1
|
Umeda T, Sakai A, Uekado R, Shigemori K, Nakajima R, Yamana K, Tomiyama T. Simply crushed zizyphi spinosi semen prevents neurodegenerative diseases and reverses age-related cognitive decline in mice. eLife 2025; 13:RP100737. [PMID: 40266679 PMCID: PMC12017767 DOI: 10.7554/elife.100737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025] Open
Abstract
Neurodegenerative diseases are age-related disorders characterized by the cerebral accumulation of amyloidogenic proteins, and cellular senescence underlies their pathogenesis. Thus, it is necessary for preventing these diseases to remove toxic proteins, repair damaged neurons, and suppress cellular senescence. As a source for such prophylactic agents, we selected zizyphi spinosi semen (ZSS), a medicinal herb used in traditional Chinese medicine. Oral administration of ZSS hot water extract ameliorated Aβ and tau pathology and cognitive impairment in mouse models of Alzheimer's disease and frontotemporal dementia. Non-extracted ZSS simple crush powder showed stronger effects than the extract and improved α-synuclein pathology and cognitive/motor function in Parkinson's disease model mice. Furthermore, when administered to normal aged mice, the ZSS powder suppressed cellular senescence, reduced DNA oxidation, promoted brain-derived neurotrophic factor expression and neurogenesis, and enhanced cognition to levels similar to those in young mice. The quantity of known active ingredients of ZSS, jujuboside A, jujuboside B, and spinosin was not proportional to the nootropic activity of ZSS. These results suggest that ZSS simple crush powder is a promising dietary material for the prevention of neurodegenerative diseases and brain aging.
Collapse
Affiliation(s)
- Tomohiro Umeda
- Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of MedicineOsakaJapan
- Cerebro Pharma IncOsakaJapan
| | - Ayumi Sakai
- Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of MedicineOsakaJapan
- Cerebro Pharma IncOsakaJapan
| | - Rumi Uekado
- Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of MedicineOsakaJapan
| | - Keiko Shigemori
- Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of MedicineOsakaJapan
| | - Ryota Nakajima
- NOMON Co., Ltd, and New Business Development Unit, Teijin Ltd, Kasumigaseki Common Gate West TowerTokyoJapan
| | - Kei Yamana
- NOMON Co., Ltd, and New Business Development Unit, Teijin Ltd, Kasumigaseki Common Gate West TowerTokyoJapan
| | - Takami Tomiyama
- Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of MedicineOsakaJapan
- Cerebro Pharma IncOsakaJapan
| |
Collapse
|
2
|
Krishnan B, Marcatti M, Fracassi A, Zhang WR, Guptarak J, Johnson K, Grant A, Kayed R, Taglialatela G, Micci MA. Hippocampal Neural Stem Cell Exosomes Promote Brain Resilience against the Impact of Tau Oligomers. J Neurosci 2025; 45:e1664242025. [PMID: 40050115 PMCID: PMC12005371 DOI: 10.1523/jneurosci.1664-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/20/2024] [Accepted: 01/28/2025] [Indexed: 04/19/2025] Open
Abstract
A promising therapeutic intervention for preventing the onset and progression of Alzheimer's disease is to protect and improve synaptic resilience, a well-established early vulnerability associated with the toxic effects of oligomers of amyloid β (AβO) and Tau (TauO). We have previously reported that exosomes from hippocampal neural stem cells (NSCs) protect synapses against AβO. Here, we demonstrate how exosomes can also shield against TauO toxicity in adult mice synapses, potentially benefiting primary and secondary tauopathies. Exosomes from hippocampal NSCs (NSCexo) or mature neurons (MNexo) were delivered intracerebroventricularly to adult wild-type male mice (C57Bl6/J). After 24 h, TauO were administered to suppress long-term potentiation (LTP) and memory, measured by electrophysiology and contextual memory deficits measured using novel object recognition test. We also assessed TauO binding to synapses using isolated synaptosomes and cultured hippocampal neurons. Furthermore, mimics of select miRNAs present in NSCexo were delivered intracerebroventricularly to mice prior to assessment of TauO-induced suppression of hippocampal LTP. Our results showed that NSC-, not MN-, derived exosomes, prevented TauO-induced memory impairment, LTP suppression, and reduced Tau accumulation and TauO internalization in synaptosomes. These findings suggest that NSC-derived exosomes can protect against synaptic dysfunction and memory deficits induced by both AβO and TauO, offering a novel therapeutic strategy for multiple neurodegenerative states.
Collapse
Affiliation(s)
- Balaji Krishnan
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Michela Marcatti
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Anna Fracassi
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Wen-Ru Zhang
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Jutatip Guptarak
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Kathia Johnson
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Auston Grant
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Giulio Taglialatela
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Maria-Adelaide Micci
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas 77555
| |
Collapse
|
3
|
Chaudhuri S, Cho M, Stumpff JC, Bice PJ, İş Ö, Ertekin-Taner N, Saykin AJ, Nho K. Cell-specific transcriptional signatures of vascular cells in Alzheimer's disease: perspectives, pathways, and therapeutic directions. Mol Neurodegener 2025; 20:12. [PMID: 39876020 PMCID: PMC11776188 DOI: 10.1186/s13024-025-00798-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025] Open
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative disease that is marked by profound neurovascular dysfunction and significant cell-specific alterations in the brain vasculature. Recent advances in high throughput single-cell transcriptomics technology have enabled the study of the human brain vasculature at an unprecedented depth. Additionally, the understudied niche of cerebrovascular cells, such as endothelial and mural cells, and their subtypes have been scrutinized for understanding cellular and transcriptional heterogeneity in AD. Here, we provide an overview of rich transcriptional signatures derived from recent single-cell and single-nucleus transcriptomic studies of human brain vascular cells and their implications for targeted therapy for AD. We conducted an in-depth literature search using Medline and Covidence to identify pertinent AD studies that utilized single-cell technologies in human post-mortem brain tissue by focusing on understanding the transcriptional differences in cerebrovascular cell types and subtypes in AD and cognitively normal older adults. We also discuss impaired cellular crosstalk between vascular cells and neuroglial units, as well as astrocytes in AD. Additionally, we contextualize the findings from single-cell studies of distinct endothelial cells, smooth muscle cells, fibroblasts, and pericytes in the human AD brain and highlight pathways for potential therapeutic interventions as a concerted multi-omic effort with spatial transcriptomics technology, neuroimaging, and neuropathology. Overall, we provide a detailed account of the vascular cell-specific transcriptional signatures in AD and their crucial cellular crosstalk with the neuroglial unit.
Collapse
Affiliation(s)
- Soumilee Chaudhuri
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Medical Neuroscience Graduate Program, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Minyoung Cho
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Julia C Stumpff
- Ruth Lilly Medical Library, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Paula J Bice
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Özkan İş
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
4
|
Sebastian A, Shanmuganathan MAA, Tripathy C, Chakravarty S, Ghosh S. Understanding Neurogenesis and Neuritogenesis via Molecular Insights, Gender Influence, and Therapeutic Implications: Intervention of Nanomaterials. ACS APPLIED BIO MATERIALS 2025; 8:12-41. [PMID: 39718903 DOI: 10.1021/acsabm.4c01079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
Neurological disorders impact global health by affecting both central and peripheral nervous systems. Understanding the neurogenic processes, i.e., neurogenesis and neuritogenesis, is of paramount importance in the context of nervous system development and regeneration as they hold promising therapeutic implications. Neurogenesis forms functional neurons from precursor cells, while neuritogenesis involves extending neurites for neuron connections. This review discusses how these processes are influenced by genetics, epigenetics, neurotrophic factors, environment, neuroinflammation, and neurotransmitters. It also covers gender-specific aspects of neurogenesis and neuritogenesis, their impact on brain plasticity, and susceptibility to neurological disorders. Alterations in these processes, under the influence of cytokines, growth factors, neurotransmitters, and aging, are linked to neurological disorders and potential therapeutic targets. Gender-specific effects of pharmacological interventions, like SSRIs, TCAs, atypical antipsychotics, and lithium, are explored in this review. Hormone-mediated effects of BDNF and PPAR-γ agonists, as well as variations in efficacy and tolerability of MAOIs, AEDs, NMDA receptor modulators, and ampakines, are detailed for accurate therapeutic design. The review also discusses nanotechnology's significant contribution to neural tissue regeneration for mending neurodegenerative disorders, enhancing neuronal connectivity, and stem cell differentiation. Gold nanoparticles support hippocampal neurogenesis, while other nanoparticles aid neuron growth and neurite outgrowth. Quantum dots and nanolayered double hydroxides assist neuroregeneration, which improves brain drug delivery. Gender-specific responses to nanomedicines designed to enhance neuroregeneration have not been extensively investigated. However, we have specified certain gender-related variables that should be taken into account during the development of nanomedicines in an aim to improve therapeutic efficacy. Further research on gender-specific responses to nanomedicines in neural processes could enhance personalized treatments for neurological disorders, paving the way for novel therapeutic approaches in neuroscience.
Collapse
Affiliation(s)
- Aishwarya Sebastian
- Polymers & Functional Materials Division, CSIR- Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Mohanraj Alias Ayyappan Shanmuganathan
- Polymers & Functional Materials Division, CSIR- Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Chinmayee Tripathy
- Applied Biology Division, CSIR- Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sumana Chakravarty
- Applied Biology Division, CSIR- Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sutapa Ghosh
- Polymers & Functional Materials Division, CSIR- Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
5
|
Li D, Rongchun W, Lu W, Ma Y. Exploring the potential of MFG-E8 in neurodegenerative diseases. Crit Rev Food Sci Nutr 2024:1-15. [PMID: 39468823 DOI: 10.1080/10408398.2024.2417800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Milk fat globule-epidermal growth factor 8 (MFG-E8) is a multifunctional glycoprotein regulating intercellular interactions in various biological and pathological processes. This review summarizes the effects and mechanisms of MFG-E8 in neurodegenerative diseases (NDDs), emphasizing its roles in inflammation, apoptosis, and oxidative stress. In this review, will also explore the potential of MFG-E8 as a diagnostic biomarker and its therapeutic applications in neurodegenerative disorders. Recent studies have revealed intriguing characteristics of using MFG-E8 as a potential drug for treating various brain disorders. While the discovery, origin, expression, and physiological functions of MFG-E8 in various organs and tissues are well defined, its role in the brain remains less understood. This is particularly true for NDDs, indicating unmet medical needs. Elucidating its role in the brain could position MFG-E8 as a potential treatment for NDDs.
Collapse
Affiliation(s)
- Dan Li
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Wang Rongchun
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Weihong Lu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Ying Ma
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
6
|
Mallick D, Acharjee A, Acharjee P, Trigun SK. Restoration of hippocampal adult neurogenesis by CDRI-08 (Bacopa monnieri extract) relates with the recovery of BDNF-TrkB levels in male rats with moderate grade hepatic encephalopathy. Int J Dev Neurosci 2024; 84:510-519. [PMID: 38795011 DOI: 10.1002/jdn.10350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/01/2024] [Accepted: 05/14/2024] [Indexed: 05/27/2024] Open
Abstract
Modulation of in vivo adult neurogenesis (AN) is an evolving concept in managing neurodegenerative diseases. CDRI-08, a bacoside-enriched fraction of Bacopa monnieri, has been demonstrated for its neuroprotective actions, but its effect on AN remains unexplored. This article describes the status of AN by monitoring neuronal stem cells (NSCs) proliferation, differentiation/maturation markers and BDNF-TrkB levels (NSCs signalling players) vs. the level of neurodegeneration and their modulations by CDRI-08 in the hippocampal dentate gyrus (DG) of male rats with moderate grade hepatic encephalopathy (MoHE). For NSC proliferation, 10 mg/kg b.w. 5-bromo-2'-deoxyuridine (BrdU) was administered i.p. during the last 3 days, and for the NSC differentiation study, it was given during the first 3 days to the control, the MoHE (developed by 100 mg/kg b.w. of thioacetamide i.p. up to 10 days) and to the MoHE male rats co-treated with 350 mg/kg b.w. CDRI-08. Compared with the control rats, the hippocampus DG region of MoHE rats showed significant decreases in the number of Nestin+/BrdU+ and SOX2+/BrdU+ (proliferating) and DCX+/BrdU+ and NeuN+/BrdU+ (differentiating) NSCs. This was consistent with a similar decline in BDNF+/TrkB+ NSCs. However, all these NSC marker positive cells were observed to be recovered to their control levels, with a concordant restoration of total cell numbers in the DG of the CDRI-08-treated MoHE rats. The findings suggest that the restoration of hippocampal AN by CDRI-08 is consistent with the recovery of BDNF-TrkB-expressing NSCs in the MoHE rat model of neurodegeneration.
Collapse
Affiliation(s)
- Debasmit Mallick
- Biochemistry Section, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Arup Acharjee
- Department of Zoology, University of Allahabad, Prayagraj, India
| | - Papia Acharjee
- Biochemistry Section, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Surendra Kumar Trigun
- Biochemistry Section, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
7
|
Geigenmüller JN, Tari AR, Wisloff U, Walker TL. The relationship between adult hippocampal neurogenesis and cognitive impairment in Alzheimer's disease. Alzheimers Dement 2024; 20:7369-7383. [PMID: 39166771 PMCID: PMC11485317 DOI: 10.1002/alz.14179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/14/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024]
Abstract
Neurogenesis persists throughout adulthood in the hippocampus and contributes to specific cognitive functions. In Alzheimer's disease (AD), the hippocampus is affected by pathology and functional impairment early in the disease. Human AD patients have reduced adult hippocampal neurogenesis (AHN) levels compared to age-matched healthy controls. Similarly, rodent AD models show a decrease in AHN before the onset of the classical hallmarks of AD pathology. Conversely, enhancement of AHN can protect against AD pathology and ameliorate memory deficits in both rodents and humans. Therefore, impaired AHN may be a contributing factor of AD-associated cognitive decline, rather than an effect of it. In this review we outline the regulation and function of AHN in healthy individuals, and highlight the relationship between AHN dysfunction and cognitive impairments in AD. The existence of AHN in humans and its relevance in AD patients will also be discussed, with an outlook toward future research directions. HIGHLIGHTS: Adult hippocampal neurogenesis occurs in the brains of mammals including humans. Adult hippocampal neurogenesis is reduced in Alzheimer's disease in humans and animal models.
Collapse
Affiliation(s)
| | - Atefe R. Tari
- The Cardiac Exercise Research Group at Department of Circulation and Medical ImagingFaculty of Medicine and Health SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
- Department of Neurology and Clinical NeurophysiologySt. Olavs University Hospital, Trondheim University HospitalTrondheimNorway
| | - Ulrik Wisloff
- The Cardiac Exercise Research Group at Department of Circulation and Medical ImagingFaculty of Medicine and Health SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
| | - Tara L. Walker
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain InstituteThe University of QueenslandBrisbaneAustralia
| |
Collapse
|
8
|
Xiang X, Jiang X, Lin H, Yu M, Wu L, Zhou R. Acylhydrazone Derivative A5 Promotes Neurogenesis by Up-Regulating Neurogenesis-Related Genes and Inhibiting Cell-Cycle Progression in Neural Stem/Progenitor Cells. Molecules 2024; 29:3330. [PMID: 39064908 PMCID: PMC11279415 DOI: 10.3390/molecules29143330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/09/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Adult neurogenesis involves the generation of functional neurons from neural progenitor cells, which have the potential to complement and restore damaged neurons and neural circuits. Therefore, the development of drugs that stimulate neurogenesis represents a promising strategy in stem cell therapy and neural regeneration, greatly facilitating the reconstruction of neural circuits in cases of neurodegeneration and brain injury. Our study reveals that compound A5, previously designed and synthesized by our team, exhibits remarkable neuritogenic activities, effectively inducing neurogenesis in neural stem/progenitor cells (NSPCs). Subsequently, transcriptome analysis using high-throughput Illumina RNA-seq technology was performed to further elucidate the underlying molecular mechanisms by which Compound A5 promotes neurogenesis. Notably, comparative transcriptome analysis showed that the up-regulated genes were mainly associated with neurogenesis, and the down-regulated genes were mainly concerned with cell cycle progression. Furthermore, we confirmed that Compound A5 significantly affected the expression of transcription factors related to neurogenesis and cell cycle regulatory proteins. Collectively, these findings identify a new compound with neurogenic activity and may provide insights into drug discovery for neural repair and regeneration.
Collapse
Affiliation(s)
- Xiaoliang Xiang
- Hunan Provincial Higher Education Key Laboratory of Intensive Processing Research on Mountain Ecological Food, College of Biological and Food Engineering, Huaihua University, Huaihua 418008, China; (X.J.); (L.W.); (R.Z.)
| | - Xia Jiang
- Hunan Provincial Higher Education Key Laboratory of Intensive Processing Research on Mountain Ecological Food, College of Biological and Food Engineering, Huaihua University, Huaihua 418008, China; (X.J.); (L.W.); (R.Z.)
