral-CA1 synapses. Pdpn deletion also enhanced the proliferative capacity of DG neural progenitor cells and diminished survival of differentiated neuronal cells in vitro. In addition, mice with podoplanin gene disruption showed increased anxiety-like behaviors in experimentally validated behavioral tests as compared to wild type littermate controls. Together, these findings broaden our knowledge on the molecular mechanisms influencing hippocampal synaptic plasticity and neurogenesis in vivo and reveal Pdpn as a novel molecular target for future studies addressing general anxiety disorder and synaptic depression-related memory dysfunctions.
Collapse
Affiliation(s)
- Ana Cicvaric
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Hannah M. Sachernegg
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Tamara Stojanovic
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Dörte Symmank
- Center for Physiology and Pharmacology, Institute for Physiology, Medical University of Vienna, Vienna, Austria
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville (IBiS)/University of Seville/CIBERCV, Seville, Spain
| | - Thomas Moeslinger
- Center for Physiology and Pharmacology, Institute for Physiology, Medical University of Vienna, Vienna, Austria
| | - Pavel Uhrin
- Center for Physiology and Pharmacology, Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Francisco J. Monje
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
129
|
Kaya I, Jennische E, Dunevall J, Lange S, Ewing AG, Malmberg P, Baykal AT, Fletcher JS. Spatial Lipidomics Reveals Region and Long Chain Base Specific Accumulations of Monosialogangliosides in Amyloid Plaques in Familial Alzheimer's Disease Mice (5xFAD) Brain. ACS Chem Neurosci 2020; 11:14-24. [PMID: 31774647 DOI: 10.1021/acschemneuro.9b00532] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ganglioside metabolism is significantly altered in Alzheimer's disease (AD), which is a progressive neurodegenerative disease prominently characterized by one of its pathological hallmarks, amyloid deposits or "senile plaques". While the plaques mainly consist of aggregated variants of amyloid-β protein (Aβ), recent studies have revealed a number of lipid species including gangliosides in amyloid plaques along with Aβ peptides. It has been widely suggested that long chain (sphingosine) base (LCBs), C18:1-LCB and C20:1-LCB, containing gangliosides might play different roles in neuronal function in vivo. In order to elucidate region-specific aspects of amyloid-plaque associated C18:1-LCB and C20:1-LCB ganglioside accumulations, high spatial resolution (10 μm per pixel) matrix assisted laser desorption ionization imaging mass spectrometry (MALDI-IMS) of gangliosides in amyloid plaques was performed in hippocampal and adjacent cortical regions of 12 month old 5xFAD mouse coronal brain sections from two different stereotaxic coordinates (bregma points, -2.2 and -2.7 mm). MALDI-IMS uncovered brain-region (2 and 3D) and/or LCB specific accumulations of monosialogangliosides (GMs): GM1, GM2, and GM3 in the hippocampal and cortical amyloid plaques. The results reveal monosialogangliosides to be an important component of amyloid plaques and the accumulation of different gangliosides is region and LCB specific in 12 month old 5xFAD mouse brain. This is discussed in relation to amyloid-associated AD pathogenesis such as lipid related immune changes in amyloid plaques, AD specific ganglioside metabolism, and, notably, AD-associated impaired neurogenesis in the subgranular zone.
Collapse
Affiliation(s)
- Ibrahim Kaya
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal 43180, Sweden
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg 405 30, Sweden
- The Gothenburg Imaging Mass Spectrometry (Go:IMS) Platform, University of Gothenburg and Chalmers University of Technology, Gothenburg, Sweden
| | - Eva Jennische
- Institute of Biomedicine, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Johan Dunevall
- Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Stefan Lange
- Institute of Biomedicine, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Andrew G. Ewing
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg 405 30, Sweden
- The Gothenburg Imaging Mass Spectrometry (Go:IMS) Platform, University of Gothenburg and Chalmers University of Technology, Gothenburg, Sweden
| | - Per Malmberg
- Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Ahmet Tarik Baykal
- Department of Medical Biochemistry, Faculty of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - John S. Fletcher
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg 405 30, Sweden
- The Gothenburg Imaging Mass Spectrometry (Go:IMS) Platform, University of Gothenburg and Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
130
|
Khacho M, Harris R, Slack RS. Mitochondria as central regulators of neural stem cell fate and cognitive function. Nat Rev Neurosci 2019; 20:34-48. [PMID: 30464208 DOI: 10.1038/s41583-018-0091-3] [Citation(s) in RCA: 229] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Emerging evidence now indicates that mitochondria are central regulators of neural stem cell (NSC) fate decisions and are crucial for both neurodevelopment and adult neurogenesis, which in turn contribute to cognitive processes in the mature brain. Inherited mutations and accumulated damage to mitochondria over the course of ageing serve as key factors underlying cognitive defects in neurodevelopmental disorders and neurodegenerative diseases, respectively. In this Review, we explore the recent findings that implicate mitochondria as crucial regulators of NSC function and cognition. In this respect, mitochondria may serve as targets for stem-cell-based therapies and interventions for cognitive defects.
Collapse
Affiliation(s)
- Mireille Khacho
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology (OISB), University of Ottawa, Ottawa, Ontario, Canada
| | - Richard Harris
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario, Canada
| | - Ruth S Slack
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
131
|
Nakafuku M, Del Águila Á. Developmental dynamics of neurogenesis and gliogenesis in the postnatal mammalian brain in health and disease: Historical and future perspectives. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 9:e369. [PMID: 31825170 DOI: 10.1002/wdev.369] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 09/16/2019] [Accepted: 10/22/2019] [Indexed: 12/21/2022]
Abstract
The mature mammalian brain has long been thought to be a structurally rigid, static organ since the era of Ramón y Cajal in the early 20th century. Evidence accumulated over the past three decades, however, has completely overturned this long-held view. We now know that new neurons and glia are continuously added to the brain at postnatal stages, even in mature adults of various mammalian species, including humans. Moreover, these newly added cells contribute to structural plasticity and play important roles in higher order brain function, as well as repair after damage. A major source of these new neurons and glia is neural stem cells (NSCs) that persist in specialized niches in the brain throughout life. With this new view, our understanding of normal brain physiology and interventional approaches to various brain disorders has changed markedly in recent years. This article provides a brief overview on the historical changes in our understanding of the developmental dynamics of neurogenesis and gliogenesis in the postnatal and adult mammalian brain and discusses the roles of NSCs and other progenitor populations in such cellular dynamics in health and disease of the postnatal mammalian brain. This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cell Differentiation and Reversion Adult Stem Cells, Tissue Renewal, and Regeneration > Tissue Stem Cells and Niches Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cells and Disease.
