1
|
Tobias IC, Moorthy SD, Shchuka VM, Langroudi L, Cherednychenko M, Gillespie ZE, Duncan AG, Tian R, Gajewska NA, Di Roberto RB, Mitchell JA. A Sox2 enhancer cluster regulates region-specific neural fates from mouse embryonic stem cells. G3 (BETHESDA, MD.) 2025; 15:jkaf012. [PMID: 39849901 PMCID: PMC12005160 DOI: 10.1093/g3journal/jkaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/14/2025] [Accepted: 01/19/2025] [Indexed: 01/25/2025]
Abstract
Sex-determining region Y box 2 (Sox2) is a critical transcription factor for embryogenesis and neural stem and progenitor cell (NSPC) maintenance. While distal enhancers control Sox2 in embryonic stem cells (ESCs), enhancers closer to the gene are implicated in Sox2 transcriptional regulation in neural development. We hypothesize that a downstream enhancer cluster, termed Sox2 regulatory regions 2-18 (SRR2-18), regulates Sox2 transcription in neural stem cells and we investigate this in NSPCs derived from mouse ESCs. Using functional genomics and CRISPR-Cas9-mediated deletion analyses, we investigate the role of SRR2-18 in Sox2 regulation during neural differentiation. Transcriptome analyses demonstrate that the loss of even 1 copy of SRR2-18 disrupts the region-specific identity of NSPCs, reducing the expression of genes associated with more anterior regions of the embryonic nervous system. Homozygous deletion of this Sox2 neural enhancer cluster causes reduced SOX2 protein, less frequent interaction with transcriptional machinery, and leads to perturbed chromatin accessibility genome-wide further affecting the expression of neurodevelopmental and anterior-posterior regionalization genes. Furthermore, homozygous NSPC deletants exhibit self-renewal defects and impaired differentiation into cell types found in the brain. Altogether, our data define a cis-regulatory enhancer cluster controlling Sox2 transcription in NSPCs and highlight the sensitivity of neural differentiation processes to decreased Sox2 transcription, which causes differentiation into posterior neural fates, specifically the caudal neural tube. This study highlights the importance of precise Sox2 regulation by SRR2-18 in neural differentiation.
Collapse
Affiliation(s)
- Ian C Tobias
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Sakthi D Moorthy
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Virlana M Shchuka
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Lida Langroudi
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Mariia Cherednychenko
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Zoe E Gillespie
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Andrew G Duncan
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Ruxiao Tian
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Natalia A Gajewska
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Raphaël B Di Roberto
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Jennifer A Mitchell
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| |
Collapse
|
2
|
Zheng H, Li L, Wang D, Zhang S, Li W, Cheng M, Ge C, Chen J, Qiang Y, Chen F, Yu Y. FoxO is required for neoblast differentiation during planarian regeneration. Int J Biol Macromol 2025; 288:138729. [PMID: 39672403 DOI: 10.1016/j.ijbiomac.2024.138729] [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: 08/19/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 12/15/2024]
Abstract
Stem cells are of great importance in the maintenance and regeneration of tissues, with Forkhead box O (FoxO) proteins emerging as pivotal regulators of their functions. However, the precise impact of FoxO proteins on stem cell behavior within regenerative environments remains ambiguous. Planarians, renowned for their abundance of adult stem cells (neoblasts), serve as an excellent model for investigating the dynamics of stem cells during regeneration. In this study, we identified DjfoxO, a conserved foxO gene in the planarian Dugesia japonica, and demonstrated its expression in neoblasts, with elevated levels detected in the regenerative blastema during the regeneration process. Using a FoxO inhibitor (AS1842856) together with RNA interference techniques, we demonstrated that inhibition of FoxO signaling in planarians hinders the regeneration of missing tissues, including the central nervous system, eyespots, anterior intestinal branches, and pharynx. It is noteworthy that the knockdown of DjfoxO does not significantly affect the mitotic activity of neoblasts. Conversely, it impedes the production of lineage-specific progenitors, potentially via modulation of the Erk pathway. These findings elucidate the instructive function of FoxO signaling in regulating stem cell differentiation and provide valuable insights into its potential for improving stem cell-based regenerative therapies.