- College of Chemistry and Materials Engineering, Huaihua University, Huaihua 418008, China;
| | - Hongwei Lin
- College of Chemistry and Materials Engineering, Huaihua University, Huaihua 418008, China;
| | - Meixing Yu
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510180, China;
| | - Liming Wu
- Hunan Provincial Higher Education Key Laboratory of Intensive Processing Research on Mountain Ecological Food, College of Biological and Food Engineering, Huaihua University, Huaihua 418008, China; (X.J.); (L.W.); (R.Z.)
| | - Rong Zhou
- Hunan Provincial Higher Education Key Laboratory of Intensive Processing Research on Mountain Ecological Food, College of Biological and Food Engineering, Huaihua University, Huaihua 418008, China; (X.J.); (L.W.); (R.Z.)
| |
Collapse
|
9
|
Umeda T, Sakai A, Shigemori K, Nakata K, Nakajima R, Yamana K, Tomiyama T. New Value of Acorus tatarinowii/ gramineus Leaves as a Dietary Source for Dementia Prevention. Nutrients 2024; 16:1589. [PMID: 38892521 PMCID: PMC11175135 DOI: 10.3390/nu16111589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
The rhizomes of Acorus tatarinowii Schott and Acorus gramineus Solander are widely used for treating amnesia in traditional Chinese medicine. In contrast, their leaves are usually discarded without their medicinal properties being known. Here, we found that the hot water extract of leaves improved cognition and tau pathology in model mice of frontotemporal dementia, similar to or even better than that of rhizomes. To explore the optimal method of processing, we made three preparations from dried leaves: hot water extract, extraction residue, and non-extracted simple crush powder. Among them, the simple crush powder had the strongest effect on tauopathy in mice. The crush powder also ameliorated Aβ and α-synuclein pathologies and restored cognition in mouse models of Alzheimer's disease and dementia with Lewy bodies. These findings suggest the potential of Acorus tatarinowii/gramineus leaves as a dietary source for dementia prevention and reveal that simple crushing is a better way to maximize their efficacy.
Collapse
Affiliation(s)
- Tomohiro Umeda
- Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan; (T.U.)
- Cerebro Pharma Inc., 4-5-6-3F Minamikyuhojimachi, Chuo-ku, Osaka 541-0058, Japan
| | - Ayumi Sakai
- Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan; (T.U.)
- Cerebro Pharma Inc., 4-5-6-3F Minamikyuhojimachi, Chuo-ku, Osaka 541-0058, Japan
| | - Keiko Shigemori
- Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan; (T.U.)
| | - Kunio Nakata
- NOMON Co., Ltd., New Business Development Unit, Teijin Ltd., Kasumigaseki Common Gate West Tower 3-2-1 Kasumigaseki, Chiyoda-ku, Tokyo 100-8585, Japan; (K.N.); (R.N.); (K.Y.)
| | - Ryota Nakajima
- NOMON Co., Ltd., New Business Development Unit, Teijin Ltd., Kasumigaseki Common Gate West Tower 3-2-1 Kasumigaseki, Chiyoda-ku, Tokyo 100-8585, Japan; (K.N.); (R.N.); (K.Y.)
| | - Kei Yamana
- NOMON Co., Ltd., New Business Development Unit, Teijin Ltd., Kasumigaseki Common Gate West Tower 3-2-1 Kasumigaseki, Chiyoda-ku, Tokyo 100-8585, Japan; (K.N.); (R.N.); (K.Y.)
| | - Takami Tomiyama
- Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan; (T.U.)
- Cerebro Pharma Inc., 4-5-6-3F Minamikyuhojimachi, Chuo-ku, Osaka 541-0058, Japan
| |
Collapse
|
10
|
Liu Q, Li W, Chen Y, Zhang S, Sun Z, Yang Y, Lv P, Yin Y. Effects of repetitive transcranial magnetic stimulation combined with music therapy in non-fluent aphasia after stroke: A randomised controlled study. INTERNATIONAL JOURNAL OF LANGUAGE & COMMUNICATION DISORDERS 2024; 59:1211-1222. [PMID: 38088533 DOI: 10.1111/1460-6984.12991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 11/11/2023] [Indexed: 05/09/2024]
Abstract
BACKGROUND Although existing studies have shown that both repetitive transcranial magnetic stimulation (rTMS) and music therapy have advantages in the treatment of non-fluent aphasia, the efficacy of the combination of these two methods remains to be investigated. AIMS To investigate the clinical efficacy of low-frequency rTMS combined with music therapy on language function and depression in patients with non-fluent aphasia after stroke. METHODS & PROCEDURES A single-blind parallel randomised controlled trial was conducted. Sixty patients (mean duration = 93.78 days) with non-fluent aphasia after stroke were randomly divided into a traditional therapy group (n = 20), a music therapy group (n = 20) and a combined therapy group (n = 20, 1 Hz). The language function and depression were evaluated before and 3 weeks after treatment with the Chinese version of the Western Aphasia Battery scale, Boston Diagnostic Aphasia Examination scale and Stroke Aphasic Depression Questionnaire Hospital Version scale. OUTCOMES & RESULTS The combined therapy group was significantly better in all outcomes than the traditional therapy group and was significantly better in depression than the music therapy group. The music therapy group was significantly better in repetition and depression than the traditional therapy group. Language improvement was positively correlated with depression improvement. For adverse events, only two patients in the combined therapy group showed slight dizziness during rTMS treatment and their symptoms improved after rest. CONCLUSIONS & IMPLICATIONS Our preliminary randomised controlled study indicates that low-frequency rTMS combined with music therapy is feasible and safe in improving language function and depression in non-fluent aphasia patients after stroke. WHAT THIS PAPER ADDS What is already known on this subject Repetitive transcranial magnetic stimulation (rTMS) and music therapy respectively have advantages in the treatment of non-fluent aphasia after stroke, but whether the combination of the two methods is more effective is still unknown. What this paper adds to the existing knowledge This is one of the first randomised control trials to investigate whether the clinical efficacy of low-frequency rTMS combined music therapy for non-fluent aphasia is better. The findings show that low-frequency rTMS combined music therapy is superior to traditional therapy in spontaneous speech, auditory comprehension, repetition, naming, aphasia quotient, functional language level and depression, and superior to music therapy in depression, while music therapy is superior to traditional therapy in repetition and depression. What are the potential or actual clinical implications of this work? Low-frequency rTMS combined music therapy may be a better method for treatment of non-fluent aphasia.
Collapse
Affiliation(s)
- Qingqing Liu
- Department of Rehabilitation Medicine, Hebei General Hospital, Shijiazhuang, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, China
| | - Weibo Li
- Department of Gastrointestinal Surgery, the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yuanwu Chen
- Department of Rehabilitation Medicine, Hebei General Hospital, Shijiazhuang, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, China
| | - Shaohua Zhang
- The Eighth People's Hospital of Hebei Province, Shijiazhuang, China
| | - Zengxin Sun
- Department of Rehabilitation Medicine, Hebei General Hospital, Shijiazhuang, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, China
| | - Yuhui Yang
- Department of Rehabilitation Medicine, Hebei General Hospital, Shijiazhuang, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, China
| | - Peiyuan Lv
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, China
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Yu Yin
- Department of Rehabilitation Medicine, Hebei General Hospital, Shijiazhuang, China
- Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, China
| |
Collapse
|
11
|
Umeda T, Shigemori K, Uekado R, Matsuda K, Tomiyama T. Hawaiian native herb Mamaki prevents dementia by ameliorating neuropathology and repairing neurons in four different mouse models of neurodegenerative diseases. GeroScience 2024; 46:1971-1987. [PMID: 37783918 PMCID: PMC10828292 DOI: 10.1007/s11357-023-00950-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 09/18/2023] [Indexed: 10/04/2023] Open
Abstract
Neurodegenerative diseases including Alzheimer's disease, frontotemporal dementia, and dementia with Lewy bodies are age-related disorders and the main cause of dementia. They are characterized by the cerebral accumulation of Aβ, tau, α-synuclein, and TDP-43. Because the accumulation begins decades before disease onset, treatment should be started in the preclinical stage. Such intervention would be long-lasting, and therefore, prophylactic agents should be safe, non-invasively taken by the patients, and inexpensive. In addition, the agents should be broadly effective against etiologic proteins and capable of repairing neurons damaged by toxic oligomers. These requirements are difficult to meet with single-ingredient pharmaceuticals but may be feasible by taking proper diets composed of multiple ingredients. As a source of such diets, we focused on the Hawaiian native herb Mamaki. From its dried leaves and fruits, we made three preparations: hot water extract of the leaves, non-extracted simple crush powder of the leaves, and simple crush powder of the fruits, and examined their effects on the cognitive function and neuropathologies in four different mouse models of neurodegenerative dementia. Hot water extract of the leaves attenuated neuropathologies, restored synaptophysin levels, suppressed microglial activation, and improved memory when orally administered for 1 month. Simply crushed leaf powder showed a higher efficacy, but simply crushed fruit powder displayed the strongest effects. Moreover, the fruit powder significantly enhanced the levels of brain-derived neurotrophic factor expression and neurogenesis, indicating its ability to repair neurons. These results suggest that crushed Mamaki leaves and fruits are promising sources of dementia-preventive diets.
Collapse
Affiliation(s)
- Tomohiro Umeda
- Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Osaka, Abeno-ku, 545-8585, Japan
- Cerebro Pharma Inc, 4-5-6-3F Minamikyuhojimachi, Osaka, Chuo-ku, 541-0058, Japan
| | - Keiko Shigemori
- Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Osaka, Abeno-ku, 545-8585, Japan
| | - Rumi Uekado
- Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Osaka, Abeno-ku, 545-8585, Japan
| | - Kazunori Matsuda
- Cerebro Pharma Inc, 4-5-6-3F Minamikyuhojimachi, Osaka, Chuo-ku, 541-0058, Japan
| | - Takami Tomiyama
- Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Osaka, Abeno-ku, 545-8585, Japan.
- Cerebro Pharma Inc, 4-5-6-3F Minamikyuhojimachi, Osaka, Chuo-ku, 541-0058, Japan.
| |
Collapse
|
12
|
Liu X, Liu Y, Liu J, Zhang H, Shan C, Guo Y, Gong X, Cui M, Li X, Tang M. Correlation between the gut microbiome and neurodegenerative diseases: a review of metagenomics evidence. Neural Regen Res 2024; 19:833-845. [PMID: 37843219 PMCID: PMC10664138 DOI: 10.4103/1673-5374.382223] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/19/2023] [Accepted: 06/17/2023] [Indexed: 10/17/2023] Open
Abstract
A growing body of evidence suggests that the gut microbiota contributes to the development of neurodegenerative diseases via the microbiota-gut-brain axis. As a contributing factor, microbiota dysbiosis always occurs in pathological changes of neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. High-throughput sequencing technology has helped to reveal that the bidirectional communication between the central nervous system and the enteric nervous system is facilitated by the microbiota's diverse microorganisms, and for both neuroimmune and neuroendocrine systems. Here, we summarize the bioinformatics analysis and wet-biology validation for the gut metagenomics in neurodegenerative diseases, with an emphasis on multi-omics studies and the gut virome. The pathogen-associated signaling biomarkers for identifying brain disorders and potential therapeutic targets are also elucidated. Finally, we discuss the role of diet, prebiotics, probiotics, postbiotics and exercise interventions in remodeling the microbiome and reducing the symptoms of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaoyan Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Yi Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
- Institute of Animal Husbandry, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu Province, China
| | - Junlin Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Hantao Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Chaofan Shan
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Yinglu Guo
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Xun Gong
- Department of Rheumatology & Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Mengmeng Cui
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Xiubin Li
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Min Tang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| |
Collapse
|
13
|
Singh L, Bhatti R. Signaling Pathways Involved in the Neuroprotective Effect of Osthole: Evidence and Mechanisms. Mol Neurobiol 2024; 61:1100-1118. [PMID: 37682453 DOI: 10.1007/s12035-023-03580-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/14/2023] [Indexed: 09/09/2023]
Abstract
Neurodegenerative diseases constitute a major threat to human health and are usually accompanied by progressive structural and functional loss of neurons. Abnormalities in synaptic plasticity are involved in neurodegenerative disorders. Aberrant cell signaling cascades play a predominant role in the initiation, progress as well as in the severity of these ailments. Notch signaling is a pivotal role in the maintenance of neural stem cells and also participates in neurogenesis. PI3k/Akt cascade regulates different biological processes including cell proliferation, apoptosis, and metabolism. It regulates neurotoxicity and mediates the survival of neurons. Moreover, the activated BDNF/TrkB cascade is involved in promoting the transcription of genes responsible for cell survival and neurogenesis. Despite significant progress made in delineating the underlying pathological mechanisms involved and derangements in cellular metabolic promenades implicated in these diseases, satisfactory strategies for the clinical management of these ailments are yet to be achieved. Therefore, the molecules targeting these cell signaling cascades may emerge as useful leads in developing newer management strategies. Osthole is an important ingredient of traditional Chinese medicinal plants, often found in various plants of the Apiaceae family and has been observed to target these aforementioned mediators. Until now, no review has been aimed to discuss the possible molecular signaling cascades involved in osthole-mediated neuroprotection at one platform. The current review aimed to explore the interplay of various mediators and the modulation of the different molecular signaling cascades in osthole-mediated neuroprotection. This review could open new insights into research involving diseases of neuronal origin, especially the effect on neurodegeneration, neurogenesis, and synaptic plasticity. The articles gathered to compose the current review were extracted by using the PubMed, Scopus, Science Direct, and Web of Science databases. A methodical approach was used to integrate and discuss all published original reports describing the modulation of different mediators by osthole to confer neuroprotection at one platform to provide possible molecular pathways. Based on the inclusion and exclusion criteria, 32 articles were included in the systematic review. Moreover, literature evidence was also used to construct the biosynthetic pathway of osthole. The current review reveals that osthole promotes neurogenesis and neuronal functioning via stimulation of Notch, BDNF/Trk, and P13k/Akt signaling pathways. It upregulates the expression of various proteins, such as BDNF, TrkB, CREB, Nrf-2, P13k, and Akt. Activation of Wnt by osthole, in turn, regulates downstream GSK-1β to inhibit tau phosphorylation and β-catenin degradation to prevent neuronal apoptosis. The activation of Wnt and inhibition of oxidative stress, Aβ, and GSK-3β mediated β-catenin degradation by osthole might also be involved in mediating the protection against neurodegenerative diseases. Furthermore, it also inhibits neuroinflammation by suppressing MAPK/NF-κB-mediated transcription of genes involved in the generation of inflammatory cytokines and NLRP-3 inflammasomes. This review delineates the various underlying signaling pathways involved in mediating the neuroprotective effect of osthole. Modulation of Notch, BDNF/Trk, MAPK/NF-κB, and P13k/Akt signaling pathways by osthole confers protection against neurodegenerative diseases. The preclinical effects of osthole suggest that it could be a valuable molecule in inspiring the development of new drugs for the management of neurodegenerative diseases and demands clinical studies to explore its potential. An effort has been made to unify the varied mechanisms and target sites involved in the neuroprotective effect of osthole. The comprehensive description of the molecular pathways in the present work reflects its originality and thoroughness. The reviewed literature findings may be extrapolated to suggest the role of othole as a "biological response modifier" which contributes to neuroprotection through kinase modulatory, immunomodulatory, and anti-oxidative activity, which is documented even at lower doses. The current review attempts to emphasize the gaps in the existing literature which can be explored in the future.
Collapse
Affiliation(s)
- Lovedeep Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, Punjab, India.
- University Institute of Pharma Sciences, Chandigarh University, Mohali, 140413, Punjab, India.
| | - Rajbir Bhatti
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, Punjab, India.
| |
Collapse
|
14
|
Sharma K, Puranik N, Yadav D. Neural Stem Cell-based Regenerative Therapy: A New Approach to Diabetes Treatment. Endocr Metab Immune Disord Drug Targets 2024; 24:531-540. [PMID: 37183465 DOI: 10.2174/1871530323666230512121416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 05/16/2023]
Abstract
Diabetes mellitus (DM) is the most common metabolic disorder that occurs due to the loss, or impaired function of insulin-secreting pancreatic beta cells, which are of two types - type 1 (T1D) and type 2 (T2D). To cure DM, the replacement of the destroyed pancreatic beta cells of islet of Langerhans is the most widely practiced treatment. For this, isolating neuronal stem cells and cultivating them as a source of renewable beta cells is a significant breakthrough in medicine. The functions, growth, and gene expression of insulin-producing pancreatic beta cells and neurons are very similar in many ways. A diabetic patient's neural stem cells (obtained from the hippocampus and olfactory bulb) can be used as a replacement source of beta cells for regenerative therapy to treat diabetes. The same protocol used to create functional neurons from progenitor cells can be used to create beta cells. Recent research suggests that replacing lost pancreatic beta cells with autologous transplantation of insulin-producing neural progenitor cells may be a perfect therapeutic strategy for diabetes, allowing for a safe and normal restoration of function and a reduction in potential risks and a long-term cure.