Collapse
Affiliation(s)
- Masato Nakafuku
- Divisions of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ángela Del Águila
- Divisions of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
132
|
Hagihara H, Murano T, Ohira K, Miwa M, Nakamura K, Miyakawa T. Expression of progenitor cell/immature neuron markers does not present definitive evidence for adult neurogenesis. Mol Brain 2019; 12:108. [PMID: 31823803 PMCID: PMC6902531 DOI: 10.1186/s13041-019-0522-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/13/2019] [Indexed: 12/14/2022] Open
Abstract
It is agreed upon that adult hippocampal neurogenesis (AHN) occurs in the dentate gyrus (DG) in rodents. However, the existence of AHN in humans, particularly in elderly individuals, remains to be determined. Recently, several studies reported that neural progenitor cells, neuroblasts, and immature neurons were detected in the hippocampus of elderly humans, based on the expressions of putative markers for these cells, claiming that this provides evidence of the persistence of AHN in humans. Herein, we briefly overview the phenomenon that we call "dematuration," in which mature neurons dedifferentiate to a pseudo-immature status and re-express the molecular markers of neural progenitor cells and immature neurons. Various conditions can easily induce dematuration, such as inflammation and hyper-excitation of neurons, and therefore, the markers for neural progenitor cells and immature neurons may not necessarily serve as markers for AHN. Thus, the aforementioned studies have not presented definitive evidence for the persistence of hippocampal neurogenesis throughout adult life in humans, and we would like to emphasize that those markers should be used cautiously when presented as evidence for AHN. Increasing AHN has been considered as a therapeutic target for Alzheimer's disease (AD); however, given that immature neuronal markers can be re-expressed in mature adult neurons, independent of AHN, in various disease conditions including AD, strategies to increase the expression of these markers in the DG may be ineffective or may worsen the symptoms of such diseases.
Collapse
Affiliation(s)
- Hideo Hagihara
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Tomoyuki Murano
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Koji Ohira
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan.,Laboratory of Nutritional Brain Science, Department of Food Science and Nutrition, Mukogawa Women's University, Nishinomiya, Hyogo, 663-8558, Japan
| | - Miki Miwa
- Cognitive Neuroscience Section, Primate Research Institute, Kyoto University, Inuyama, Aichi, 484-8506, Japan
| | - Katsuki Nakamura
- Cognitive Neuroscience Section, Primate Research Institute, Kyoto University, Inuyama, Aichi, 484-8506, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan.
| |
Collapse
|
133
|
The puzzle of preserved cognition in the oldest old. Neurol Sci 2019; 41:441-447. [PMID: 31713754 DOI: 10.1007/s10072-019-04111-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 10/15/2019] [Indexed: 02/07/2023]
Abstract
Although epidemiological studies predict an exponential increase in the prevalence of dementia with age, recent studies have demonstrated that the oldest old are actually less frequently affected by dementia than the younger elderly. To explain this, I suggest a parallel between brain ageing and Alzheimer's disease (AD) and assume that theories concerning the brain's vulnerability to AD and its individual variability may also explain why some of the oldest old remain cognitively efficient. Some theories argue that AD is due to the continuing presence of the immature neurones vulnerable to amyloid beta protein (Aß) that are normally involved in brain development and then removed as a result of cell selection by the proteins associated with both brain development and AD. If a dysfunction in cell selection allows these immature neurones to survive, they degenerate early as a result of the neurotoxic action of Aß accumulation, which their mature counterparts can withstand. Consequently, age at the time of onset of AD and its clinical presentations depend on the number and location of such immature cells. I speculate that the same mechanism is responsible for the variability of normal brain ageing: the oldest old with well-preserved cognitive function are people genetically programmed for extreme ageing who have benefited from better cell selection during prenatal and neonatal life and therefore have fewer surviving neurones vulnerable to amyloid-promoted degeneration, whereas the process of early life cell selection was less successful in the oldest old who develop dementia.
Collapse
|
134
|
von Rüden EL, Zellinger C, Gedon J, Walker A, Bierling V, Deeg CA, Hauck SM, Potschka H. Regulation of Alzheimer's disease-associated proteins during epileptogenesis. Neuroscience 2019; 424:102-120. [PMID: 31705965 DOI: 10.1016/j.neuroscience.2019.08.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 07/26/2019] [Accepted: 08/20/2019] [Indexed: 12/12/2022]
Abstract
Clinical evidence and pathological studies suggest a bidirectional link between temporal lobe epilepsy and Alzheimer's disease (AD). Data analysis from omic studies offers an excellent opportunity to identify the overlap in molecular alterations between the two pathologies. We have subjected proteomic data sets from a rat model of epileptogenesis to a bioinformatics analysis focused on proteins functionally linked with AD. The data sets have been obtained for hippocampus (HC) and parahippocampal cortex samples collected during the course of epileptogenesis. Our study confirmed a relevant dysregulation of proteins linked with Alzheimer pathogenesis. When comparing the two brain areas, a more prominent regulation was evident in parahippocampal cortex samples as compared to the HC. Dysregulated protein groups comprised those affecting mitochondrial function and calcium homeostasis. Differentially expressed mitochondrial proteins included proteins of the mitochondrial complexes I, III, IV, and V as well as of the accessory subunit of complex I. The analysis also revealed a regulation of the microtubule associated protein Tau in parahippocampal cortex tissue during the latency phase. This was further confirmed by immunohistochemistry. Moreover, we demonstrated a complex epileptogenesis-associated dysregulation of proteins involved in amyloid β processing and its regulation. Among others, the amyloid precursor protein and the α-secretase alpha disintegrin metalloproteinase 17 were included. Our analysis revealed a relevant regulation of key proteins known to be associated with AD pathogenesis. The analysis provides a comprehensive overview of shared molecular alterations characterizing epilepsy development and manifestation as well as AD development and progression.