Collapse
Affiliation(s)
- Hanxue Zheng
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Linfeng Li
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Du Wang
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Shengchao Zhang
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Wenhui Li
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Mengdi Cheng
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Cui Ge
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Jiayi Chen
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Yanmei Qiang
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Fulin Chen
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China; Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China
| | - Yuan Yu
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China; Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China.
| |
Collapse
|
3
|
Oh M, Yoon NH, Kim SA, Yoo HJ. Epigenetic Insights into Autism Spectrum Disorder: DNA Methylation Levels of NR3C1, ASCL1, and FOXO3 in Korean Autism Spectrum Disorder Sibling Pairs. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2024; 22:635-645. [PMID: 39420610 PMCID: PMC11494426 DOI: 10.9758/cpn.24.1188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 10/19/2024]
Abstract
Objective Previous research on autism spectrum disorder (ASD) in Koreans has primarily focused on genetic diversity because of its high heritability. However, the emerging recognition of transgenerational epigenetic changes has recently shifted research attention towards epigenetic perspectives. Methods This study investigated the DNA methylation patterns of the promoter regions of candidate genes such as NR3C1, ASCL1, and FOXO3 in blood samples from ASD probands and their unaffected siblings. The analysis included 54 families (ASD proband group: 54; unaffected biological sibling group: 63). The diagnostic process involved screening the probands and their siblings for ASD based on the Diagnostic and Statistical Manual of Mental Disorders 5th edition. Intelligence, social ability, and medical history were thoroughly assessed using various scales and questionnaires. Genomic DNA from blood samples was analyzed using a methylation-sensitive quantitative polymerase chain reaction to examine the DNA methylation status of candidate genes. Results Methylation levels in candidate gene promoter regions differed significantly between the proband and sibling groups for all candidate genes. Correlation analysis between the proband and sibling groups revealed strong and significant correlations in NR3C1 and ASCL1 methylation. Additionally, in the analysis of the relationship between DNA and ASD phenotypes, FOXO3 methylation correlated with social quotient in probands, and ASCL1 methylation was associated with nonverbal communication, and daily living skills as measured by the Korean Vineland Adaptive Behavior Scale. Notably, ASCL1 methylation was significantly associated with parental age at pregnancy. Conclusion This study proposes DNA methylation of NR3C1, ASCL1, and FOXO3 in peripheral blood samples is a potential epigenetic biomarker of ASD.
Collapse
Affiliation(s)
- Miae Oh
- Department of Psychiatry, Kyung Hee University Hospital, Seoul, Korea
| | - Nan-He Yoon
- Division of Social Welfare and Health Administration, Wonkwang University, Iksan, Korea
| | - Soon Ae Kim
- Department of Pharmacology, School of Medicine, Eulji University, Daejon, Korea
| | - Hee Jeong Yoo
- Department of Psychiatry, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
4
|
Samoilova EM, Chudakova DA, Dashinimaev EB, Snezhkina AV, Kudryashova OM, Lipatova AV, Soboleva AV, Vorob’yev PO, Valuev-Elliston VT, Zakirova NF, Ivanov AV, Baklaushev VP. A Snapshot of Early Transcriptional Changes Accompanying the Pro-Neural Phenotype Switch by NGN2, ASCL1, SOX2, and MSI1 in Human Fibroblasts: An RNA-Seq Study. Int J Mol Sci 2024; 25:12385. [PMID: 39596450 PMCID: PMC11594342 DOI: 10.3390/ijms252212385] [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: 10/04/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Direct pro-neural reprogramming is a conversion of differentiated somatic cells to neural cells without an intermediate pluripotency stage. It is usually achieved via ectopic expression (EE) of certain transcription factors (TFs) or other reprogramming factors (RFs). Determining the transcriptional changes (TCs) caused by particular RFs in a given cell line enables an informed approach to reprogramming initiation. Here, we characterized TCs in the human fibroblast cell line LF1 on the 5th day after EE of the single well-known pro-neural RFs NGN2, ASCL1, SOX2, and MSI1. As assessed by expression analysis of the bona fide neuronal markers nestin and beta-III tubulin, all four RFs initiated pro-neuronal phenotype conversion; analysis by RNA-seq revealed striking differences in the resulting TCs, although some pathways were overlapping. ASCL1 and SOX2 were not sufficient to induce significant pro-neural phenotype switches using our EE system. NGN2 induced TCs indicative of cell phenotype changes towards neural crest cells, neural stem cells, mature neurons, as well as radial glia, astrocytes, and oligodendrocyte precursors and their mature forms. MSI1 mainly induced a switch towards early stem-like cells, such as radial glia.
Collapse
Affiliation(s)
- Ekaterina M. Samoilova
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia; (E.M.S.); (O.M.K.); (A.V.S.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
| | - Daria A. Chudakova
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia; (E.M.S.); (O.M.K.); (A.V.S.)