Collapse
Affiliation(s)
- Kajal Sharma
- School of Sciences in Biotechnology, Jiwaji University, Gwalior, 474011, Madhya Pradesh, India
| | - Nidhi Puranik
- Department of Bio-logical Sciences, Bharathiar University, Tamil Nadu, India
| | - Dhananjay Yadav
- Department of Life Science, Yeungnam University, Gyeongsan, 38541, Korea
| |
Collapse
|
15
|
Hussain G, Akram R, Anwar H, Sajid F, Iman T, Han HS, Raza C, De Aguilar JLG. Adult neurogenesis: a real hope or a delusion? Neural Regen Res 2024; 19:6-15. [PMID: 37488837 PMCID: PMC10479850 DOI: 10.4103/1673-5374.375317] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/27/2023] [Accepted: 04/10/2023] [Indexed: 07/26/2023] Open
Abstract
Adult neurogenesis, the process of creating new neurons, involves the coordinated division, migration, and differentiation of neural stem cells. This process is restricted to neurogenic niches located in two distinct areas of the brain: the subgranular zone of the dentate gyrus of the hippocampus and the subventricular zone of the lateral ventricle, where new neurons are generated and then migrate to the olfactory bulb. Neurogenesis has been thought to occur only during the embryonic and early postnatal stages and to decline with age due to a continuous depletion of neural stem cells. Interestingly, recent years have seen tremendous progress in our understanding of adult brain neurogenesis, bridging the knowledge gap between embryonic and adult neurogenesis. Here, we discuss the current status of adult brain neurogenesis in light of what we know about neural stem cells. In this notion, we talk about the importance of intracellular signaling molecules in mobilizing endogenous neural stem cell proliferation. Based on the current understanding, we can declare that these molecules play a role in targeting neurogenesis in the mature brain. However, to achieve this goal, we need to avoid the undesired proliferation of neural stem cells by controlling the necessary checkpoints, which can lead to tumorigenesis and prove to be a curse instead of a blessing or hope.
Collapse
Affiliation(s)
- Ghulam Hussain
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Punjab, Pakistan
| | - Rabia Akram
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Punjab, Pakistan
| | - Haseeb Anwar
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Punjab, Pakistan
| | - Faiqa Sajid
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Punjab, Pakistan
| | - Tehreem Iman
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Punjab, Pakistan
| | - Hyung Soo Han
- Department of Physiology, School of Medicine, Clinical Omics Institute, Kyungpook National University, Daegu, Korea
| | - Chand Raza
- Department of Zoology, Faculty of Chemistry and Life Sciences, Government College University, Lahore, Pakistan
| | - Jose-Luis Gonzalez De Aguilar
- INSERM, U1118, Mécanismes Centraux et Péripheriques de la Neurodégénérescence, Strasbourg, France, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
16
|
Wu A, Zhang J. Neuroinflammation, memory, and depression: new approaches to hippocampal neurogenesis. J Neuroinflammation 2023; 20:283. [PMID: 38012702 PMCID: PMC10683283 DOI: 10.1186/s12974-023-02964-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023] Open
Abstract
As one of most common and severe mental disorders, major depressive disorder (MDD) significantly increases the risks of premature death and other medical conditions for patients. Neuroinflammation is the abnormal immune response in the brain, and its correlation with MDD is receiving increasing attention. Neuroinflammation has been reported to be involved in MDD through distinct neurobiological mechanisms, among which the dysregulation of neurogenesis in the dentate gyrus (DG) of the hippocampus (HPC) is receiving increasing attention. The DG of the hippocampus is one of two niches for neurogenesis in the adult mammalian brain, and neurotrophic factors are fundamental regulators of this neurogenesis process. The reported cell types involved in mediating neuroinflammation include microglia, astrocytes, oligodendrocytes, meningeal leukocytes, and peripheral immune cells which selectively penetrate the blood-brain barrier and infiltrate into inflammatory regions. This review summarizes the functions of the hippocampus affected by neuroinflammation during MDD progression and the corresponding influences on the memory of MDD patients and model animals.
Collapse
Affiliation(s)
- Anbiao Wu
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Jiyan Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China.
| |
Collapse
|
17
|
Assogna M, Premi E, Gazzina S, Benussi A, Ashton NJ, Zetterberg H, Blennow K, Gasparotti R, Padovani A, Tadayon E, Romanella S, Sprugnoli G, Pascual-Leone A, Di Lorenzo F, Koch G, Borroni B, Santarnecchi E. Association of Choroid Plexus Volume With Serum Biomarkers, Clinical Features, and Disease Severity in Patients With Frontotemporal Lobar Degeneration Spectrum. Neurology 2023; 101:e1218-e1230. [PMID: 37500561 PMCID: PMC10516270 DOI: 10.1212/wnl.0000000000207600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 05/15/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Choroid plexus (ChP) is emerging as a key brain structure in the pathophysiology of neurodegenerative disorders. In this observational study, we investigated ChP volume in a large cohort of patients with frontotemporal lobar degeneration (FTLD) spectrum to explore a possible link between ChP volume and other disease-specific biomarkers. METHODS Participants included patients meeting clinical criteria for a probable syndrome in the FTLD spectrum. Structural brain MRI imaging, serum neurofilament light (NfL), serum phosphorylated-Tau181 (p-Tau181), and cognitive and behavioral data were collected. MRI ChP volumes were obtained from an ad-hoc segmentation model based on a Gaussian Mixture Models algorithm. RESULTS Three-hundred and sixteen patients within FTLD spectrum were included in this study, specifically 135 patients diagnosed with behavioral variant frontotemporal dementia (bvFTD), 75 primary progressive aphasia, 46 progressive supranuclear palsy, and 60 corticobasal syndrome. In addition, 82 age-matched healthy participants were recruited as controls (HCs). ChP volume was significantly larger in patients with FTLD compared with HC, across the clinical subtype. Moreover, we found a significant difference in ChP volume between HC and patients stratified for disease-severity based on CDR plus NACC FTLD, including patients at very early stage of the disease. Interestingly, ChP volume correlated with serum NfL, cognitive/behavioral deficits, and with patterns of cortical atrophy. Finally, ChP volume seemed to discriminate HC from patients with FTLD better than other previously identified brain structure volumes. DISCUSSION Considering the clinical, pathologic, and genetic heterogeneity of the disease, ChP could represent a potential biomarker across the FTLD spectrum, especially at the early stage of disease. Further longitudinal studies are needed to establish its role in disease onset and progression. CLASSIFICATION OF EVIDENCE This study provides Class III evidence that choroid plexus volume, as measured on MRI scan, can assist in differentiating patients with FTLD from healthy controls and in characterizing disease severity.
Collapse
Affiliation(s)
- Martina Assogna
- From the Precision Neuroscience & Neuromodulation Program (M.A., S.R., G.S., E.S.), Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Non-Invasive Brain Stimulation Unit (M.A., F.D.L., G.K.), Department of Behavioural and Clinical Neurology, Santa Lucia Foundation IRCCS; Memory Clinic (M.A.), Department of Systems Medicine, University of Tor Vergata, Rome; Neurology Unit (E.P., S.G., A.B., A.P., B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; Institute of Neuroscience and Physiology (N.J.A.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Wallenberg Centre for Molecular and Translational Medicine (N.J.A.), University of Gothenburg, Mӧlndal, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), United Kingdom; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Clear Water Bay, Hong Kong, China; Neuroradiology Unit (R.G.), University of Brescia, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation (E.T.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine (G.S.), Surgery and Neuroscience, Siena Brain Investigation & Neuromodulation Laboratory, University of Siena, Siena, Italy; Hinda and Arthur Marcus Institute for Aging Research at Hebrew SeniorLife (A.P.-L.); Department of Neurology (A.P.-L.), Harvard MedicalSchool, Boston, MA, USA; and Department of Neuroscience and Rehabilitation (G.K.), University of Ferrara, Italy
| | - Enrico Premi
- From the Precision Neuroscience & Neuromodulation Program (M.A., S.R., G.S., E.S.), Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Non-Invasive Brain Stimulation Unit (M.A., F.D.L., G.K.), Department of Behavioural and Clinical Neurology, Santa Lucia Foundation IRCCS; Memory Clinic (M.A.), Department of Systems Medicine, University of Tor Vergata, Rome; Neurology Unit (E.P., S.G., A.B., A.P., B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; Institute of Neuroscience and Physiology (N.J.A.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Wallenberg Centre for Molecular and Translational Medicine (N.J.A.), University of Gothenburg, Mӧlndal, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), United Kingdom; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Clear Water Bay, Hong Kong, China; Neuroradiology Unit (R.G.), University of Brescia, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation (E.T.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine (G.S.), Surgery and Neuroscience, Siena Brain Investigation & Neuromodulation Laboratory, University of Siena, Siena, Italy; Hinda and Arthur Marcus Institute for Aging Research at Hebrew SeniorLife (A.P.-L.); Department of Neurology (A.P.-L.), Harvard MedicalSchool, Boston, MA, USA; and Department of Neuroscience and Rehabilitation (G.K.), University of Ferrara, Italy
| | - Stefano Gazzina
- From the Precision Neuroscience & Neuromodulation Program (M.A., S.R., G.S., E.S.), Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Non-Invasive Brain Stimulation Unit (M.A., F.D.L., G.K.), Department of Behavioural and Clinical Neurology, Santa Lucia Foundation IRCCS; Memory Clinic (M.A.), Department of Systems Medicine, University of Tor Vergata, Rome; Neurology Unit (E.P., S.G., A.B., A.P., B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; Institute of Neuroscience and Physiology (N.J.A.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Wallenberg Centre for Molecular and Translational Medicine (N.J.A.), University of Gothenburg, Mӧlndal, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), United Kingdom; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Clear Water Bay, Hong Kong, China; Neuroradiology Unit (R.G.), University of Brescia, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation (E.T.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine (G.S.), Surgery and Neuroscience, Siena Brain Investigation & Neuromodulation Laboratory, University of Siena, Siena, Italy; Hinda and Arthur Marcus Institute for Aging Research at Hebrew SeniorLife (A.P.-L.); Department of Neurology (A.P.-L.), Harvard MedicalSchool, Boston, MA, USA; and Department of Neuroscience and Rehabilitation (G.K.), University of Ferrara, Italy
| | - Alberto Benussi
- From the Precision Neuroscience & Neuromodulation Program (M.A., S.R., G.S., E.S.), Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Non-Invasive Brain Stimulation Unit (M.A., F.D.L., G.K.), Department of Behavioural and Clinical Neurology, Santa Lucia Foundation IRCCS; Memory Clinic (M.A.), Department of Systems Medicine, University of Tor Vergata, Rome; Neurology Unit (E.P., S.G., A.B., A.P., B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; Institute of Neuroscience and Physiology (N.J.A.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Wallenberg Centre for Molecular and Translational Medicine (N.J.A.), University of Gothenburg, Mӧlndal, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), United Kingdom; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Clear Water Bay, Hong Kong, China; Neuroradiology Unit (R.G.), University of Brescia, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation (E.T.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine (G.S.), Surgery and Neuroscience, Siena Brain Investigation & Neuromodulation Laboratory, University of Siena, Siena, Italy; Hinda and Arthur Marcus Institute for Aging Research at Hebrew SeniorLife (A.P.-L.); Department of Neurology (A.P.-L.), Harvard MedicalSchool, Boston, MA, USA; and Department of Neuroscience and Rehabilitation (G.K.), University of Ferrara, Italy
| | - Nicholas J Ashton
- From the Precision Neuroscience & Neuromodulation Program (M.A., S.R., G.S., E.S.), Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Non-Invasive Brain Stimulation Unit (M.A., F.D.L., G.K.), Department of Behavioural and Clinical Neurology, Santa Lucia Foundation IRCCS; Memory Clinic (M.A.), Department of Systems Medicine, University of Tor Vergata, Rome; Neurology Unit (E.P., S.G., A.B., A.P., B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; Institute of Neuroscience and Physiology (N.J.A.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Wallenberg Centre for Molecular and Translational Medicine (N.J.A.), University of Gothenburg, Mӧlndal, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), United Kingdom; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Clear Water Bay, Hong Kong, China; Neuroradiology Unit (R.G.), University of Brescia, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation (E.T.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine (G.S.), Surgery and Neuroscience, Siena Brain Investigation & Neuromodulation Laboratory, University of Siena, Siena, Italy; Hinda and Arthur Marcus Institute for Aging Research at Hebrew SeniorLife (A.P.-L.); Department of Neurology (A.P.-L.), Harvard MedicalSchool, Boston, MA, USA; and Department of Neuroscience and Rehabilitation (G.K.), University of Ferrara, Italy
| | - Henrik Zetterberg
- From the Precision Neuroscience & Neuromodulation Program (M.A., S.R., G.S., E.S.), Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Non-Invasive Brain Stimulation Unit (M.A., F.D.L., G.K.), Department of Behavioural and Clinical Neurology, Santa Lucia Foundation IRCCS; Memory Clinic (M.A.), Department of Systems Medicine, University of Tor Vergata, Rome; Neurology Unit (E.P., S.G., A.B., A.P., B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; Institute of Neuroscience and Physiology (N.J.A.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Wallenberg Centre for Molecular and Translational Medicine (N.J.A.), University of Gothenburg, Mӧlndal, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), United Kingdom; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Clear Water Bay, Hong Kong, China; Neuroradiology Unit (R.G.), University of Brescia, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation (E.T.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine (G.S.), Surgery and Neuroscience, Siena Brain Investigation & Neuromodulation Laboratory, University of Siena, Siena, Italy; Hinda and Arthur Marcus Institute for Aging Research at Hebrew SeniorLife (A.P.-L.); Department of Neurology (A.P.-L.), Harvard MedicalSchool, Boston, MA, USA; and Department of Neuroscience and Rehabilitation (G.K.), University of Ferrara, Italy
| | - Kaj Blennow
- From the Precision Neuroscience & Neuromodulation Program (M.A., S.R., G.S., E.S.), Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Non-Invasive Brain Stimulation Unit (M.A., F.D.L., G.K.), Department of Behavioural and Clinical Neurology, Santa Lucia Foundation IRCCS; Memory Clinic (M.A.), Department of Systems Medicine, University of Tor Vergata, Rome; Neurology Unit (E.P., S.G., A.B., A.P., B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; Institute of Neuroscience and Physiology (N.J.A.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Wallenberg Centre for Molecular and Translational Medicine (N.J.A.), University of Gothenburg, Mӧlndal, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), United Kingdom; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Clear Water Bay, Hong Kong, China; Neuroradiology Unit (R.G.), University of Brescia, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation (E.T.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine (G.S.), Surgery and Neuroscience, Siena Brain Investigation & Neuromodulation Laboratory, University of Siena, Siena, Italy; Hinda and Arthur Marcus Institute for Aging Research at Hebrew SeniorLife (A.P.-L.); Department of Neurology (A.P.-L.), Harvard MedicalSchool, Boston, MA, USA; and Department of Neuroscience and Rehabilitation (G.K.), University of Ferrara, Italy
| | - Roberto Gasparotti