Collapse
Affiliation(s)
- Eva-Lotta von Rüden
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Christina Zellinger
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Julia Gedon
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Andreas Walker
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Vera Bierling
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Cornelia A Deeg
- Institute of Animal Physiology, Department of Veterinary Sciences, Ludwig-Maximilians-University (LMU), Munich, Germany; Experimental Ophthalmology, Philipps University of Marburg, Marburg, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Center Munich, Neuherberg, Germany
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University (LMU), Munich, Germany.
| |
Collapse
|
135
|
Jeon SG, Hong SB, Nam Y, Tae J, Yoo A, Song EJ, Kim KI, Lee D, Park J, Lee SM, Kim JI, Moon M. Ghrelin in Alzheimer's disease: Pathologic roles and therapeutic implications. Ageing Res Rev 2019; 55:100945. [PMID: 31434007 DOI: 10.1016/j.arr.2019.100945] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/25/2019] [Accepted: 08/16/2019] [Indexed: 12/11/2022]
Abstract
Ghrelin, which has many important physiological roles, such as stimulating food intake, regulating energy homeostasis, and releasing insulin, has recently been studied for its roles in a diverse range of neurological disorders. Despite the several functions of ghrelin in the central nervous system, whether it works as a therapeutic agent for neurological dysfunction has been unclear. Altered levels and various roles of ghrelin have been reported in Alzheimer's disease (AD), which is characterized by the accumulation of misfolded proteins resulting in synaptic loss and cognitive decline. Interestingly, treatment with ghrelin or with the agonist of ghrelin receptor showed attenuation in several cases of AD-related pathology. These findings suggest the potential therapeutic implications of ghrelin in the pathogenesis of AD. In the present review, we summarized the roles of ghrelin in AD pathogenesis, amyloid beta (Aβ) homeostasis, tau hyperphosphorylation, neuroinflammation, mitochondrial deficit, synaptic dysfunction and cognitive impairment. The findings from this review suggest that ghrelin has a novel therapeutic potential for AD treatment. Thus, rigorously designed studies are needed to establish an effective AD-modifying strategy.
Collapse
|
136
|
Cipriani S, Ferrer I, Aronica E, Kovacs GG, Verney C, Nardelli J, Khung S, Delezoide AL, Milenkovic I, Rasika S, Manivet P, Benifla JL, Deriot N, Gressens P, Adle-Biassette H. Hippocampal Radial Glial Subtypes and Their Neurogenic Potential in Human Fetuses and Healthy and Alzheimer's Disease Adults. Cereb Cortex 2019; 28:2458-2478. [PMID: 29722804 DOI: 10.1093/cercor/bhy096] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Indexed: 02/06/2023] Open
Abstract
Neuropathological conditions might affect adult granulogenesis in the adult human dentate gyrus. However, radial glial cells (RGCs) have not been well characterized during human development and aging. We have previously described progenitor and neuronal layer establishment in the hippocampal pyramidal layer and dentate gyrus from embryonic life until mid-gestation. Here, we describe RGC subtypes in the hippocampus from 13 gestational weeks (GW) to mid-gestation and characterize their evolution and the dynamics of neurogenesis from mid-gestation to adulthood in normal and Alzheimer's disease (AD) subjects. In the pyramidal ventricular zone (VZ), RGC density declined with neurogenesis from mid-gestation until the perinatal period. In the dentate area, morphologic and antigenic differences among RGCs were observed from early ages of development to adulthood. Density and proliferative capacity of dentate RGCs as well as neurogenesis were strongly reduced during childhood until 5 years, few DCX+ cells are seen in adults. The dentate gyrus of both control and AD individuals showed Nestin+ and/or GFAPδ+ cells displaying different morphologies. In conclusion, pools of morphologically, antigenically, and topographically diverse neural progenitor cells are present in the human hippocampus from early developmental stages until adulthood, including in AD patients, while their neurogenic potential seems negligible in the adult.
Collapse
Affiliation(s)
- Sara Cipriani
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Isidre Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Bellvitge Campus, L'Hospitalet de Llobregat, Spain; Centre for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), Institute Carlos III, Madrid, Spain
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Gabor G Kovacs
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Catherine Verney
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Jeannette Nardelli
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Suonavy Khung
- APHP, Service de Biologie du Développement, Hôpital Robert-Debré, APHP, Paris, France
| | - Anne-Lise Delezoide
- APHP, Service de Biologie du Développement, Hôpital Robert-Debré, APHP, Paris, France
| | - Ivan Milenkovic
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | | | - Philippe Manivet
- APHP, Plateforme de Bio-Pathologie et de Technologies Innovantes en Santé, Centre de Ressources Biologiques BB-0033-00064, Hôpital Lariboisière, Paris, France
| | - Jean-Louis Benifla
- APHP, Service de Gynécologie-Obstétrique, Hôpital Lariboisère, Paris, France
| | - Nicolas Deriot
- APHP, Plateforme de Bio-Pathologie et de Technologies Innovantes en Santé, Centre de Ressources Biologiques BB-0033-00064, Hôpital Lariboisière, Paris, France.,Service d'Anatomie et de Cytologie Pathologiques, Hôpital Lariboisère, Paris, France
| | - Pierre Gressens
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Department of Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
| | - Homa Adle-Biassette
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,APHP, Plateforme de Bio-Pathologie et de Technologies Innovantes en Santé, Centre de Ressources Biologiques BB-0033-00064, Hôpital Lariboisière, Paris, France.,Service d'Anatomie et de Cytologie Pathologiques, Hôpital Lariboisère, Paris, France
| |
Collapse
|
137
|
Hwang SJ, Tao Z, Kim WH, Singh V. Conditional Recurrent Flow: Conditional Generation of Longitudinal Samples with Applications to Neuroimaging. PROCEEDINGS. IEEE INTERNATIONAL CONFERENCE ON COMPUTER VISION 2019; 2019:10691-10700. [PMID: 32405276 PMCID: PMC7220239 DOI: 10.1109/iccv.2019.01079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
We develop a conditional generative model for longitudinal image datasets based on sequential invertible neural networks. Longitudinal image acquisitions are common in various scientific and biomedical studies where often each image sequence sample may also come together with various secondary (fixed or temporally dependent) measurements. The key goal is not only to estimate the parameters of a deep generative model for the given longitudinal data, but also to enable evaluation of how the temporal course of the generated longitudinal samples are influenced as a function of induced changes in the (secondary) temporal measurements (or events). Our proposed formulation incorporates recurrent subnetworks and temporal context gating, which provide a smooth transition in a temporal sequence of generated data that can be easily informed or modulated by secondary temporal conditioning variables. We show that the formulation works well despite the smaller sample sizes common in these applications. Our model is validated on two video datasets and a longitudinal Alzheimer's disease (AD) dataset for both quantitative and qualitative evaluations of the generated samples. Further, using our generated longitudinal image samples, we show that we can capture the pathological progressions in the brain that turn out to be consistent with the existing literature, and could facilitate various types of downstream statistical analysis.
Collapse
|
138
|
Chavoshinezhad S, Mohseni Kouchesfahani H, Ahmadiani A, Dargahi L. Interferon beta ameliorates cognitive dysfunction in a rat model of Alzheimer's disease: Modulation of hippocampal neurogenesis and apoptosis as underlying mechanism. Prog Neuropsychopharmacol Biol Psychiatry 2019; 94:109661. [PMID: 31152860 DOI: 10.1016/j.pnpbp.2019.109661] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 05/08/2019] [Accepted: 05/28/2019] [Indexed: 12/21/2022]
Abstract
Neuronal apoptosis and impaired hippocampal neurogenesis are major players in cognitive/memory dysfunctions including Alzheimer's disease (AD). Interferon beta (IFNβ) is a cytokine with anti-apoptotic and neuroprotective properties on the central nervous system (CNS) cells which specifically affects neural progenitor cells (NPCs) even in the adult brain. In this study, we examined the effect of IFNβ on memory impairment as well as hippocampal neurogenesis and apoptosis in a rat model of AD. AD model was induced by lentiviral-mediated overexpression of mutant APP in the hippocampus of adult rats. Intranasal (IN) administration of IFNβ (0.5 μg/kg and 1 μg/kg doses) was started from day 23 after virus injection and continued every other day to the final day of experiments. The expression levels of APP, neurogenesis (Nestin, Ki67, DCX, and Reelin) and apoptosis (Bax/Bcl-2 ratio, cleaved-caspase-3 and seladin-1) markers were evaluated by immunohistochemistry, real-time PCR, immunofluorescence and western blotting. Moreover, thioflavin T and Nissl stainings were used to assess Aβ plaque levels and neuronal degeneration in the hippocampus, respectively. Our results showed that IFNβ treatment reduced APP expression and Aβ plaque formation, and concomitantly ameliorated spatial learning and memory deficits examined in Y-maze and Morris water maze tests. Moreover, in parallel with reducing apoptosis and neural loss in the hippocampal subfields, IFNβ decreased ectopic neurogenesis in the CA1 and CA3 regions of the AD rat hippocampus. However, IFNβ increased neurogenesis in the dentate gyrus neurogenic niche. Our findings suggest that IFNβ exerts neuroprotective effects at least partly by inhibition of apoptosis and modulation of neurogenesis. Taken together, IFNβ can be a promising therapeutic approach to improve cognitive performance in AD-like neurodegenerative context.
Collapse
Affiliation(s)
- Sara Chavoshinezhad
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | | | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
139
|
Boldrini M, Fulmore CA, Tartt AN, Simeon LR, Pavlova I, Poposka V, Rosoklija GB, Stankov A, Arango V, Dwork AJ, Hen R, Mann JJ. Human Hippocampal Neurogenesis Persists throughout Aging. Cell Stem Cell 2019; 22:589-599.e5. [PMID: 29625071 PMCID: PMC5957089 DOI: 10.1016/j.stem.2018.03.015] [Citation(s) in RCA: 852] [Impact Index Per Article: 170.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/24/2017] [Accepted: 03/19/2018] [Indexed: 12/12/2022]
Abstract
Adult hippocampal neurogenesis declines in aging rodents and primates. Aging humans are thought to exhibit waning neurogenesis and exercise-induced angiogenesis, with a resulting volumetric decrease in the neurogenic hippocampal dentate gyrus (DG) region, although concurrent changes in these parameters are not well studied. Here we assessed whole autopsy hippocampi from healthy human individuals ranging from 14 to 79 years of age. We found similar numbers of intermediate neural progenitors and thousands of immature neurons in the DG, comparable numbers of glia and mature granule neurons, and equivalent DG volume across ages. Nevertheless, older individuals have less angiogenesis and neuroplasticity and a smaller quiescent progenitor pool in anterior-mid DG, with no changes in posterior DG. Thus, healthy older subjects without cognitive impairment, neuropsychiatric disease, or treatment display preserved neurogenesis. It is possible that ongoing hippocampal neurogenesis sustains human-specific cognitive function throughout life and that declines may be linked to compromised cognitive-emotional resilience.