- From the Precision Neuroscience & Neuromodulation Program (M.A., S.R., G.S., E.S.), Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Non-Invasive Brain Stimulation Unit (M.A., F.D.L., G.K.), Department of Behavioural and Clinical Neurology, Santa Lucia Foundation IRCCS; Memory Clinic (M.A.), Department of Systems Medicine, University of Tor Vergata, Rome; Neurology Unit (E.P., S.G., A.B., A.P., B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; Institute of Neuroscience and Physiology (N.J.A.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Wallenberg Centre for Molecular and Translational Medicine (N.J.A.), University of Gothenburg, Mӧlndal, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), United Kingdom; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Clear Water Bay, Hong Kong, China; Neuroradiology Unit (R.G.), University of Brescia, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation (E.T.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine (G.S.), Surgery and Neuroscience, Siena Brain Investigation & Neuromodulation Laboratory, University of Siena, Siena, Italy; Hinda and Arthur Marcus Institute for Aging Research at Hebrew SeniorLife (A.P.-L.); Department of Neurology (A.P.-L.), Harvard MedicalSchool, Boston, MA, USA; and Department of Neuroscience and Rehabilitation (G.K.), University of Ferrara, Italy
| | - Alessandro Padovani
- From the Precision Neuroscience & Neuromodulation Program (M.A., S.R., G.S., E.S.), Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Non-Invasive Brain Stimulation Unit (M.A., F.D.L., G.K.), Department of Behavioural and Clinical Neurology, Santa Lucia Foundation IRCCS; Memory Clinic (M.A.), Department of Systems Medicine, University of Tor Vergata, Rome; Neurology Unit (E.P., S.G., A.B., A.P., B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; Institute of Neuroscience and Physiology (N.J.A.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Wallenberg Centre for Molecular and Translational Medicine (N.J.A.), University of Gothenburg, Mӧlndal, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), United Kingdom; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Clear Water Bay, Hong Kong, China; Neuroradiology Unit (R.G.), University of Brescia, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation (E.T.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine (G.S.), Surgery and Neuroscience, Siena Brain Investigation & Neuromodulation Laboratory, University of Siena, Siena, Italy; Hinda and Arthur Marcus Institute for Aging Research at Hebrew SeniorLife (A.P.-L.); Department of Neurology (A.P.-L.), Harvard MedicalSchool, Boston, MA, USA; and Department of Neuroscience and Rehabilitation (G.K.), University of Ferrara, Italy
| | - Ehsan Tadayon
- From the Precision Neuroscience & Neuromodulation Program (M.A., S.R., G.S., E.S.), Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Non-Invasive Brain Stimulation Unit (M.A., F.D.L., G.K.), Department of Behavioural and Clinical Neurology, Santa Lucia Foundation IRCCS; Memory Clinic (M.A.), Department of Systems Medicine, University of Tor Vergata, Rome; Neurology Unit (E.P., S.G., A.B., A.P., B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; Institute of Neuroscience and Physiology (N.J.A.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Wallenberg Centre for Molecular and Translational Medicine (N.J.A.), University of Gothenburg, Mӧlndal, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), United Kingdom; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Clear Water Bay, Hong Kong, China; Neuroradiology Unit (R.G.), University of Brescia, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation (E.T.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine (G.S.), Surgery and Neuroscience, Siena Brain Investigation & Neuromodulation Laboratory, University of Siena, Siena, Italy; Hinda and Arthur Marcus Institute for Aging Research at Hebrew SeniorLife (A.P.-L.); Department of Neurology (A.P.-L.), Harvard MedicalSchool, Boston, MA, USA; and Department of Neuroscience and Rehabilitation (G.K.), University of Ferrara, Italy
| | - Sara Romanella
- From the Precision Neuroscience & Neuromodulation Program (M.A., S.R., G.S., E.S.), Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Non-Invasive Brain Stimulation Unit (M.A., F.D.L., G.K.), Department of Behavioural and Clinical Neurology, Santa Lucia Foundation IRCCS; Memory Clinic (M.A.), Department of Systems Medicine, University of Tor Vergata, Rome; Neurology Unit (E.P., S.G., A.B., A.P., B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; Institute of Neuroscience and Physiology (N.J.A.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Wallenberg Centre for Molecular and Translational Medicine (N.J.A.), University of Gothenburg, Mӧlndal, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), United Kingdom; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Clear Water Bay, Hong Kong, China; Neuroradiology Unit (R.G.), University of Brescia, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation (E.T.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine (G.S.), Surgery and Neuroscience, Siena Brain Investigation & Neuromodulation Laboratory, University of Siena, Siena, Italy; Hinda and Arthur Marcus Institute for Aging Research at Hebrew SeniorLife (A.P.-L.); Department of Neurology (A.P.-L.), Harvard MedicalSchool, Boston, MA, USA; and Department of Neuroscience and Rehabilitation (G.K.), University of Ferrara, Italy
| | - Giulia Sprugnoli
- From the Precision Neuroscience & Neuromodulation Program (M.A., S.R., G.S., E.S.), Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Non-Invasive Brain Stimulation Unit (M.A., F.D.L., G.K.), Department of Behavioural and Clinical Neurology, Santa Lucia Foundation IRCCS; Memory Clinic (M.A.), Department of Systems Medicine, University of Tor Vergata, Rome; Neurology Unit (E.P., S.G., A.B., A.P., B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; Institute of Neuroscience and Physiology (N.J.A.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Wallenberg Centre for Molecular and Translational Medicine (N.J.A.), University of Gothenburg, Mӧlndal, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), United Kingdom; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Clear Water Bay, Hong Kong, China; Neuroradiology Unit (R.G.), University of Brescia, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation (E.T.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine (G.S.), Surgery and Neuroscience, Siena Brain Investigation & Neuromodulation Laboratory, University of Siena, Siena, Italy; Hinda and Arthur Marcus Institute for Aging Research at Hebrew SeniorLife (A.P.-L.); Department of Neurology (A.P.-L.), Harvard MedicalSchool, Boston, MA, USA; and Department of Neuroscience and Rehabilitation (G.K.), University of Ferrara, Italy
| | - Alvaro Pascual-Leone
- From the Precision Neuroscience & Neuromodulation Program (M.A., S.R., G.S., E.S.), Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Non-Invasive Brain Stimulation Unit (M.A., F.D.L., G.K.), Department of Behavioural and Clinical Neurology, Santa Lucia Foundation IRCCS; Memory Clinic (M.A.), Department of Systems Medicine, University of Tor Vergata, Rome; Neurology Unit (E.P., S.G., A.B., A.P., B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; Institute of Neuroscience and Physiology (N.J.A.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Wallenberg Centre for Molecular and Translational Medicine (N.J.A.), University of Gothenburg, Mӧlndal, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), United Kingdom; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Clear Water Bay, Hong Kong, China; Neuroradiology Unit (R.G.), University of Brescia, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation (E.T.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine (G.S.), Surgery and Neuroscience, Siena Brain Investigation & Neuromodulation Laboratory, University of Siena, Siena, Italy; Hinda and Arthur Marcus Institute for Aging Research at Hebrew SeniorLife (A.P.-L.); Department of Neurology (A.P.-L.), Harvard MedicalSchool, Boston, MA, USA; and Department of Neuroscience and Rehabilitation (G.K.), University of Ferrara, Italy
| | - Francesco Di Lorenzo
- From the Precision Neuroscience & Neuromodulation Program (M.A., S.R., G.S., E.S.), Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Non-Invasive Brain Stimulation Unit (M.A., F.D.L., G.K.), Department of Behavioural and Clinical Neurology, Santa Lucia Foundation IRCCS; Memory Clinic (M.A.), Department of Systems Medicine, University of Tor Vergata, Rome; Neurology Unit (E.P., S.G., A.B., A.P., B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; Institute of Neuroscience and Physiology (N.J.A.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Wallenberg Centre for Molecular and Translational Medicine (N.J.A.), University of Gothenburg, Mӧlndal, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), United Kingdom; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Clear Water Bay, Hong Kong, China; Neuroradiology Unit (R.G.), University of Brescia, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation (E.T.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine (G.S.), Surgery and Neuroscience, Siena Brain Investigation & Neuromodulation Laboratory, University of Siena, Siena, Italy; Hinda and Arthur Marcus Institute for Aging Research at Hebrew SeniorLife (A.P.-L.); Department of Neurology (A.P.-L.), Harvard MedicalSchool, Boston, MA, USA; and Department of Neuroscience and Rehabilitation (G.K.), University of Ferrara, Italy
| | - Giacomo Koch
- From the Precision Neuroscience & Neuromodulation Program (M.A., S.R., G.S., E.S.), Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Non-Invasive Brain Stimulation Unit (M.A., F.D.L., G.K.), Department of Behavioural and Clinical Neurology, Santa Lucia Foundation IRCCS; Memory Clinic (M.A.), Department of Systems Medicine, University of Tor Vergata, Rome; Neurology Unit (E.P., S.G., A.B., A.P., B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; Institute of Neuroscience and Physiology (N.J.A.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Wallenberg Centre for Molecular and Translational Medicine (N.J.A.), University of Gothenburg, Mӧlndal, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), United Kingdom; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Clear Water Bay, Hong Kong, China; Neuroradiology Unit (R.G.), University of Brescia, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation (E.T.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine (G.S.), Surgery and Neuroscience, Siena Brain Investigation & Neuromodulation Laboratory, University of Siena, Siena, Italy; Hinda and Arthur Marcus Institute for Aging Research at Hebrew SeniorLife (A.P.-L.); Department of Neurology (A.P.-L.), Harvard MedicalSchool, Boston, MA, USA; and Department of Neuroscience and Rehabilitation (G.K.), University of Ferrara, Italy
| | - Barbara Borroni
- From the Precision Neuroscience & Neuromodulation Program (M.A., S.R., G.S., E.S.), Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Non-Invasive Brain Stimulation Unit (M.A., F.D.L., G.K.), Department of Behavioural and Clinical Neurology, Santa Lucia Foundation IRCCS; Memory Clinic (M.A.), Department of Systems Medicine, University of Tor Vergata, Rome; Neurology Unit (E.P., S.G., A.B., A.P., B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; Institute of Neuroscience and Physiology (N.J.A.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Wallenberg Centre for Molecular and Translational Medicine (N.J.A.), University of Gothenburg, Mӧlndal, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), United Kingdom; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Clear Water Bay, Hong Kong, China; Neuroradiology Unit (R.G.), University of Brescia, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation (E.T.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine (G.S.), Surgery and Neuroscience, Siena Brain Investigation & Neuromodulation Laboratory, University of Siena, Siena, Italy; Hinda and Arthur Marcus Institute for Aging Research at Hebrew SeniorLife (A.P.-L.); Department of Neurology (A.P.-L.), Harvard MedicalSchool, Boston, MA, USA; and Department of Neuroscience and Rehabilitation (G.K.), University of Ferrara, Italy
| | - Emiliano Santarnecchi
- From the Precision Neuroscience & Neuromodulation Program (M.A., S.R., G.S., E.S.), Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Non-Invasive Brain Stimulation Unit (M.A., F.D.L., G.K.), Department of Behavioural and Clinical Neurology, Santa Lucia Foundation IRCCS; Memory Clinic (M.A.), Department of Systems Medicine, University of Tor Vergata, Rome; Neurology Unit (E.P., S.G., A.B., A.P., B.B.), Department of Clinical and Experimental Sciences, University of Brescia, Italy; Institute of Neuroscience and Physiology (N.J.A.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg; Wallenberg Centre for Molecular and Translational Medicine (N.J.A.), University of Gothenburg, Mӧlndal, Sweden; King's College London (N.J.A.), Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation (N.J.A.), United Kingdom; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Clear Water Bay, Hong Kong, China; Neuroradiology Unit (R.G.), University of Brescia, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation (E.T.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine (G.S.), Surgery and Neuroscience, Siena Brain Investigation & Neuromodulation Laboratory, University of Siena, Siena, Italy; Hinda and Arthur Marcus Institute for Aging Research at Hebrew SeniorLife (A.P.-L.); Department of Neurology (A.P.-L.), Harvard MedicalSchool, Boston, MA, USA; and Department of Neuroscience and Rehabilitation (G.K.), University of Ferrara, Italy.
| |
Collapse
|
18
|
Stepanichev M, Aniol V, Lazareva N, Gulyaeva N. Decreased Hippocampal Neurogenesis in Aged Male Wistar Rats Is Not Associated with Memory Acquisition in a Water Maze. Int J Mol Sci 2023; 24:13276. [PMID: 37686083 PMCID: PMC10487931 DOI: 10.3390/ijms241713276] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/12/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Brain aging is associated with a progressive decrease in learning abilities, memory, attention, decision making, and sensory perception. Age-related cognitive disturbances may be related to a decrease in the functional capacities of the hippocampus. This brain region is essential for learning and memory, and the lifelong neurogenesis occurring in the subgranular zone of the dentate gyrus may be a key event mediating the mnemonic functions of the hippocampus. In the present study, we investigated whether age-related changes in hippocampal neurogenesis are associated with learning and memory disturbances. Four- and 24-month-old rats were trained to find a hidden platform in a water maze. Though the older group showed higher latency to search the platform as compared to the younger group, both groups learned the task. However, the density of proliferating (PCNA-positive), differentiating (Dcx-positive), and new neurons (pre-labeled BrdU-positive) was significantly lower in the hippocampus of aged rats as compared to young ones. This inhibition of neurogenesis could be related to increased local production of nitric oxide since the density of neurons expressing neuronal NO-synthase was higher in the aged hippocampus. Thus, we can suggest that an age-related decrease in neurogenesis is not directly associated with place learning in aged rats.
Collapse
Affiliation(s)
- Mikhail Stepanichev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerova Str., 5a, Moscow 117485, Russia; (V.A.); (N.L.); (N.G.)
| | | | | | | |
Collapse
|
19
|
Andrade V, Wong-Guerra M, Cortés N, Pastor G, González A, Calfío C, Guzmán-Martínez L, Navarrete LP, Ramos-Escobar N, Morales I, Santander R, Andrades-Lagos J, Bacho M, Rojo LE, Maccioni RB. Scaling the Andean Shilajit: A Novel Neuroprotective Agent for Alzheimer's Disease. Pharmaceuticals (Basel) 2023; 16:960. [PMID: 37513872 PMCID: PMC10383824 DOI: 10.3390/ph16070960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/12/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder without a cure, despite the enormous number of investigations and therapeutic approaches. AD is a consequence of microglial responses to "damage signals", such as aggregated tau oligomers, which trigger a neuro-inflammatory reaction, promoting the misfolding of cytoskeleton structure. Since AD is the most prevalent cause of dementia in the elderly (>60 years old), new treatments are essential to improve the well-being of affected subjects. The pharmaceutical industry has not developed new drugs with efficacy for controlling AD. In this context, major attention has been given to nutraceuticals and novel bioactive compounds, such as molecules from the Andean Shilajit (AnSh), obtained from the Andes of Chile. Primary cultures of rat hippocampal neurons and mouse neuroblastoma cells were evaluated to examine the functional and neuroprotective role of different AnSh fractions. Our findings show that AnSh fractions increase the number and length of neuronal processes at a differential dose. All fractions were viable in neurons. The AnSh fractions inhibit tau self-aggregation after 10 days of treatment. Finally, we identified two candidate molecules in M3 fractions assayed by UPLC/MS. Our research points to a novel AnSh-derived fraction that is helpful in AD. Intensive work toward elucidation of the molecular mechanisms is being carried out. AnSh is an alternative for AD treatment or as a coadjuvant for an effective treatment.