Collapse
Affiliation(s)
- Maura Boldrini
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Molecular Imaging and Neuropathology, NYS Psychiatric Institute, New York, NY 10032, USA.
| | - Camille A Fulmore
- Division of Molecular Imaging and Neuropathology, NYS Psychiatric Institute, New York, NY 10032, USA
| | - Alexandria N Tartt
- Division of Molecular Imaging and Neuropathology, NYS Psychiatric Institute, New York, NY 10032, USA
| | - Laika R Simeon
- Division of Molecular Imaging and Neuropathology, NYS Psychiatric Institute, New York, NY 10032, USA
| | - Ina Pavlova
- Division of Integrative Neuroscience, NYS Psychiatric Institute, New York, NY 10032, USA
| | - Verica Poposka
- Institute for Forensic Medicine, Ss. Cyril & Methodius University, Skopje 1000, Republic of Macedonia
| | - Gorazd B Rosoklija
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Molecular Imaging and Neuropathology, NYS Psychiatric Institute, New York, NY 10032, USA; Macedonian Academy of Sciences & Arts, 2, Ss. Cyril & Methodius University, Skopje 1000, Republic of Macedonia
| | - Aleksandar Stankov
- Institute for Forensic Medicine, Ss. Cyril & Methodius University, Skopje 1000, Republic of Macedonia
| | - Victoria Arango
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Molecular Imaging and Neuropathology, NYS Psychiatric Institute, New York, NY 10032, USA
| | - Andrew J Dwork
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA; Division of Molecular Imaging and Neuropathology, NYS Psychiatric Institute, New York, NY 10032, USA; Macedonian Academy of Sciences & Arts, 2, Ss. Cyril & Methodius University, Skopje 1000, Republic of Macedonia
| | - René Hen
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Department of Neuroscience, Columbia University, New York, NY 10032, USA; Department of Pharmacology, Columbia University, New York, NY 10032, USA; Division of Integrative Neuroscience, NYS Psychiatric Institute, New York, NY 10032, USA
| | - J John Mann
- Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Molecular Imaging and Neuropathology, NYS Psychiatric Institute, New York, NY 10032, USA
| |
Collapse
|
140
|
Kandasamy M, Radhakrishnan RK, Poornimai Abirami GP, Roshan SA, Yesudhas A, Balamuthu K, Prahalathan C, Shanmugaapriya S, Moorthy A, Essa MM, Anusuyadevi M. Possible Existence of the Hypothalamic-Pituitary-Hippocampal (HPH) Axis: A Reciprocal Relationship Between Hippocampal Specific Neuroestradiol Synthesis and Neuroblastosis in Ageing Brains with Special Reference to Menopause and Neurocognitive Disorders. Neurochem Res 2019; 44:1781-1795. [PMID: 31254250 DOI: 10.1007/s11064-019-02833-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/13/2019] [Accepted: 06/19/2019] [Indexed: 12/18/2022]
|
141
|
Liu Z, Wu P, Yin Y, Tian Y. A ratiometric fluorescent DNA nanoprobe for cerebral adenosine triphosphate assay. Chem Commun (Camb) 2019; 55:9955-9958. [PMID: 31364619 DOI: 10.1039/c9cc05046a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A silver nanocluster-based ratiometric fluorescent nanosensor was developed for the determination of ATP in the cerebrospinal fluid of a mouse brain. Using this useful tool with good stability and high selectivity as well as a wide linear detection range, it was found that the ATP concentration in a mouse brain with Alzheimer's disease was 2300-fold higher than that in a normal one.
Collapse
Affiliation(s)
- Zhichao Liu
- Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P. R. China.
| | - Peicong Wu
- Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P. R. China.
| | - Yaoyao Yin
- Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P. R. China.
| | - Yang Tian
- Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, P. R. China.
| |
Collapse
|
142
|
Myhre CL, Thygesen C, Villadsen B, Vollerup J, Ilkjær L, Krohn KT, Grebing M, Zhao S, Khan AM, Dissing-Olesen L, Jensen MS, Babcock AA, Finsen B. Microglia Express Insulin-Like Growth Factor-1 in the Hippocampus of Aged APP swe/PS1 ΔE9 Transgenic Mice. Front Cell Neurosci 2019; 13:308. [PMID: 31417357 PMCID: PMC6682662 DOI: 10.3389/fncel.2019.00308] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 06/24/2019] [Indexed: 11/14/2022] Open
Abstract
Insulin-like growth factor-1 (IGF-1) is a pleiotropic molecule with neurotrophic and immunomodulatory functions. Knowing the capacity of chronically activated microglia to produce IGF-1 may therefore show essential to promote beneficial microglial functions in Alzheimer's disease (AD). Here, we investigated the expression of IGF-1 mRNA and IGF-1 along with the expression of tumor necrosis factor (TNF) mRNA, and the amyloid-β (Aβ) plaque load in the hippocampus of 3- to 24-month-old APPswe/PS1ΔE9 transgenic (Tg) and wild-type (WT) mice. As IGF-1, in particular, is implicated in neurogenesis we also monitored the proliferation of cells in the subgranular zone (sgz) of the dentate gyrus. We found that the Aβ plaque load reached its maximum in aged 21- and 24-month-old APPswe/PS1ΔE9 Tg mice, and that microglial reactivity and hippocampal IGF-1 and TNF mRNA levels were significantly elevated in aged APPswe/PS1ΔE9 Tg mice. The sgz cell proliferation decreased with age, regardless of genotype and increased IGF-1/TNF mRNA levels. Interestingly, IGF-1 mRNA was expressed in subsets of sgz cells, likely neuroblasts, and neurons in both genotypes, regardless of age, as well as in glial-like cells. By double in situ hybridization these were shown to be IGF1 mRNA+ CD11b mRNA+ cells, i.e., IGF-1 mRNA-expressing microglia. Quantification showed a 2-fold increase in the number of microglia and IGF-1 mRNA-expressing microglia in the molecular layer of the dentate gyrus in aged APPswe/PS1ΔE9 Tg mice. Double-immunofluorescence showed that IGF-1 was expressed in a subset of Aβ plaque-associated CD11b+ microglia and in several subsets of neurons. Exposure of primary murine microglia and BV2 cells to Aβ42 did not affect IGF-1 mRNA expression. IGF-1 mRNA levels remained constant in WT mice with aging, unlike TNF mRNA levels which increased with aging. In conclusion, our results suggest that the increased IGF-1 mRNA levels can be ascribed to a larger number of IGF-1 mRNA-expressing microglia in the aged APPswe/PS1ΔE9 Tg mice. The finding that subsets of microglia retain the capacity to express IGF-1 mRNA and IGF-1 in the aged APPswe/PS1ΔE9 Tg mice is encouraging, considering the beneficial therapeutic potential of modulating microglial production of IGF-1 in AD.