Collapse
Affiliation(s)
- Víctor Andrade
- Laboratory of Neuroscience and Functional Medicine, International Center for Biomedicine, Faculty of Sciences, University of Chile, Santiago 7800003, Chile
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, 50923 Köln, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, 53127 Bonn, Germany
| | - Maylin Wong-Guerra
- Laboratory of Neuroscience and Functional Medicine, International Center for Biomedicine, Faculty of Sciences, University of Chile, Santiago 7800003, Chile
- Laboratory of Toxicology and Metabolism, Faculty of Chemistry and Biology, University of Santiago of Chile, Santiago 9170022, Chile
| | - Nicole Cortés
- Laboratory of Neuroscience and Functional Medicine, International Center for Biomedicine, Faculty of Sciences, University of Chile, Santiago 7800003, Chile
| | - Gabriela Pastor
- Laboratory of Neuroscience and Functional Medicine, International Center for Biomedicine, Faculty of Sciences, University of Chile, Santiago 7800003, Chile
- Laboratory of Toxicology and Metabolism, Faculty of Chemistry and Biology, University of Santiago of Chile, Santiago 9170022, Chile
| | - Andrea González
- Laboratory of Neuroscience and Functional Medicine, International Center for Biomedicine, Faculty of Sciences, University of Chile, Santiago 7800003, Chile
| | - Camila Calfío
- Laboratory of Neuroscience and Functional Medicine, International Center for Biomedicine, Faculty of Sciences, University of Chile, Santiago 7800003, Chile
| | - Leonardo Guzmán-Martínez
- Laboratory of Neuroscience and Functional Medicine, International Center for Biomedicine, Faculty of Sciences, University of Chile, Santiago 7800003, Chile
| | - Leonardo P Navarrete
- Laboratory of Neuroscience and Functional Medicine, International Center for Biomedicine, Faculty of Sciences, University of Chile, Santiago 7800003, Chile
- Biochemistry School, Faculty of Health Sciences, Andres Bello University, Santiago 8370035, Chile
| | - Nicolas Ramos-Escobar
- Laboratory of Neuroscience and Functional Medicine, International Center for Biomedicine, Faculty of Sciences, University of Chile, Santiago 7800003, Chile
| | - Inelia Morales
- Laboratory of Neuroscience and Functional Medicine, International Center for Biomedicine, Faculty of Sciences, University of Chile, Santiago 7800003, Chile
| | - Rocío Santander
- Laboratory of Kinetics and Photochemistry, Faculty of Chemistry and Biology, University of Santiago of Chile, Santiago 9170022, Chile
| | - Juan Andrades-Lagos
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510157, Chile
- Drug Development Laboratory, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8380492, Chile
| | - Mitchell Bacho
- Organic and Organometallic Synthesis Laboratory, Faculty of Chemistry, Andrés Bello University, Santiago 8370186, Chile
- Laboratory of Natural Resources, Faculty of Sciences, University of Chile, Santiago 7750000, Chile
| | - Leonel E Rojo
- Laboratory of Toxicology and Metabolism, Faculty of Chemistry and Biology, University of Santiago of Chile, Santiago 9170022, Chile
| | - Ricardo Benjamín Maccioni
- Laboratory of Neuroscience and Functional Medicine, International Center for Biomedicine, Faculty of Sciences, University of Chile, Santiago 7800003, Chile
| |
Collapse
|
20
|
Arce B, Grañana N. [Cognitive deficits with chemotherapy in women with breast cancer: a bibliographic revisión]. REVISTA DE LA FACULTAD DE CIENCIAS MÉDICAS 2023; 80:126-133. [PMID: 37402295 PMCID: PMC10443418 DOI: 10.31053/1853.0605.v80.n2.40533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/07/2023] [Indexed: 07/06/2023] Open
Abstract
Background Patients with breast cancer may experience cognitive difficulties from chemotherapy. This alteration is called Chemoinduced Cognitive Impairment, also known as Chemobrain or Chemofog. Objective To identify the cognitive profile and the characteristics of the neuropsychological assessment in this population. Method: PubMed, SpringerLink and SciELO databases were revised. Articles from 1994 to September 2021 were selected. Keywords related to the study topic were used. Results Chemotherapy can cause cognitive impairment between 15 and 50% of women. This disturbance may be from multiple aetiologies and can be associated with biological factors and functional and/or structural changes of the CNS. Sociodemographic, clinical and psychological factors should be considered as modulating variables. It manifests mainly with memory problems, executive function, attention and processing speed impairment. It can be measured through neuropsychological evaluation instruments. Discussion and conclusion We suggest that chemo-induced cognitive impairment should be included to the informed consent. Further development of longitudinal studies complemented with neuroimages that allow us to advance in the knowledge of this problem is recommended. A neuropsychological protocol is proposed, which includes screening tests, clinical scales, specific cognitive tests and quality of life questionnaires, within the recommendations of the International Cognition and Cancer Task Force.
Collapse
|
21
|
Alam MR, Singh S. Neuromodulation in Parkinson's disease targeting opioid and cannabinoid receptors, understanding the role of NLRP3 pathway: a novel therapeutic approach. Inflammopharmacology 2023:10.1007/s10787-023-01259-0. [PMID: 37318694 DOI: 10.1007/s10787-023-01259-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/26/2023] [Indexed: 06/16/2023]
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta, resulting in motor and non-motor symptoms. Although levodopa is the primary medication for PD, its long-term use is associated with complications such as dyskinesia and drug resistance, necessitating novel therapeutic approaches. Recent research has highlighted the potential of targeting opioid and cannabinoid receptors as innovative strategies for PD treatment. Modulating opioid transmission, particularly through activating µ (MOR) and δ (DOR) receptors while inhibiting κ (KOR) receptors, shows promise in preventing motor complications and reducing L-DOPA-induced dyskinesia. Opioids also possess neuroprotective properties and play a role in neuroprotection and seizure control. Similar to this, endocannabinoid signalling via CB1 and CB2 receptors influences the basal ganglia and may contribute to PD pathophysiology, making it a potential therapeutic target. In addition to opioid and cannabinoid receptor targeting, the NLRP3 pathway, implicated in neuroinflammation and neurodegeneration, emerges as another potential therapeutic avenue for PD. Recent studies suggest that targeting this pathway holds promise as a therapeutic strategy for PD management. This comprehensive review focuses on neuromodulation and novel therapeutic approaches for PD, specifically highlighting the targeting of opioid and cannabinoid receptors and the NLRP3 pathway. A better understanding of these mechanisms has the potential to enhance the quality of life for PD patients.
Collapse
Affiliation(s)
- Md Reyaz Alam
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
22
|
Alonso Bellido IM, Posada-Pérez M, Hernández-Rasco F, Vázquez-Reyes S, Cabanillas M, Herrera AJ, Bachiller S, Soldán-Hidalgo J, Espinosa-Oliva AM, Joseph B, de Pablos RM, Venero JL, Ruiz R. Microglial Caspase-3 is essential for modulating hippocampal neurogenesis. Brain Behav Immun 2023:S0889-1591(23)00157-5. [PMID: 37327833 DOI: 10.1016/j.bbi.2023.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 06/07/2023] [Accepted: 06/10/2023] [Indexed: 06/18/2023] Open
Abstract
Adult hippocampal neurogenesis (AHN) is a process involved in numerous neurodegenerative diseases. Many researchers have described microglia as a key component in regulating the formation and migration of new neurons along the rostral migratory stream. Caspase-3 is a cysteine-aspartate-protease classically considered as one of the main effector caspases in the cell death program process. In addition to this classical function, we have identified the role of this protein as a modulator of microglial function; however, its action on neurogenic processes is unknown. The aim of the present study is to identify the role of Caspase-3 in neurogenesis-related microglial functions. To address this study, Caspase-3 conditional knockout mice in the microglia cell line were used. Using this tool, we wanted to elucidate the role of this protein in microglial function in the hippocampus, the main region in which adult neurogenesis takes place. After the reduction of Caspase-3 in microglia, mutant mice showed a reduction of microglia in the hippocampus, especially in the dentate gyrus region, a region inherently associated to neurogenesis. In addition, we found a reduction in doublecortin-positive neurons in conditional Caspase-3 knockout mice, which corresponds to a reduction in neurogenic neurons. Furthermore, using high-resolution image analysis, we also observed a reduction in the phagocytic capacity of microglia lacking Caspase-3. Behavioral analysis using object recognition and Y-maze tests showed altered memory and learning in the absence of Caspase-3. Finally, we identified specific microglia located specifically in neurogenic niche positive for Galectin 3 which colocalized with Cleaved-Caspase-3 in control mice. Taken together, these results showed the essential role of Caspase-3 in microglial function and highlight the relevant role of this specific microglial phenotype in the maintenance of AHN in the hippocampus.
Collapse
Affiliation(s)
- Isabel M Alonso Bellido
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - Mercedes Posada-Pérez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden
| | - Francisco Hernández-Rasco
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - Sandra Vázquez-Reyes
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - María Cabanillas
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - Antonio J Herrera
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - Sara Bachiller
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Laboratory of Immunovirology, Virgen del Rocío University Hospital, Seville, Spain; Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Jesús Soldán-Hidalgo
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - Ana M Espinosa-Oliva
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - Bertrand Joseph
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden
| | - Rocío M de Pablos
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - José L Venero
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - Rocío Ruiz
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain.
| |
Collapse
|
23
|
Abbate C. The Adult Neurogenesis Theory of Alzheimer's Disease. J Alzheimers Dis 2023:JAD221279. [PMID: 37182879 DOI: 10.3233/jad-221279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Alzheimer's disease starts in neural stem cells (NSCs) in the niches of adult neurogenesis. All primary factors responsible for pathological tau hyperphosphorylation are inherent to adult neurogenesis and migration. However, when amyloid pathology is present, it strongly amplifies tau pathogenesis. Indeed, the progressive accumulation of extracellular amyloid-β deposits in the brain triggers a state of chronic inflammation by microglia. Microglial activation has a significant pro-neurogenic effect that fosters the process of adult neurogenesis and supports neuronal migration. Unfortunately, this "reactive" pro-neurogenic activity ultimately perturbs homeostatic equilibrium in the niches of adult neurogenesis by amplifying tau pathogenesis in AD. This scenario involves NSCs in the subgranular zone of the hippocampal dentate gyrus in late-onset AD (LOAD) and NSCs in the ventricular-subventricular zone along the lateral ventricles in early-onset AD (EOAD), including familial AD (FAD). Neuroblasts carrying the initial seed of tau pathology travel throughout the brain via neuronal migration driven by complex signals and convey the disease from the niches of adult neurogenesis to near (LOAD) or distant (EOAD) brain regions. In these locations, or in close proximity, a focus of degeneration begins to develop. Then, tau pathology spreads from the initial foci to large neuronal networks along neural connections through neuron-to-neuron transmission.
Collapse
Affiliation(s)
- Carlo Abbate
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| |
Collapse
|
24
|
Zhang B, Zhang Y, Zuo Z, Xiong G, Luo H, Song B, Zhao L, Zhou Z, Chang X. Paraquat-induced neurogenesis abnormalities via Drp1-mediated mitochondrial fission. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 257:114939. [PMID: 37087969 DOI: 10.1016/j.ecoenv.2023.114939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 05/03/2023]
Abstract
Neurogenesis is a fundamental process in the development and plasticity of the nervous system, and its regulation is tightly linked to mitochondrial dynamics. Imbalanced mitochondrial dynamics can result in oxidative stress, which has been implicated in various neurological disorders. Paraquat (PQ), a commonly used agricultural chemical known to be neurotoxic, induces oxidative stress that can lead to mitochondrial fragmentation. In this study, we investigated the effects of PQ on neurogenesis in primary murine neural progenitor cells (mNPCs) isolated from neonatal C57BL/6 mice. We treated the mNPCs with 0-40 μM PQ for 24 h and observed that PQ inhibited their proliferation, migration, and differentiation into neurons in a concentration-dependent manner. Moreover, PQ induced excessive mitochondrial fragmentation and upregulated the expression of Drp-1, p-Drp1, and Fis-1, while downregulating the expression of Mfn2 and Opa1. To confirm our findings, we used Mdivi-1, an inhibitor of mitochondrial fission, which reversed the adverse effects of PQ on neurogenesis, particularly differentiation into neurons and migration of mNPCs. Additionally, we found that Mito-TEMPO, a mitochondria-targeted antioxidant, ameliorated excessive mitochondrial fragmentation caused by PQ. Our study suggests that PQ exposure impairs neurogenesis by inducing excessive mitochondrial fission and abnormal mitochondrial fragmentation via oxidative stress. These findings identify mitochondrial fission as a potential therapeutic target for PQ-induced neurotoxicity. Further research is needed to elucidate the underlying mechanisms of mitochondrial dynamics and neurogenesis in the context of oxidative stress-induced neurological disorders.
Collapse
Affiliation(s)
- Bing Zhang
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai 200032, China
| | - Yuwei Zhang
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai 200032, China
| | - Zhenzi Zuo
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai 200032, China
| | - Guiya Xiong
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai 200032, China
| | - Huan Luo
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai 200032, China
| | - Bo Song
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai 200032, China
| | - Lina Zhao
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai 200032, China
| | - Zhijun Zhou
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai 200032, China
| | - Xiuli Chang
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai 200032, China.
| |
Collapse
|
25
|
Fan X, Zhao Z, Huang Z, Wu M, Wang D, Xiao J. Mineralocorticoid receptor agonist aldosterone rescues hippocampal neural stem cell proliferation defects and improves postoperative cognitive function in aged mice. World J Biol Psychiatry 2023; 24:149-161. [PMID: 35615969 DOI: 10.1080/15622975.2022.2082524] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES Hippocampal neurogenesis is closely related to learning and memory, and hippocampal neurogenesis disorders are involved in the development of many neurodegenerative diseases. Mineralocorticoid receptor (MR) plays a vital role in regulating stress response, neuroendocrine and cognitive functions, and is involved in regulating the integrity and stability of neural networks. However, the potential role of MR in the pathogenesis of postoperative cognitive dysfunction (POCD) is unclear. Therefore, this study evaluated the effect and mechanism of MR activation on postoperative hippocampal neurogenesis and cognitive function in aged mice. METHODS 18-month-old male Kunming mice were randomly divided into Control group (C group), Surgery group (S group), Surgery+ Aldosterone group (S+Aldo group), Surgery + Wortmannin group (S+Wort group), Surgery + Aldosterone + Wortmannin group (S+Aldo+Wort group). Laparotomy was used to establish an animal model of postoperative cognitive dysfunction. After surgery, mice were intraperitoneally injected with aldosterone (100 ug/kg,150 ug/kg,200 ug/kg) and / or wortmannin (1 mg/kg); One day before the sacrifice, mice were injected intraperitoneally with BrdU (100 mg / kg / time, 3 times in total). Mice were subjected to Morris water maze and field tests at 1, 3, 7, and 14 days after surgery. Immunofluorescence was used to detect the number of BrdU +, Nestin +, BrdU/Nestin + positive cells in the hippocampal dentate gyrus of mice at 1, 3, 7 and 14 days after surgery. Western-blot was used to detect PI3K/Akt/GSK-3β signaling pathway related proteins Akt, p-Akt, GSK-3β, P-GSK-3β expression. RESULTS Stress impairs the performance of aged mice in water maze and open field tests, reduces the number of BrdU/Nestin+ cells in the hippocampal dentate gyrus, and inhibits the phosphorylation of Akt and GSK-3β proteins in the hippocampus. Aldosterone treatment promotes P-Akt, P-GSK-3β protein expression and hippocampal neural stem cell proliferation, and improves postoperative cognitive dysfunction. However, wortmannin treatment significantly reversed these effects of aldosterone. CONCLUSIONS The mineralocorticoid receptor agonist aldosterone promotes the proliferation of hippocampal neural stem cells and improves cognitive dysfunction in aged mice after surgery, and the mechanism may be related to activation of PI3K/Akt/GSK-3β signaling.
Collapse
Affiliation(s)
- Xuhong Fan
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Zhenyu Zhao
- Department of Anesthesiology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Zixia Huang
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Mingyue Wu
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Deming Wang
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Ji Xiao
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China
| |
Collapse
|
26
|
Shen W, Jiang N, Zhou W. What can traditional Chinese medicine do for adult neurogenesis? Front Neurosci 2023; 17:1158228. [PMID: 37123359 PMCID: PMC10130459 DOI: 10.3389/fnins.2023.1158228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/13/2023] [Indexed: 05/02/2023] Open
Abstract
Adult neurogenesis plays a crucial role in cognitive function and mood regulation, while aberrant adult neurogenesis contributes to various neurological and psychiatric diseases. With a better understanding of the significance of adult neurogenesis, the demand for improving adult neurogenesis is increasing. More and more research has shown that traditional Chinese medicine (TCM), including TCM prescriptions (TCMPs), Chinese herbal medicine, and bioactive components, has unique advantages in treating neurological and psychiatric diseases by regulating adult neurogenesis at various stages, including proliferation, differentiation, and maturation. In this review, we summarize the progress of TCM in improving adult neurogenesis and the key possible mechanisms by which TCM may benefit it. Finally, we suggest the possible strategies of TCM to improve adult neurogenesis in the treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Wei Shen
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Ning Jiang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
- *Correspondence: Ning Jiang, ; Wenxia Zhou,
| | - Wenxia Zhou
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
- *Correspondence: Ning Jiang, ; Wenxia Zhou,
| |
Collapse
|
27
|
Méresse S, Larrigaldie V, Oummadi A, de Concini V, Morisset-Lopez S, Reverchon F, Menuet A, Montécot-Dubourg C, Mortaud S. β-N-Methyl-Amino-L-Alanine cyanotoxin promotes modification of undifferentiated cells population and disrupts the inflammatory status in primary cultures of neural stem cells. Toxicology 2022; 482:153358. [DOI: 10.1016/j.tox.2022.153358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/06/2022] [Accepted: 10/24/2022] [Indexed: 11/07/2022]
|
28
|
Ruiz-Contreras HA, Santamaría A, Arellano-Mendoza MG, Sánchez-Chapul L, Robles-Bañuelos B, Rangel-López E. Modulatory Activity of the Endocannabinoid System in the Development and Proliferation of Cells in the CNS. Neurotox Res 2022; 40:1690-1706. [PMID: 36522511 DOI: 10.1007/s12640-022-00592-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/21/2022] [Accepted: 10/08/2022] [Indexed: 12/23/2022]
Abstract
The Endocannabinoid System (ECS, also known as Endocannabinoidome) plays a key role in the function of the Central Nervous System, though the participation of this system on the early development - specifically in neuroprotection and proliferation of nerve cells - has been poorly studied. Here, we collect and describe evidence regarding how cannabinoid receptors CB1R and CB2R regulate several cell markers related to proliferation. While CB1R participates in the modulation of neuronal and glial proliferation, CB2R is involved in the proliferation of glial cells. The endocannabinoids anandamide (AEA) and 2-arachidonoylglycerol (2-AG) exert significant effects on nerve cell proliferation. AEA generated during embryogenesis induces major effects on the differentiation of neuronal progenitor cells, whereas 2-AG participates in modulating cell migration events rather than affecting the neural proliferation rate. However, although the ECS has been demonstrated to participate in neuroprotection, more characterization on its role in neuronal and glial proliferation and differentiation is needed, especially in brain areas with recognized high neurogenesis rates. This has encouraged scientists to elucidate and propose specific mechanisms related with these cell proliferation mechanisms to better understand some neurodegenerative disorders such as Parkinson, Huntington and Alzheimer diseases, in which neuronal loss and poor neurogenesis are crucial factors for their onset and progression. In this review, we collect and present recent evidence published pointing to an active role of the ECS in the development and proliferation of nerve cells.
Collapse
Affiliation(s)
- Hipolito A Ruiz-Contreras
- Maestría en Ciencias en Farmacología, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores/Laboratorio de Neurofarmacología Molecular Y Nanotecnología, Instituto Nacional de Neurología Y Neurocirugía Manuel Velasco Suárez, Insurgentes Sur 3877, 14269, Mexico City, Mexico.
| | - Mónica G Arellano-Mendoza
- Laboratorio de Investigación en Enfermedades Crónico Degenerativas, Sección de Estudios de Posgrado E Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Laura Sánchez-Chapul
- Laboratorio de Enfermedades Neuromusculares, División de Neurociencias Clínicas, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Benjamín Robles-Bañuelos
- Laboratorio de Aminoácidos Excitadores/Laboratorio de Neurofarmacología Molecular Y Nanotecnología, Instituto Nacional de Neurología Y Neurocirugía Manuel Velasco Suárez, Insurgentes Sur 3877, 14269, Mexico City, Mexico
| | - Edgar Rangel-López
- Laboratorio de Aminoácidos Excitadores/Laboratorio de Neurofarmacología Molecular Y Nanotecnología, Instituto Nacional de Neurología Y Neurocirugía Manuel Velasco Suárez, Insurgentes Sur 3877, 14269, Mexico City, Mexico.
| |
Collapse
|
29
|
Hossain MM, Belkadi A, Zhou X, DiCicco-Bloom E. Exposure to deltamethrin at the NOAEL causes ER stress and disruption of hippocampal neurogenesis in adult mice. Neurotoxicology 2022; 93:233-243. [DOI: 10.1016/j.neuro.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/29/2022] [Accepted: 10/07/2022] [Indexed: 11/15/2022]
|
30
|
Downey J, Lam JC, Li VO, Gozes I. Somatic Mutations and Alzheimer’s Disease. J Alzheimers Dis 2022; 90:475-493. [DOI: 10.3233/jad-220643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alzheimer’s disease (AD) represents a global health challenge, with an estimated 55 million people suffering from the non-curable disease across the world. While amyloid-β plaques and tau neurofibrillary tangles in the brain define AD proteinopathy, it has become evident that diverse coding and non-coding regions of the genome may significantly contribute to AD neurodegeneration. The diversity of factors associated with AD pathogenesis, coupled with age-associated damage, suggests that a series of triggering events may be required to initiate AD. Since somatic mutations accumulate with aging, and aging is a major risk factor for AD, there is a great potential for somatic mutational events to drive disease. Indeed, recent data from the Gozes team/laboratories as well as other leading laboratories correlated the accumulation of somatic brain mutations with the progression of tauopathy. In this review, we lay the current perspectives on the principal genetic factors associated with AD and the potential causes, highlighting the contribution of somatic mutations to the pathogenesis of late onset Alzheimer’s disease. The roles that artificial intelligence and big data can play in accelerating the progress of causal somatic mutation markers/biomarkers identification, and the associated drug discovery/repurposing, have been highlighted for future AD and other neurodegenerative studies, with the aim to bring hope for the vulnerable aging population.
Collapse
Affiliation(s)
- Jocelyn Downey
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| | - Jacqueline C.K. Lam
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
- Department of Computer Science and Technology, University of Cambridge, UK
| | - Victor O.K. Li
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| | - Illana Gozes
- The Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
31
|
Huang M, Bargues-Carot A, Riaz Z, Wickham H, Zenitsky G, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Impact of Environmental Risk Factors on Mitochondrial Dysfunction, Neuroinflammation, Protein Misfolding, and Oxidative Stress in the Etiopathogenesis of Parkinson's Disease. Int J Mol Sci 2022; 23:10808. [PMID: 36142718 PMCID: PMC9505762 DOI: 10.3390/ijms231810808] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/25/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
As a prevalent progressive neurodegenerative disorder, Parkinson's disease (PD) is characterized by the neuropathological hallmark of the loss of nigrostriatal dopaminergic (DAergic) innervation and the appearance of Lewy bodies with aggregated α-synuclein. Although several familial forms of PD have been reported to be associated with several gene variants, most cases in nature are sporadic, triggered by a complex interplay of genetic and environmental risk factors. Numerous epidemiological studies during the past two decades have shown positive associations between PD and several environmental factors, including exposure to neurotoxic pesticides/herbicides and heavy metals as well as traumatic brain injury. Other environmental factors that have been implicated as potential risk factors for PD include industrial chemicals, wood pulp mills, farming, well-water consumption, and rural residence. In this review, we summarize the environmental toxicology of PD with the focus on the elaboration of chemical toxicity and the underlying pathogenic mechanisms associated with exposure to several neurotoxic chemicals, specifically 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), rotenone, paraquat (PQ), dichloro-diphenyl-trichloroethane (DDT), dieldrin, manganese (Mn), and vanadium (V). Our overview of the current findings from cellular, animal, and human studies of PD provides information for possible intervention strategies aimed at halting the initiation and exacerbation of environmentally linked PD.
Collapse
Affiliation(s)
- Minhong Huang
- Department of Biomedical Sciences, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Alejandra Bargues-Carot
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Zainab Riaz
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Hannah Wickham
- Department of Biomedical Sciences, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Gary Zenitsky
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Huajun Jin
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Vellareddy Anantharam
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Arthi Kanthasamy
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Anumantha G. Kanthasamy
- Department of Biomedical Sciences, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| |
Collapse
|
32
|
Examination of Longitudinal Alterations in Alzheimer’s Disease-Related Neurogenesis in an APP/PS1 Transgenic Mouse Model, and the Effects of P33, a Putative Neuroprotective Agent Thereon. Int J Mol Sci 2022; 23:ijms231810364. [PMID: 36142277 PMCID: PMC9499399 DOI: 10.3390/ijms231810364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/05/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Neurogenesis plays a crucial role in cognitive processes. During aging and in Alzheimer’s disease (AD), altered neurogenesis and neuroinflammation are evident both in C57BL/6J, APPSwe/PS1dE9 (Tg) mice and humans. AD pathology may slow down upon drug treatment, for example, in a previous study of our group P33, a putative neuroprotective agent was found to exert advantageous effects on the elevated levels of APP, Aβ, and neuroinflammation. In the present study, we aimed to examine longitudinal alterations in neurogenesis, neuroinflammation and AD pathology in a transgenic (Tg) mouse model, and assessed the putative beneficial effects of long-term P33 treatment on AD-specific neurological alterations. Hippocampal cell proliferation and differentiation were significantly reduced between 8 and 12 months of age. Regarding neuroinflammation, significantly elevated astrogliosis and microglial activation were observed in 6- to 7-month-old Tg animals. The amounts of the molecules involved in the amyloidogenic pathway were altered from 4 months of age in Tg animals. P33-treatment led to significantly increased neurogenesis in 9-month-old animals. Our data support the hypothesis that altered neurogenesis may be a consequence of AD pathology. Based on our findings in the transgenic animal model, early pharmacological treatment before the manifestation of AD symptoms might ameliorate neurological decline.
Collapse
|
33
|
Britton R, Liu AT, Rege SV, Adams JM, Akrapongpisak L, Le D, Alcantara-Lee R, Estrada RA, Ray R, Ahadi S, Gallager I, Yang CF, Minami SS, Braithwaite SP, Czirr E, Campbell MK. Molecular and histological correlates of cognitive decline across age in male C57BL/6J mice. Brain Behav 2022; 12:e2736. [PMID: 35971662 PMCID: PMC9480918 DOI: 10.1002/brb3.2736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 07/01/2022] [Accepted: 07/20/2022] [Indexed: 11/09/2022] Open
Abstract
INTRODUCTION Increasing age is the number one risk factor for developing cognitive decline and neurodegenerative disease. Aged humans and mice exhibit numerous molecular changes that contribute to a decline in cognitive function and increased risk of developing age-associated diseases. Here, we characterize multiple age-associated changes in male C57BL/6J mice to understand the translational utility of mouse aging. METHODS Male C57BL/6J mice from various ages between 2 and 24 months of age were used to assess behavioral, as well as, histological and molecular changes across three modalities: neuronal, microgliosis/neuroinflammation, and the neurovascular unit (NVU). Additionally, a cohort of 4- and 22-month-old mice was used to assess blood-brain barrier (BBB) breakdown. Mice in this cohort were treated with a high, acute dose of lipopolysaccharide (LPS, 10 mg/kg) or saline control 6 h prior to sacrifice followed by tail vein injection of 0.4 kDa sodium fluorescein (100 mg/kg) 2 h later. RESULTS Aged mice showed a decline in cognitive and motor abilities alongside decreased neurogenesis, proliferation, and synapse density. Further, neuroinflammation and circulating proinflammatory cytokines were increased in aged mice. Additionally, we found changes at the BBB, including increased T cell infiltration in multiple brain regions and an exacerbation in BBB leakiness following chemical insult with age. There were also a number of readouts that were unchanged with age and have limited utility as markers of aging in male C57BL/6J mice. CONCLUSIONS Here we propose that these changes may be used as molecular and histological readouts that correspond to aging-related behavioral decline. These comprehensive findings, in the context of the published literature, are an important resource toward deepening our understanding of normal aging and provide an important tool for studying aging in mice.
Collapse
Affiliation(s)
| | - Angela T Liu
- Alkahest, Inc., San Carlos, California, USA.,Coda Biotherapeutics, South San Francisco, California, USA
| | | | | | - Lily Akrapongpisak
- Alkahest, Inc., San Carlos, California, USA.,University of Queensland, Herston, Queensland, Australia
| | - David Le
- Alkahest, Inc., San Carlos, California, USA.,Fountain Therapeutics, South San Francisco, California, USA
| | | | | | - Rebecca Ray
- Alkahest, Inc., San Carlos, California, USA.,202 Chives Way, Walnut Creek, California, USA
| | - Sara Ahadi
- Alkahest, Inc., San Carlos, California, USA
| | | | | | | | | | - Eva Czirr
- Alkahest, Inc., San Carlos, California, USA.,Confluence Therapeutics, South San Francisco, California, USA
| | | |
Collapse
|
34
|
Shahid SS, Wen Q, Risacher SL, Farlow MR, Unverzagt FW, Apostolova LG, Foroud TM, Zetterberg H, Blennow K, Saykin AJ, Wu YC. Hippocampal-subfield microstructures and their relation to plasma biomarkers in Alzheimer's disease. Brain 2022; 145:2149-2160. [PMID: 35411392 PMCID: PMC9630875 DOI: 10.1093/brain/awac138] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 11/14/2022] Open
Abstract
Hippocampal subfields exhibit differential vulnerabilities to Alzheimer's disease-associated pathology including abnormal accumulation of amyloid-β deposition and neurofibrillary tangles. These pathological processes extensively impact on the structural and functional interconnectivities of the subfields and may explain the association between hippocampal dysfunction and cognitive deficits. In this study, we investigated the degree of alterations in the microstructure of hippocampal subfields across the clinical continuum of Alzheimer's disease. We applied a grey matter-specific multi-compartment diffusion model (Cortical-Neurite orientation dispersion and density imaging) to understand the differential effects of Alzheimer's disease pathology on the hippocampal subfield microstructure. A total of 119 participants were included in this cross-sectional study. Participants were stratified into three categories, cognitively normal (n = 47), mild cognitive impairment (n = 52), and Alzheimer's disease (n = 19). Diffusion MRI, plasma biomarkers and neuropsychological test scores were used to determine the association between the microstructural integrity and Alzheimer's disease-associated molecular indicators and cognition. For Alzheimer's disease-related plasma biomarkers, we studied amyloid-β, total tau and neurofilament light; for Alzheimer's disease-related neuropsychological tests, we included the Trail Making Test, Rey Auditory Verbal Learning Test, Digit Span and Montreal Cognitive Assessment. Comparisons between cognitively normal subjects and those with mild cognitive impairment showed significant microstructural alterations in the hippocampal cornu ammonis (CA) 4 and dentate gyrus region, whereas CA 1-3 was the most sensitive region for the later stages in the Alzheimer's disease clinical continuum. Among imaging metrics for microstructures, the volume fraction of isotropic diffusion for interstitial free water demonstrated the largest effect size in between-group comparisons. Regarding the plasma biomarkers, neurofilament light appeared to be the most sensitive biomarker for associations with microstructural imaging findings in CA4-dentate gyrus. CA 1-3 was the subfield which had stronger correlations between cognitive performance and microstructural metrics. Particularly, poor performance on the Rey Auditory Verbal Learning Test and Montreal Cognitive Assessment was associated with decreased intracellular volume fraction. Overall, our findings support the value of tissue-specific microstructural imaging for providing pathologically relevant information manifesting in the plasma biomarkers and neuropsychological outcomes across various stages of Alzheimer's disease.
Collapse
Affiliation(s)
- Syed Salman Shahid
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Qiuting Wen
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shannon L Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Martin R Farlow
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Frederick W Unverzagt
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Liana G Apostolova
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tatiana M Foroud
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yu Chien Wu
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
35
|
Coelho P, Fão L, Mota S, Rego AC. Mitochondrial function and dynamics in neural stem cells and neurogenesis: Implications for neurodegenerative diseases. Ageing Res Rev 2022; 80:101667. [PMID: 35714855 DOI: 10.1016/j.arr.2022.101667] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 05/21/2022] [Accepted: 06/09/2022] [Indexed: 11/28/2022]
Abstract
Mitochondria have been largely described as the powerhouse of the cell and recent findings demonstrate that this organelle is fundamental for neurogenesis. The mechanisms underlying neural stem cells (NSCs) maintenance and differentiation are highly regulated by both intrinsic and extrinsic factors. Mitochondrial-mediated switch from glycolysis to oxidative phosphorylation, accompanied by mitochondrial remodeling and dynamics are vital to NSCs fate. Deregulation of mitochondrial proteins, mitochondrial DNA, function, fission/fusion and metabolism underly several neurodegenerative diseases; data show that these impairments are already present in early developmental stages and NSC fate decisions. However, little is known about mitochondrial role in neurogenesis. In this Review, we describe the recent evidence covering mitochondrial role in neurogenesis, its impact in selected neurodegenerative diseases, for which aging is the major risk factor, and the recent advances in stem cell-based therapies that may alleviate neurodegenerative disorders-related neuronal deregulation through improvement of mitochondrial function and dynamics.
Collapse
Affiliation(s)
- Patrícia Coelho
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra Polo 1, Coimbra, Portugal.
| | - Lígia Fão
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra Polo 1, Coimbra, Portugal; FMUC- Faculty of Medicine, University of Coimbra Polo 3, Coimbra, Portugal.
| | - Sandra Mota
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra Polo 1, Coimbra, Portugal; III, Institute of Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
| | - A Cristina Rego
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra Polo 1, Coimbra, Portugal; FMUC- Faculty of Medicine, University of Coimbra Polo 3, Coimbra, Portugal.
| |
Collapse
|
36
|
Ozgen S, Krigman J, Zhang R, Sun N. Significance of mitochondrial activity in neurogenesis and neurodegenerative diseases. Neural Regen Res 2022; 17:741-747. [PMID: 34472459 PMCID: PMC8530128 DOI: 10.4103/1673-5374.322429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/13/2021] [Accepted: 03/13/2021] [Indexed: 12/11/2022] Open
Abstract
Mitochondria play a multidimensional role in the function and the vitality of the neurological system. From the generation of neural stem cells to the maintenance of neurons and their ultimate demise, mitochondria play a critical role in regulating our neural pathways' homeostasis, a task that is critical to our cognitive health and neurological well-being. Mitochondria provide energy via oxidative phosphorylation for the neurotransmission and generation of an action potential along the neuron's axon. This paper will first review and examine the molecular subtleties of the mitochondria's role in neurogenesis and neuron vitality, as well as outlining the impact of defective mitochondria in neural aging. The authors will then summarize neurodegenerative diseases related to either neurogenesis or homeostatic dysfunction. Because of the significant detriment neurodegenerative diseases have on the quality of life, it is essential to understand their etiology and ongoing molecular mechanics. The mitochondrial role in neurogenesis and neuron vitality is essential. Dissecting and understanding this organelle's role in the genesis and homeostasis of neurons should assist in finding pharmaceutical targets for neurodegenerative diseases.