Collapse
Affiliation(s)
- Christa Løth Myhre
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Camilla Thygesen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Brain Research – Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Birgitte Villadsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Brain Research – Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jeanette Vollerup
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Brain Research – Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Laura Ilkjær
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Katrine Tækker Krohn
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Brain Research – Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Manuela Grebing
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Shuainan Zhao
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Brain Research – Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Asif Manzoor Khan
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Lasse Dissing-Olesen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Alicia A. Babcock
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Bente Finsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Brain Research – Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
143
|
Song J. Pineal gland dysfunction in Alzheimer's disease: relationship with the immune-pineal axis, sleep disturbance, and neurogenesis. Mol Neurodegener 2019; 14:28. [PMID: 31296240 PMCID: PMC6624939 DOI: 10.1186/s13024-019-0330-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/01/2019] [Indexed: 12/15/2022] Open
Abstract
Alzheimer’s disease (AD) is a globally common neurodegenerative disease, which is accompanied by alterations to various lifestyle patterns, such as sleep disturbance. The pineal gland is the primary endocrine organ that secretes hormones, such as melatonin, and controls the circadian rhythms. The decrease in pineal gland volume and pineal calcification leads to the reduction of melatonin production. Melatonin has been reported to have multiple roles in the central nervous system (CNS), including improving neurogenesis and synaptic plasticity, suppressing neuroinflammation, enhancing memory function, and protecting against oxidative stress. Recently, reduced pineal gland volume and pineal calcification, accompanied by cognitive decline and sleep disturbances have been observed in AD patients. Here, I review current significant evidence of the contribution of pineal dysfunction in AD to the progress of AD neuropathology. I suggest new insights to understanding the relationship between AD pathogenesis and pineal gland function.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun, 58128, Jeollanam-do, Republic of Korea.
| |
Collapse
|
144
|
Interleukine-17 Administration Modulates Adult Hippocampal Neurogenesis and Improves Spatial Learning in Mice. J Mol Neurosci 2019; 69:254-263. [PMID: 31254254 DOI: 10.1007/s12031-019-01354-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/29/2019] [Indexed: 01/17/2023]
Abstract
Adult hippocampal neurogenesis plays an important role in health and disease. Regulating neurogenesis may be a key mechanism in the pathophysiology and treatment of several neurobehavioral disorders such as schizophrenia, depression, autism spectrum disorders and Alzheimer's disease. Cytokines are known to affect adult neurogenesis, but conflicting studies have been reported with regard to their actual role. Interleukine-17 (IL-17), a potent pro-inflammatory cytokine, has been shown to inhibit proliferation of neuroprogenitors and thus reduce hippocampal neurogenesis, while other studies suggested it can promote neurite outgrowth. In the present study we sought to explore the possible effect of a single dose administration of IL-17 on neurogenesis related behavior, i.e. spatial learning. Surprisingly, ICR mice injected with IL-17 (8 μg) had a significant slight improvement in spatial learning in the Morris water maze paradigm, without any changes in general locomotion compared with control mice. Indeed, the expression of neurogenesis related genes was down regulated following IL-17 treatment. However, we detected an upregulation in the expression of FGF-13, a gene promoting microtubule polymerization and neurite outgrowth, thus supporting neuronal maturation. We thus suggest that IL-17 has a complex role in regulating adult neurogenesis: inhibiting neuroprogenitors proliferation on one hand, while promoting maturation of already formed neuroblasts on the other hand. Our findings suggest that these roles can potentially affect neurogenesis related behavior. Its actual role in health and disease is yet to be determined.
Collapse
|
145
|
Fu CH, Iascone DM, Petrof I, Hazra A, Zhang X, Pyfer MS, Tosi U, Corbett BF, Cai J, Lee J, Park J, Iacovitti L, Scharfman HE, Enikolopov G, Chin J. Early Seizure Activity Accelerates Depletion of Hippocampal Neural Stem Cells and Impairs Spatial Discrimination in an Alzheimer's Disease Model. Cell Rep 2019; 27:3741-3751.e4. [PMID: 31242408 PMCID: PMC6697001 DOI: 10.1016/j.celrep.2019.05.101] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 04/24/2019] [Accepted: 05/28/2019] [Indexed: 12/21/2022] Open
Abstract
Adult hippocampal neurogenesis has been reported to be decreased, increased, or not changed in Alzheimer's disease (AD) patients and related transgenic mouse models. These disparate findings may relate to differences in disease stage, or the presence of seizures, which are associated with AD and can stimulate neurogenesis. In this study, we investigate a transgenic mouse model of AD that exhibits seizures similarly to AD patients and find that neurogenesis is increased in early stages of disease, as spontaneous seizures became evident, but is decreased below control levels as seizures recur. Treatment with the antiseizure drug levetiracetam restores neurogenesis and improves performance in a neurogenesis-associated spatial discrimination task. Our results suggest that seizures stimulate, and later accelerate the depletion of, the hippocampal neural stem cell pool. These results have implications for AD as well as any disorder accompanied by recurrent seizures, such as epilepsy.