Collapse
Affiliation(s)
- Serra Ozgen
- Departments of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- College of Medicine, Graduate Research in the Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Judith Krigman
- Departments of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ruohan Zhang
- Departments of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- College of Pharmacy, Department of Graduate Research, The Ohio State University, Columbus, OH, USA
| | - Nuo Sun
- Departments of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
37
|
Jo T, Nho K, Bice P, Saykin AJ. Deep learning-based identification of genetic variants: application to Alzheimer's disease classification. Brief Bioinform 2022; 23:bbac022. [PMID: 35183061 PMCID: PMC8921609 DOI: 10.1093/bib/bbac022] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 01/29/2023] Open
Abstract
Deep learning is a promising tool that uses nonlinear transformations to extract features from high-dimensional data. Deep learning is challenging in genome-wide association studies (GWAS) with high-dimensional genomic data. Here we propose a novel three-step approach (SWAT-CNN) for identification of genetic variants using deep learning to identify phenotype-related single nucleotide polymorphisms (SNPs) that can be applied to develop accurate disease classification models. In the first step, we divided the whole genome into nonoverlapping fragments of an optimal size and then ran convolutional neural network (CNN) on each fragment to select phenotype-associated fragments. In the second step, using a Sliding Window Association Test (SWAT), we ran CNN on the selected fragments to calculate phenotype influence scores (PIS) and identify phenotype-associated SNPs based on PIS. In the third step, we ran CNN on all identified SNPs to develop a classification model. We tested our approach using GWAS data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) including (N = 981; cognitively normal older adults (CN) = 650 and AD = 331). Our approach identified the well-known APOE region as the most significant genetic locus for AD. Our classification model achieved an area under the curve (AUC) of 0.82, which was compatible with traditional machine learning approaches, random forest and XGBoost. SWAT-CNN, a novel deep learning-based genome-wide approach, identified AD-associated SNPs and a classification model for AD and may hold promise for a range of biomedical applications.
Collapse
Affiliation(s)
- Taeho Jo
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Network Science Institute, Bloomington, IN, USA
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Network Science Institute, Bloomington, IN, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Paula Bice
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Network Science Institute, Bloomington, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | |
Collapse
|
38
|
Karvelas N, Bennett S, Politis G, Kouris NI, Kole C. Advances in stem cell therapy in Alzheimer's disease: a comprehensive clinical trial review. Stem Cell Investig 2022; 9:2. [PMID: 35280344 PMCID: PMC8898169 DOI: 10.21037/sci-2021-063] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/27/2022] [Indexed: 07/30/2023]
Abstract
Alzheimer's disease (AD) is the most common type of dementia responsible for more than 121,499 deaths from AD in 2019 making AD the sixth-leading cause in the United States. AD is a progressive neurodegenerative disorder characterized by decline of memory, behavioral impairments that affects a person's ability to function independently ultimately leading to death. The current pressing need for a treatment for (AD) and advances in the field of cell therapy, has rendered stem cell therapeutics a promising field of research. Despite advancements in stem cell technology, confirmed by encouraging pre-clinical utilization of stem cells in AD animal models, the number of clinical trials evaluating the efficacy of stem cell therapy is limited, with the results of many ongoing clinical trials on cell therapy for AD still pending. Mesenchymal stem cells (MSCs) have been the main focus in these studies, reporting encouraging results concerning safety profile, however their efficacy remains unproven. In the current article we review the latest advances regarding different sources of stem cell therapy and present a comprehensive list of every available clinical trial in national and international registries. Finally, we discuss drawbacks arising from AD pathology and technical limitations that hinder the transition of stem cell technology from bench to bedside. Our findings emphasize the need to increase clinical trials towards uncovering the mode of action and the underlying therapeutic mechanisms of transplanted cells as well as the molecular mechanisms controlling regeneration and neuronal microenvironment.
Collapse
Affiliation(s)
- Nikolaos Karvelas
- Faculty of Medicine, National and Kapodistrian University of Athens, Athina, Greece
| | | | - Georgios Politis
- Faculty of Medicine, National and Kapodistrian University of Athens, Athina, Greece
| | | | - Christo Kole
- Faculty of Medicine, National and Kapodistrian University of Athens, Athina, Greece
| |
Collapse
|
39
|
Rao YL, Ganaraja B, Murlimanju BV, Joy T, Krishnamurthy A, Agrawal A. Hippocampus and its involvement in Alzheimer's disease: a review. 3 Biotech 2022; 12:55. [PMID: 35116217 PMCID: PMC8807768 DOI: 10.1007/s13205-022-03123-4] [Citation(s) in RCA: 205] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/16/2022] [Indexed: 12/12/2022] Open
Abstract
Hippocampus is the significant component of the limbic lobe, which is further subdivided into the dentate gyrus and parts of Cornu Ammonis. It is the crucial region for learning and memory; its sub-regions aid in the generation of episodic memory. However, the hippocampus is one of the brain areas affected by Alzheimer's (AD). In the early stages of AD, the hippocampus shows rapid loss of its tissue, which is associated with the functional disconnection with other parts of the brain. In the progression of AD, atrophy of medial temporal and hippocampal regions are the structural markers in magnetic resonance imaging (MRI). Lack of sirtuin (SIRT) expression in the hippocampal neurons will impair cognitive function, including recent memory and spatial learning. Proliferation, differentiation, and migrations are the steps involved in adult neurogenesis. The microglia in the hippocampal region are more immunologically active than the other regions of the brain. Intrinsic factors like hormones, glia, and vascular nourishment are instrumental in the neural stem cell (NSC) functions by maintaining the brain's microenvironment. Along with the intrinsic factors, many extrinsic factors like dietary intake and physical activity may also influence the NSCs. Hence, pro-neurogenic lifestyle could delay neurodegeneration.
Collapse
Affiliation(s)
- Y. Lakshmisha Rao
- Department of Anatomy, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, Karnataka India
| | - B. Ganaraja
- Department of Physiology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, Karnataka India
| | - B. V. Murlimanju
- Department of Anatomy, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, Karnataka India
| | - Teresa Joy
- Department of Anatomy, College of Medicine, American University of Antigua, Coolidge, Antigua, Antigua and Barbuda
| | - Ashwin Krishnamurthy
- Department of Anatomy, K.S. Hegde Medical Academy, Deralakatte, Nitte University, Mangalore, Karnataka India
| | - Amit Agrawal
- Department of Neurosurgery, All India Institute of Medical Sciences, Saket Nagar, Bhopal, 462020 Madhya Pradesh India
| |
Collapse
|
40
|
Ishimoto T, Kato Y. Ergothioneine in the brain. FEBS Lett 2022; 596:1290-1298. [PMID: 34978075 DOI: 10.1002/1873-3468.14271] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 12/22/2022]
Abstract
Ergothioneine (ERGO) is a naturally occurring food-derived antioxidant. Despite its extremely hydrophilic properties, ERGO is easily absorbed from the gastrointestinal tract and distributed to various organs, including the brain. This is primarily because its entry into brain cells is mediated by the ERGO-specific transporter OCTN1/SLC22A4. Octn1 gene knockout mice do not have ERGO in the brain, due to the absence of OCTN1 in neurons, neural stem cells, and microglia. The existence of OCTN1 and uptake of ERGO into the brain parenchymal cells may suggest that ERGO and its transporter play a pivotal role in brain function. Oral administration of ERGO has antidepressant activities in mice. Furthermore, repeated oral administration of ERGO and ERGO-containing food extract tablets enhance memory function in mice and humans, respectively. ERGO also protects against stress-induced sleep disturbance and neuronal injury induced by amyloid β in rodents. In vitro observations suggest that ERGO benefits brain function through both its antioxidative activity and by promoting neurogenesis and neuronal maturation. This review discusses the possible involvement of ERGO in brain function and its potential therapeutic properties.
Collapse
Affiliation(s)
| | - Yukio Kato
- Faculty of Pharmacy, Kanazawa University, Japan
| |
Collapse
|
41
|
Teplyashina EA, Komleva YK, Lychkovskaya EV, Deikhina AS, Salmina AB. Regulation of neurogenesis and cerebral angiogenesis by cell protein proteolysis products. RUDN JOURNAL OF MEDICINE 2021. [DOI: 10.22363/2313-0245-2021-25-2-114-126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Brain development is a unique process characterized by mechanisms defined as neuroplasticity (synaptogenesis, synapse elimination, neurogenesis, and cerebral angiogenesis). Numerous neurodevelopmental disorders brain damage, and aging are manifested by neurological deficits that are caused by aberrant neuroplasticity. The presence of stem and progenitor cells in neurogenic niches of the brain is responsible for the formation of new neurons capable of integrating into preexisting synaptic assemblies. The determining factors for the cells within the neurogenic niche are the activity of the vascular scaffold and the availability of active regulatory molecules that establish the optimal microenvironment. It has been found that regulated intramembrane proteolysis plays an important role in the control of neurogenesis in brain neurogenic niches. Molecules generated by the activity of specific proteases can stimulate or suppress the activity of neural stem and progenitor cells, their proliferation and differentiation, migration and integration of newly formed neurons into synaptic networks. Local neoangiogenesis supports the processes of neurogenesis in neurogenic niches, which is guaranteed by the multivalent action of peptides formed from transmembrane proteins. Identification of new molecules regulating the neuroplasticity (neurogenesis and angiogenesis). i. e. enzymes, substrates, and products of intramembrane proteolysis, will ensure the development of protocols for detecting the neuroplasticity markers and targets for efficient pharmacological modulation.
Collapse
|
42
|
Metformin and fluoxetine improve depressive-like behavior in a murine model of Parkinsońs disease through the modulation of neuroinflammation, neurogenesis and neuroplasticity. Int Immunopharmacol 2021; 102:108415. [PMID: 34890997 DOI: 10.1016/j.intimp.2021.108415] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/10/2021] [Accepted: 11/25/2021] [Indexed: 02/01/2023]
Abstract
Thereabout 30-40% of patients with Parkinson's Disease (PD) also have depression contributing to the loss of quality of life. Among the patients who treat depression, about 50% do not show significant improvement due to the limited efficacy of the treatment. So far, there are no effective disease-modifying treatments that can impede its progression. The current clinical approach is based on symptom management. Nonetheless, the reuse of drugs with excellent safety profiles represents an attractive alternative strategy for treating of different clinical aspects of PD. In this study, we evaluated the effects of metformin separately and associated with fluoxetine on depressive like-behavior and motor alterations in experimental Parkinson's disease. C57BL6 mice were induced with rotenone (2.5 mg/kg/day) for 20 days and treated with metformin (200 mg/kg/day) and fluoxetine (10 mg/kg/day) from the 5th day of induction. The animals were submitted to Sucrose Preference, Tail Suspension, and rotarod tests. Hippocampus, prefrontal cortex, and substantia nigra were dissected for molecular and morphological analysis. Metformin and fluoxetine prevented depressive-like behavior and improved motor impairment and increased TH nigral positive cells. Metformin and fluoxetine also reduced IBA-1 and GFAP positive cells in the hippocampus. Moreover, metformin reduced the phospho-NF-kB, IL-1β in the prefrontal cortex and iNOS levels in the hippocampus. Both metformin and fluoxetine increased neurogenesis by increasing KI67, but only the combined treatment increased neuronal survival by NeuN positive cells in the hippocampus. In addition, fluoxetine reduced cell death, decreasing caspase-3 and PARP-1 levels. Lastly, metformin potentiated the effect of fluoxetine on neuroplasticity by increasing BDNF positive cells. Metformin has antidepressant and antiparkinsonian potential due to anti-inflammatory neurogenic, and neuroplasticity-inducing effects when combined with fluoxetine.
Collapse
|
43
|
Visvanathar R, Papanikolaou M, Nôga DA, Pádua-Reis M, Tort ABL, Blunder M. Hippocampal Cb 2 receptors: an untold story. Rev Neurosci 2021; 33:413-426. [PMID: 34717053 DOI: 10.1515/revneuro-2021-0109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/01/2021] [Indexed: 12/13/2022]
Abstract
The field of cannabinoid research has been receiving ever-growing interest. Ongoing debates worldwide about the legislation of medical cannabis further motivates research into cannabinoid function within the central nervous system (CNS). To date, two well-characterized cannabinoid receptors exist. While most research has investigated Cb1 receptors (Cb1Rs), Cb2 receptors (Cb2Rs) in the brain have started to attract considerable interest in recent years. With indisputable evidence showing the wide-distribution of Cb2Rs in the brain of different species, they are no longer considered just peripheral receptors. However, in contrast to Cb1Rs, the functionality of central Cb2Rs remains largely unexplored. Here we review recent studies on hippocampal Cb2Rs. While conflicting results about their function have been reported, we have made significant progress in understanding the involvement of Cb2Rs in modulating cellular properties and network excitability. Moreover, Cb2Rs have been shown to be expressed in different subregions of the hippocampus, challenging our prior understanding of the endocannabinoid system. Although more insight into their functional roles is necessary, we propose that targeting hippocampal Cb2Rs may offer novel therapies for diseases related to memory and adult neurogenesis deficits.
Collapse
Affiliation(s)
- Robin Visvanathar
- Behavioral Neurophysiology, Department of Neuroscience, Biomedical Center, Uppsala University, Husargatan 3, 751 23, Uppsala, Sweden
| | - Maria Papanikolaou
- Behavioral Neurophysiology, Department of Neuroscience, Biomedical Center, Uppsala University, Husargatan 3, 751 23, Uppsala, Sweden
| | - Diana Aline Nôga
- Behavioral Neurophysiology, Department of Neuroscience, Biomedical Center, Uppsala University, Husargatan 3, 751 23, Uppsala, Sweden
| | - Marina Pádua-Reis
- Behavioral Neurophysiology, Department of Neuroscience, Biomedical Center, Uppsala University, Husargatan 3, 751 23, Uppsala, Sweden
| | | | - Martina Blunder
- Behavioral Neurophysiology, Department of Neuroscience, Biomedical Center, Uppsala University, Husargatan 3, 751 23, Uppsala, Sweden
| |
Collapse
|
44
|
Leal-Galicia P, Chávez-Hernández ME, Mata F, Mata-Luévanos J, Rodríguez-Serrano LM, Tapia-de-Jesús A, Buenrostro-Jáuregui MH. Adult Neurogenesis: A Story Ranging from Controversial New Neurogenic Areas and Human Adult Neurogenesis to Molecular Regulation. Int J Mol Sci 2021; 22:11489. [PMID: 34768919 PMCID: PMC8584254 DOI: 10.3390/ijms222111489] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/16/2022] Open
Abstract
The generation of new neurons in the adult brain is a currently accepted phenomenon. Over the past few decades, the subventricular zone and the hippocampal dentate gyrus have been described as the two main neurogenic niches. Neurogenic niches generate new neurons through an asymmetric division process involving several developmental steps. This process occurs throughout life in several species, including humans. These new neurons possess unique properties that contribute to the local circuitry. Despite several efforts, no other neurogenic zones have been observed in many years; the lack of observation is probably due to technical issues. However, in recent years, more brain niches have been described, once again breaking the current paradigms. Currently, a debate in the scientific community about new neurogenic areas of the brain, namely, human adult neurogenesis, is ongoing. Thus, several open questions regarding new neurogenic niches, as well as this phenomenon in adult humans, their functional relevance, and their mechanisms, remain to be answered. In this review, we discuss the literature and provide a compressive overview of the known neurogenic zones, traditional zones, and newly described zones. Additionally, we will review the regulatory roles of some molecular mechanisms, such as miRNAs, neurotrophic factors, and neurotrophins. We also join the debate on human adult neurogenesis, and we will identify similarities and differences in the literature and summarize the knowledge regarding these interesting topics.
Collapse
Affiliation(s)
- Perla Leal-Galicia
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (M.E.C.-H.); (F.M.); (J.M.-L.); (L.M.R.-S.); (A.T.-d.-J.)
| | - María Elena Chávez-Hernández
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (M.E.C.-H.); (F.M.); (J.M.-L.); (L.M.R.-S.); (A.T.-d.-J.)
| | - Florencia Mata
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (M.E.C.-H.); (F.M.); (J.M.-L.); (L.M.R.-S.); (A.T.-d.-J.)
| | - Jesús Mata-Luévanos
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (M.E.C.-H.); (F.M.); (J.M.-L.); (L.M.R.-S.); (A.T.-d.-J.)
| | - Luis Miguel Rodríguez-Serrano
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (M.E.C.-H.); (F.M.); (J.M.-L.); (L.M.R.-S.); (A.T.-d.-J.)