Collapse
Affiliation(s)
- Chia-Hsuan Fu
- Memory & Brain Research Center, Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Daniel Maxim Iascone
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Iraklis Petrof
- Memory & Brain Research Center, Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Anupam Hazra
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Xiaohong Zhang
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Mark S Pyfer
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Umberto Tosi
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Brian F Corbett
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jingli Cai
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jason Lee
- Memory & Brain Research Center, Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jin Park
- Memory & Brain Research Center, Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lorraine Iacovitti
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Helen E Scharfman
- Departments of Psychiatry, Neuroscience, and Physiology and the Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Grigori Enikolopov
- Center for Developmental Genetics and Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jeannie Chin
- Memory & Brain Research Center, Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
146
|
Shin SJ, Jeon SG, Kim JI, Jeong YO, Kim S, Park YH, Lee SK, Park HH, Hong SB, Oh S, Hwang JY, Kim HS, Park H, Nam Y, Lee YY, Kim JJ, Park SH, Kim JS, Moon M. Red Ginseng Attenuates Aβ-Induced Mitochondrial Dysfunction and Aβ-mediated Pathology in an Animal Model of Alzheimer's Disease. Int J Mol Sci 2019; 20:E3030. [PMID: 31234321 PMCID: PMC6627470 DOI: 10.3390/ijms20123030] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/05/2019] [Accepted: 06/19/2019] [Indexed: 12/03/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease and is characterized by neurodegeneration and cognitive deficits. Amyloid beta (Aβ) peptide is known to be a major cause of AD pathogenesis. However, recent studies have clarified that mitochondrial deficiency is also a mediator or trigger for AD development. Interestingly, red ginseng (RG) has been demonstrated to have beneficial effects on AD pathology. However, there is no evidence showing whether RG extract (RGE) can inhibit the mitochondrial deficit-mediated pathology in the experimental models of AD. The effects of RGE on Aβ-mediated mitochondrial deficiency were investigated in both HT22 mouse hippocampal neuronal cells and the brains of 5XFAD Aβ-overexpressing transgenic mice. To examine whether RGE can affect mitochondria-related pathology, we used immunohistostaining to study the effects of RGE on Aβ accumulation, neuroinflammation, neurodegeneration, and impaired adult hippocampal neurogenesis in hippocampal formation of 5XFAD mice. In vitro and in vivo findings indicated that RGE significantly improves Aβ-induced mitochondrial pathology. In addition, RGE significantly ameliorated AD-related pathology, such as Aβ deposition, gliosis, and neuronal loss, and deficits in adult hippocampal neurogenesis in brains with AD. Our results suggest that RGE may be a mitochondria-targeting agent for the treatment of AD.
Collapse
Affiliation(s)
- Soo Jung Shin
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Seong Gak Jeon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Jin-Il Kim
- Department of Nursing, College of Nursing, Jeju National University, Jeju-si 63243, Korea.
| | - Yu-On Jeong
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Yong Ho Park
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Seong-Kyung Lee
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Hyun Ha Park
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Sang Bum Hong
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Sua Oh
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Ji-Young Hwang
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Hyeon Soo Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - HyunHee Park
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Yong Yook Lee
- The Korean Ginseng Research Institute, Korea Ginseng Corporation, Gajeong-ro, Shinseong-dong, Yuseong-gu, Daejeon 34128, Korea.
| | - Jwa-Jin Kim
- Department of Nephrology, School of Medicine, Chungnam National University, Daejeon 35015, Korea.
| | - Sun-Hyun Park
- R&D center for Advanced Pharmaceuticals & Evaluation, Korea Institute of toxicology, 141, Gajeong-ro, Yuseong-gu, Daejeon 34114, Korea.
| | - Jong-Seok Kim
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| |
Collapse
|
147
|
Altuna M, Urdánoz-Casado A, Sánchez-Ruiz de Gordoa J, Zelaya MV, Labarga A, Lepesant JMJ, Roldán M, Blanco-Luquin I, Perdones Á, Larumbe R, Jericó I, Echavarri C, Méndez-López I, Di Stefano L, Mendioroz M. DNA methylation signature of human hippocampus in Alzheimer's disease is linked to neurogenesis. Clin Epigenetics 2019; 11:91. [PMID: 31217032 PMCID: PMC6585076 DOI: 10.1186/s13148-019-0672-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/25/2019] [Indexed: 12/22/2022] Open
Abstract
Background Drawing the epigenome landscape of Alzheimer’s disease (AD) still remains a challenge. To characterize the epigenetic molecular basis of the human hippocampus in AD, we profiled genome-wide DNA methylation levels in hippocampal samples from a cohort of pure AD patients and controls by using the Illumina 450K methylation arrays. Results Up to 118 AD-related differentially methylated positions (DMPs) were identified in the AD hippocampus, and extended mapping of specific regions was obtained by bisulfite cloning sequencing. AD-related DMPs were significantly correlated with phosphorylated tau burden. Functional analysis highlighted that AD-related DMPs were enriched in poised promoters that were not generally maintained in committed neural progenitor cells, as shown by ChiP-qPCR experiments. Interestingly, AD-related DMPs preferentially involved neurodevelopmental and neurogenesis-related genes. Finally, InterPro ontology analysis revealed enrichment in homeobox-containing transcription factors in the set of AD-related DMPs. Conclusions These results suggest that altered DNA methylation in the AD hippocampus occurs at specific regulatory regions crucial for neural differentiation supporting the notion that adult hippocampal neurogenesis may play a role in AD through epigenetic mechanisms. Graphical abstract ![]()
Electronic supplementary material The online version of this article (10.1186/s13148-019-0672-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Miren Altuna
- Neuroepigenetics Laboratory, Navarrabiomed, Public University of Navarre (UPNA), IdiSNA (Navarra Institute for Health Research), c/ Irunlarrea, 3, 31008, Pamplona, Spain.,Department of Neurology, Complejo Hospitalario de Navarra, IdiSNA (Navarra Institute for Health Research), Pamplona, Spain
| | - Amaya Urdánoz-Casado
- Neuroepigenetics Laboratory, Navarrabiomed, Public University of Navarre (UPNA), IdiSNA (Navarra Institute for Health Research), c/ Irunlarrea, 3, 31008, Pamplona, Spain
| | - Javier Sánchez-Ruiz de Gordoa
- Neuroepigenetics Laboratory, Navarrabiomed, Public University of Navarre (UPNA), IdiSNA (Navarra Institute for Health Research), c/ Irunlarrea, 3, 31008, Pamplona, Spain.,Department of Neurology, Complejo Hospitalario de Navarra, IdiSNA (Navarra Institute for Health Research), Pamplona, Spain
| | - María V Zelaya
- Department of Pathology, Complejo Hospitalario de Navarra- IdiSNA (Navarra Institute for Health Research), Pamplona, Spain
| | - Alberto Labarga
- Bioinformatics Unit, Navarrabiomed, Public University of Navarre (UPNA), IdiSNA (Navarra Institute for Health Research), Pamplona, Spain
| | - Julie M J Lepesant
- Laboratoire de biologie cellulaire et moléculaire du contrôle de la prolifération (LBCMCP), Université Paul Sabatier, CNRS, Toulouse, France
| | - Miren Roldán
- Neuroepigenetics Laboratory, Navarrabiomed, Public University of Navarre (UPNA), IdiSNA (Navarra Institute for Health Research), c/ Irunlarrea, 3, 31008, Pamplona, Spain
| | - Idoia Blanco-Luquin
- Neuroepigenetics Laboratory, Navarrabiomed, Public University of Navarre (UPNA), IdiSNA (Navarra Institute for Health Research), c/ Irunlarrea, 3, 31008, Pamplona, Spain
| | - Álvaro Perdones
- Bioinformatics Unit, Navarrabiomed, Public University of Navarre (UPNA), IdiSNA (Navarra Institute for Health Research), Pamplona, Spain
| | - Rosa Larumbe
- Neuroepigenetics Laboratory, Navarrabiomed, Public University of Navarre (UPNA), IdiSNA (Navarra Institute for Health Research), c/ Irunlarrea, 3, 31008, Pamplona, Spain.,Department of Neurology, Complejo Hospitalario de Navarra, IdiSNA (Navarra Institute for Health Research), Pamplona, Spain
| | - Ivonne Jericó
- Department of Neurology, Complejo Hospitalario de Navarra, IdiSNA (Navarra Institute for Health Research), Pamplona, Spain
| | - Carmen Echavarri
- Neuroepigenetics Laboratory, Navarrabiomed, Public University of Navarre (UPNA), IdiSNA (Navarra Institute for Health Research), c/ Irunlarrea, 3, 31008, Pamplona, Spain.,Department of Neurology, Complejo Hospitalario de Navarra, IdiSNA (Navarra Institute for Health Research), Pamplona, Spain
| | - Iván Méndez-López
- Neuroepigenetics Laboratory, Navarrabiomed, Public University of Navarre (UPNA), IdiSNA (Navarra Institute for Health Research), c/ Irunlarrea, 3, 31008, Pamplona, Spain.,Department of Internal Medicine, Hospital García-Orcoyen, Estella, Spain
| | - Luisa Di Stefano
- Laboratoire de biologie cellulaire et moléculaire du contrôle de la prolifération (LBCMCP), Université Paul Sabatier, CNRS, Toulouse, France
| | - Maite Mendioroz
- Neuroepigenetics Laboratory, Navarrabiomed, Public University of Navarre (UPNA), IdiSNA (Navarra Institute for Health Research), c/ Irunlarrea, 3, 31008, Pamplona, Spain. .,Department of Neurology, Complejo Hospitalario de Navarra, IdiSNA (Navarra Institute for Health Research), Pamplona, Spain.
| |
Collapse
|
148
|
Chao F, Jiang L, Zhang Y, Zhou C, Zhang L, Tang J, Liang X, Qi Y, Zhu Y, Ma J, Tang Y. Stereological Investigation of the Effects of Treadmill Running Exercise on the Hippocampal Neurons in Middle-Aged APP/PS1 Transgenic Mice. J Alzheimers Dis 2019; 63:689-703. [PMID: 29689723 DOI: 10.3233/jad-171017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The risk of cognitive decline during Alzheimer's disease (AD) can be reduced if physical activity is maintained; however, the specific neural events underlying this beneficial effect are still uncertain. To quantitatively investigate the neural events underlying the effect of running exercise on middle-aged AD subjects, 12-month-old male APP/PS1 mice were randomly assigned to a control group or running group, and age-matched non-transgenic littermates were used as a wild-type group. AD running group mice were subjected to a treadmill running protocol (regular and moderate intensity) for four months. Spatial learning and memory abilities were assessed using the Morris water maze. Hippocampal amyloid plaques were observed using Thioflavin S staining and immunohistochemistry. Hippocampal volume, number of neurons, and number of newborn cells (BrdU+ cells) in the hippocampus were estimated using stereological techniques, and newborn neurons were observed using double-labelling immunofluorescence. Marked neuronal loss in both the CA1 field and dentate gyrus (DG) and deficits in both the neurogenesis and survival of new neurons in the DG of middle-aged APP/PS1 mice were observed. Running exercise could improve the spatial learning and memory abilities, reduce amyloid plaques in the hippocampi, delay neuronal loss, induce neurogenesis, and promote the survival of newborn neurons in the DG of middle-aged APP/PS1 mice. Exercise-induced protection of neurons and adult neurogenesis within the DG might be part of the important structural basis of the improved spatial learning and memory abilities observed in AD mice.
Collapse
Affiliation(s)
- Fenglei Chao
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Lin Jiang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Yi Zhang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Chunni Zhou
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Lei Zhang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Jing Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Xin Liang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Yingqiang Qi
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Yanqing Zhu
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Jing Ma
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Yong Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| |
Collapse
|
149
|
Arellano JI, Harding B, Thomas JL. Adult Human Hippocampus: No New Neurons in Sight. Cereb Cortex 2019; 28:2479-2481. [PMID: 29746611 DOI: 10.1093/cercor/bhy106] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 04/20/2018] [Indexed: 12/27/2022] Open
Abstract
In this issue of Cerebral Cortex, Cipriani et al. are following up on the recent report of Sorrels et al. to add novel immunohistological observations indicating that, unlike rodents, adult and aging humans do not acquire new neurons in the hippocampus. The common finding emerging from these 2 different, but almost simultaneous studies is highly significant because the dentate gyrus of the hippocampus was, until recently, considered as the only structure in the human brain that may continue neurogenesis throughout the full life span.
Collapse
Affiliation(s)
- Jon I Arellano
- Department of Neuroscience, Yale University, New Haven, CT, USA
| | - Brian Harding
- Department of Pathology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jean-Leon Thomas
- Department of Neurology, Yale University, New Haven, CT, USA.,Université Pierre et Marie Curie, Paris 06, UMR S 1127, Sorbonne Université, Institut du Cerveau et de la Moelle Epinière, Paris, France
| |
Collapse
|
150
|
Protective effects of melatonin against valproic acid-induced memory impairments and reductions in adult rat hippocampal neurogenesis. Neuroscience 2019; 406:580-593. [DOI: 10.1016/j.neuroscience.2019.02.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 01/12/2023]
|