- Laboratorio de Neurobiología de la Alimentación, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
| | - Alejandro Tapia-de-Jesús
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (M.E.C.-H.); (F.M.); (J.M.-L.); (L.M.R.-S.); (A.T.-d.-J.)
| | - Mario Humberto Buenrostro-Jáuregui
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (M.E.C.-H.); (F.M.); (J.M.-L.); (L.M.R.-S.); (A.T.-d.-J.)
| |
Collapse
|
45
|
Sun D, Guo H, Womer FY, Yang J, Tang J, Liu J, Zhu Y, Duan J, Peng Z, Wang H, Tan Q, Zhu Q, Wei Y, Xu K, Zhang Y, Tang Y, Zhang X, Xu F, Wang J, Wang F. Frontal-posterior functional imbalance and aberrant function developmental patterns in schizophrenia. Transl Psychiatry 2021; 11:495. [PMID: 34580274 PMCID: PMC8476507 DOI: 10.1038/s41398-021-01617-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/28/2021] [Accepted: 08/20/2021] [Indexed: 12/01/2022] Open
Abstract
Schizophrenia (SZ) is a neurodevelopmental disorder. There remain significant gaps in understanding the neural trajectory across development in SZ. A major research focus is to clarify the developmental functional changes of SZ and to identify the specific timing, the specific brain regions, and the underlying mechanisms of brain alterations during SZ development. Regional homogeneity (ReHo) characterizing brain function was collected and analyzed on humans with SZ (hSZ) and healthy controls (HC) cross-sectionally, and methylazoxymethanol acetate (MAM) rats, a neurodevelopmental model of SZ, and vehicle rats longitudinally from adolescence to adulthood. Metabolomic and proteomic profiling in adult MAM rats and vehicle rats was examined and bioanalyzed. Compared to HC or adult vehicle rats, similar ReHo alterations were observed in hSZ and adult MAM rats, characterized by increased frontal (medial prefrontal and orbitofrontal cortices) and decreased posterior (visual and associated cortices) ReHo. Longitudinal analysis of MAM rats showed aberrant ReHo patterns as decreased posterior ReHo in adolescence and increased frontal and decreased posterior ReHo in adulthood. Accordingly, it was suggested that the visual cortex was a critical locus and adolescence was a sensitive window in SZ development. In addition, metabolic and proteomic alterations in adult MAM rats suggested that central carbon metabolism disturbance and mitochondrial dysfunction were the potential mechanisms underlying the ReHo alterations. This study proposed frontal-posterior functional imbalance and aberrant function developmental patterns in SZ, suggesting that the adolescent visual cortex was a critical locus and a sensitive window in SZ development. These findings from linking data between hSZ and MAM rats may have a significant translational contribution to the development of effective therapies in SZ.
Collapse
Affiliation(s)
- Dandan Sun
- Department of Cardiovascular Ultrasound, The People's Hospital of China Medical University & The People's Hospital of Liaoning Province, Shenyang, China
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Huiling Guo
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Fay Y Womer
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - Jingyu Yang
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Jingwei Tang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Juan Liu
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Yue Zhu
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Jia Duan
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Zhengwu Peng
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Huaning Wang
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Qingrong Tan
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Qiwen Zhu
- Liaoning Key Laboratory of Cognitive Neuroscience, Shenyang Medical College, Shenyang, China
| | - Yange Wei
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Ke Xu
- Department of Radiology, The First Hospital of China Medical University, Shenyang, China
| | - Yanbo Zhang
- Department of Psychiatry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Yanqing Tang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
| | - Xizhe Zhang
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Fuqiang Xu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
- Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jie Wang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Fei Wang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China.
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China.
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
46
|
Miglis MG, Adler CH, Antelmi E, Arnaldi D, Baldelli L, Boeve BF, Cesari M, Dall'Antonia I, Diederich NJ, Doppler K, Dušek P, Ferri R, Gagnon JF, Gan-Or Z, Hermann W, Högl B, Hu MT, Iranzo A, Janzen A, Kuzkina A, Lee JY, Leenders KL, Lewis SJG, Liguori C, Liu J, Lo C, Ehgoetz Martens KA, Nepozitek J, Plazzi G, Provini F, Puligheddu M, Rolinski M, Rusz J, Stefani A, Summers RLS, Yoo D, Zitser J, Oertel WH. Biomarkers of conversion to α-synucleinopathy in isolated rapid-eye-movement sleep behaviour disorder. Lancet Neurol 2021; 20:671-684. [PMID: 34302789 DOI: 10.1016/s1474-4422(21)00176-9] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/19/2022]
Abstract
Patients with isolated rapid-eye-movement sleep behaviour disorder (RBD) are commonly regarded as being in the early stages of a progressive neurodegenerative disease involving α-synuclein pathology, such as Parkinson's disease, dementia with Lewy bodies, or multiple system atrophy. Abnormal α-synuclein deposition occurs early in the neurodegenerative process across the central and peripheral nervous systems and might precede the appearance of motor symptoms and cognitive decline by several decades. These findings provide the rationale to develop reliable biomarkers that can better predict conversion to clinically manifest α-synucleinopathies. In addition, biomarkers of disease progression will be essential to monitor treatment response once disease-modifying therapies become available, and biomarkers of disease subtype will be essential to enable prediction of which subtype of α-synucleinopathy patients with isolated RBD might develop.
Collapse
Affiliation(s)
- Mitchell G Miglis
- Department of Neurology and Neurological Sciences and Department of Psychiatry and Behavioral Science, Stanford University, Palo Alto, CA, USA.
| | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Scottsdale, AZ, USA
| | - Elena Antelmi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Dario Arnaldi
- Clinical Neurology, DINOGMI, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Luca Baldelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Bradley F Boeve
- Department of Neurology and Center for Sleep Medicine, Mayo Clinic, Rochester, MN, USA
| | - Matteo Cesari
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Irene Dall'Antonia
- Department of Neurology and Center of Clinical Neuroscience, Charles University First Faculty of Medicine, Prague, Czech Republic
| | - Nico J Diederich
- Department of Neuroscience, Centre Hospitalier de Luxembourg, Luxembourg City, Luxembourg
| | - Kathrin Doppler
- Department of Neurology, University of Würzburg, Würzburg, Germany
| | - Petr Dušek
- Department of Neurology and Center of Clinical Neuroscience, Charles University First Faculty of Medicine, Prague, Czech Republic
| | | | - Jean-François Gagnon
- Centre for Advanced Research in Sleep Medicine, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Île-de-Montréal-Hôpital du Sacré-Coeur de Montréal, Montreal, QC, Canada
| | - Ziv Gan-Or
- The Neuro-Montreal Neurological Institute-Hospital, Department of Neurology and Neurosurgery, and Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Wiebke Hermann
- Department of Neurology, University of Rostock, Rostock, Germany; German Center for Neurodegenerative Diseases (DZNE), Research Site Rostock, Rostock, Germany
| | - Birgit Högl
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michele T Hu
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Alex Iranzo
- Sleep Disorders Center, Neurology Service, Hospital Clínic Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Annette Janzen
- Department of Neurology and Section on Clinical Neuroscience, Philipps University Marburg, Marburg, Germany
| | | | - Jee-Young Lee
- Department of Neurology, Seoul National University College of Medicine, Seoul, South Korea
| | - Klaus L Leenders
- Department of Nuclear Medicine and Biomedical Imaging, University Medical Center Groningen, Groningen, Netherlands
| | - Simon J G Lewis
- ForeFront Parkinson's Disease Research Clinic, Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - Claudio Liguori
- Sleep Medicine Center, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Jun Liu
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Christine Lo
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Kaylena A Ehgoetz Martens
- Department of Kinesiology, Faculty of Applied Health Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Jiri Nepozitek
- Department of Neurology and Center of Clinical Neuroscience, Charles University First Faculty of Medicine, Prague, Czech Republic
| | - Giuseppe Plazzi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy; Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Federica Provini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy; IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy; UOC Clinica Neurologica Rete Metropolitana NEUROMET, Bellaria Hospital, Bologna, Italy
| | - Monica Puligheddu
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy
| | - Michal Rolinski
- Institute of Clinical Neurosciences, University of Bristol, Bristol, UK
| | - Jan Rusz
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Ambra Stefani
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Dallah Yoo
- Department of Neurology, Kyung Hee University Hospital, Seoul, South Korea
| | - Jennifer Zitser
- Department of Neurology and Neurological Sciences, University of California, San Francisco, CA, USA; Department of Neurology, Tel Aviv Sourasky Medical Center, Affiliate of Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Wolfgang H Oertel
- Department of Neurology and Section on Clinical Neuroscience, Philipps University Marburg, Marburg, Germany; Institute for Neurogenomics, Helmholtz Center for Health and Environment, München-Neuherberg, Germany
| |
Collapse
|
47
|
Zingg JM, Vlad A, Ricciarelli R. Oxidized LDLs as Signaling Molecules. Antioxidants (Basel) 2021; 10:antiox10081184. [PMID: 34439432 PMCID: PMC8389018 DOI: 10.3390/antiox10081184] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022] Open
Abstract
Levels of oxidized low-density lipoproteins (oxLDLs) are usually low in vivo but can increase whenever the balance between formation and scavenging of free radicals is impaired. Under normal conditions, uptake and degradation represent the physiological cellular response to oxLDL exposure. The uptake of oxLDLs is mediated by cell surface scavenger receptors that may also act as signaling molecules. Under conditions of atherosclerosis, monocytes/macrophages and vascular smooth muscle cells highly exposed to oxLDLs tend to convert to foam cells due to the intracellular accumulation of lipids. Moreover, the atherogenic process is accelerated by the increased expression of the scavenger receptors CD36, SR-BI, LOX-1, and SRA in response to high levels of oxLDL and oxidized lipids. In some respects, the effects of oxLDLs, involving cell proliferation, inflammation, apoptosis, adhesion, migration, senescence, and gene expression, can be seen as an adaptive response to the rise of free radicals in the vascular system. Unlike highly reactive radicals, circulating oxLDLs may signal to cells at more distant sites and possibly trigger a systemic antioxidant defense, thus elevating the role of oxLDLs to that of signaling molecules with physiological relevance.
Collapse
Affiliation(s)
- Jean-Marc Zingg
- Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Correspondence: (J.-M.Z.); (R.R.); Tel.: +1-(305)-2433531 (J.-M.Z.); +39-010-3538831 (R.R.)
| | - Adelina Vlad
- Physiology Department, “Carol Davila” UMPh, 020021 Bucharest, Romania;
| | - Roberta Ricciarelli
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Correspondence: (J.-M.Z.); (R.R.); Tel.: +1-(305)-2433531 (J.-M.Z.); +39-010-3538831 (R.R.)
| |
Collapse
|
48
|
Wang G, Wang C, Chen H, Chen L, Li J. Activation of 6-8-week-old new mature adult-born dentate granule cells contributes to anxiety-like behavior. Neurobiol Stress 2021; 15:100358. [PMID: 34195305 PMCID: PMC8240024 DOI: 10.1016/j.ynstr.2021.100358] [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: 02/16/2021] [Revised: 06/17/2021] [Accepted: 06/17/2021] [Indexed: 11/28/2022] Open
Abstract
Adult-born dentate granule cells (aDGCs) at 4–6 weeks of age are particularly excitable but subsequently develop the quiet properties of mature cells. Most existing studies have focused on the hyperactivity of 4–6-week-old aDGCs or neurogenesis, which confers stress resilience or buffers stress responses. However, the function of the quiet property of new mature aDGCs remains unclear. Here we used a retrovirus expressing cre recombinase in combination with an associated-adenovirus to specifically interfere with the activity of new mature aDGCs, and estimated anxiety-like behaviors by the open-field test and elevated plus maze test, antidepressant-like behaviors by the tail suspension test, and spatial memory by the Barnes maze test. We found that sustained hyperactivity of 6–8-week-old, but not 8–10-week-old, aDGCs induced anxiety-like behaviors, and suppression of the activity of 6–8-week-old aDGCs disturbed spatial memory. Meanwhile, sustained hyperactivity of 6–8-week-old aDGCs induced activation of mature dentate gyrus (DG) neurons and inhibition of immature aDGCs. Additionally, the mice showing anxiety-like behaviors induced by chronic mild immobilization stress exhibited increased activity in 6–8-week-old aDGCs. Furthermore, the sustained hyperactivity of mature DG neurons also induced anxiety-like behaviors and decreased the activity of immature aDGCs. Our results combined show that the excitation of 6–8-week-old new mature aDGCs, which prohibits them from normally entering the resting state, determines anxiety-like behavior, while the maintenance of normal excitation ability of 6–8-week-old new mature aDGCs confers memory. Our results suggests that strategies aimed at inhibiting unusual hyperactive new mature aDGCs at a restricted time window may protect against stress-related psychiatric disorders, such as anxiety and depression.
Collapse
Affiliation(s)
- Guohua Wang
- 502 Room, 28 Yunjing Road, Guangzhou, 510515, China
| | - Canmao Wang
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - He Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Limei Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Juan Li
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
49
|
Abstract
With extensive use in industrial and agriculture applications, overexposure to heavy metals has become a global public health concern. The nervous system is vulnerable to many heavy metals, including cadmium, lead, and mercury. However, the knowledge about the underlying mechanisms of these metals' neurotoxicity is still very limited. Adult neurogenesis is a process of generating functional neurons from adult neural progenitor/stem cells (aNPCs), which plays an important role in cognitive function and olfaction. The studies of adult neurogenesis provide new insights into mechanisms of heavy metal neurotoxicity. This review summarizes the current research about the effects of heavy metals on adult neurogenesis and discusses their importance in understanding the mechanisms of heavy metals neurotoxicity, as well as challenges and future directions.
Collapse
Affiliation(s)
- Hao Wang
- Toxicology Program, Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Megumi T. Matsushita
- Toxicology Program, Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| |
Collapse
|
50
|
Chua JP, De Calbiac H, Kabashi E, Barmada SJ. Autophagy and ALS: mechanistic insights and therapeutic implications. Autophagy 2021; 18:254-282. [PMID: 34057020 PMCID: PMC8942428 DOI: 10.1080/15548627.2021.1926656] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mechanisms of protein homeostasis are crucial for overseeing the clearance of misfolded and toxic proteins over the lifetime of an organism, thereby ensuring the health of neurons and other cells of the central nervous system. The highly conserved pathway of autophagy is particularly necessary for preventing and counteracting pathogenic insults that may lead to neurodegeneration. In line with this, mutations in genes that encode essential autophagy factors result in impaired autophagy and lead to neurodegenerative conditions such as amyotrophic lateral sclerosis (ALS). However, the mechanistic details underlying the neuroprotective role of autophagy, neuronal resistance to autophagy induction, and the neuron-specific effects of autophagy-impairing mutations remain incompletely defined. Further, the manner and extent to which non-cell autonomous effects of autophagy dysfunction contribute to ALS pathogenesis are not fully understood. Here, we review the current understanding of the interplay between autophagy and ALS pathogenesis by providing an overview of critical steps in the autophagy pathway, with special focus on pivotal factors impaired by ALS-causing mutations, their physiologic effects on autophagy in disease models, and the cell type-specific mechanisms regulating autophagy in non-neuronal cells which, when impaired, can contribute to neurodegeneration. This review thereby provides a framework not only to guide further investigations of neuronal autophagy but also to refine therapeutic strategies for ALS and related neurodegenerative diseases.Abbreviations: ALS: amyotrophic lateral sclerosis; Atg: autophagy-related; CHMP2B: charged multivesicular body protein 2B; DPR: dipeptide repeat; FTD: frontotemporal dementia; iPSC: induced pluripotent stem cell; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; PINK1: PTEN induced kinase 1; RNP: ribonuclear protein; sALS: sporadic ALS; SPHK1: sphingosine kinase 1; TARDBP/TDP-43: TAR DNA binding protein; TBK1: TANK-binding kinase 1; TFEB: transcription factor EB; ULK: unc-51 like autophagy activating kinase; UPR: unfolded protein response; UPS: ubiquitin-proteasome system; VCP: valosin containing protein.
Collapse
Affiliation(s)
- Jason P Chua
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Hortense De Calbiac
- Recherche translationnelle sur les maladies neurologiques, Institut Imagine, UMR-1163 INSERM et Université Paris Descartes, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Edor Kabashi
- Recherche translationnelle sur les maladies neurologiques, Institut Imagine, UMR-1163 INSERM et Université Paris Descartes, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Sami J Barmada
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|