- National Medical Research Center of Children’s Health of the Ministry of Health of the Russian Federation, 119296 Moscow, Russia
| | - Erdem B. Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Anastasiya V. Snezhkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
| | - Olga M. Kudryashova
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia; (E.M.S.); (O.M.K.); (A.V.S.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Anastasia V. Lipatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
| | - Alesya V. Soboleva
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia; (E.M.S.); (O.M.K.); (A.V.S.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
| | - Pavel O. Vorob’yev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
| | - Vladimir T. Valuev-Elliston
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
| | - Natalia F. Zakirova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
| | - Alexander V. Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
| | - Vladimir P. Baklaushev
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia; (E.M.S.); (O.M.K.); (A.V.S.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
- Pulmonology Research Institute, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
- Department of Medical Nanobiotechnology of Medical and Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, 117997 Moscow, Russia
| |
Collapse
|
5
|
Hlavac K, Pavelkova P, Ondrisova L, Mraz M. FoxO1 signaling in B cell malignancies and its therapeutic targeting. FEBS Lett 2024. [PMID: 39533662 DOI: 10.1002/1873-3468.15057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/09/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
FoxO transcription factors (FoxO1, FoxO3a, FoxO4, FoxO6) are a highly evolutionary conserved subfamily of the 'forkhead' box proteins. They have traditionally been considered tumor suppressors, but FoxO1 also exhibits oncogenic properties. The complex nature of FoxO1 is illustrated by its various roles in B cell development and differentiation, immunoglobulin gene rearrangement and cell-surface B cell receptor (BCR) structure, DNA damage control, cell cycle regulation, and germinal center reaction. FoxO1 is tightly regulated at a transcriptional (STAT3, HEB, EBF, FoxOs) and post-transcriptional level (Akt, AMPK, CDK2, GSK3, IKKs, JNK, MAPK/Erk, SGK1, miRNA). In B cell malignancies, recurrent FoxO1 activating mutations (S22/T24) and aberrant nuclear export and activity have been described, underscoring the potential of its therapeutic inhibition. Here, we review FoxO1's roles across B cell and myeloid malignancies, namely acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), follicular lymphoma (FL), diffuse large B cell lymphoma (DLBCL), mantle cell lymphoma (MCL), Burkitt lymphoma (BL), Hodgkin lymphoma (HL), and multiple myeloma (MM). We also discuss preclinical evidence for FoxO1 targeting by currently available inhibitors (AS1708727, AS1842856, cpd10).
Collapse
Affiliation(s)
- Krystof Hlavac
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Brno, Czech Republic
| | - Petra Pavelkova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Brno, Czech Republic
| | - Laura Ondrisova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Brno, Czech Republic
| | - Marek Mraz
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Brno, Czech Republic
| |
Collapse
|
6
|
Özgüldez HÖ, Bulut-Karslioğlu A. Dormancy, Quiescence, and Diapause: Savings Accounts for Life. Annu Rev Cell Dev Biol 2024; 40:25-49. [PMID: 38985838 DOI: 10.1146/annurev-cellbio-112122-022528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Life on Earth has been through numerous challenges over eons and, one way or another, has always triumphed. From mass extinctions to more daily plights to find food, unpredictability is everywhere. The adaptability of life-forms to ever-changing environments is the key that confers life's robustness. Adaptability has become synonymous with Darwinian evolution mediated by heritable genetic changes. The extreme gene-centric view, while being of central significance, at times has clouded our appreciation of the cell as a self-regulating entity informed of, and informing, the genetic data. An essential element that powers adaptability is the ability to regulate cell growth. In this review, we provide an extensive overview of growth regulation spanning species, tissues, and regulatory mechanisms. We aim to highlight the commonalities, as well as differences, of these phenomena and their molecular regulators. Finally, we curate open questions and areas for further exploration.
Collapse
Affiliation(s)
- Hatice Özge Özgüldez
- Stem Cell Chromatin Group, Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany;
| | - Aydan Bulut-Karslioğlu
- Stem Cell Chromatin Group, Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany;
| |
Collapse
|
7
|
Santini L, Kowald S, Cerron-Alvan LM, Huth M, Fabing AP, Sestini G, Rivron N, Leeb M. FoxO transcription factors actuate the formative pluripotency specific gene expression programme. Nat Commun 2024; 15:7879. [PMID: 39251582 PMCID: PMC11384738 DOI: 10.1038/s41467-024-51794-9] [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: 01/24/2024] [Accepted: 08/16/2024] [Indexed: 09/11/2024] Open
Abstract
Naïve pluripotency is sustained by a self-reinforcing gene regulatory network (GRN) comprising core and naïve pluripotency-specific transcription factors (TFs). Upon exiting naïve pluripotency, embryonic stem cells (ESCs) transition through a formative post-implantation-like pluripotent state, where they acquire competence for lineage choice. However, the mechanisms underlying disengagement from the naïve GRN and initiation of the formative GRN are unclear. Here, we demonstrate that phosphorylated AKT acts as a gatekeeper that prevents nuclear localisation of FoxO TFs in naïve ESCs. PTEN-mediated reduction of AKT activity upon exit from naïve pluripotency allows nuclear entry of FoxO TFs, enforcing a cell fate transition by binding and activating formative pluripotency-specific enhancers. Indeed, FoxO TFs are necessary and sufficient for the activation of the formative pluripotency-specific GRN. Our work uncovers a pivotal role for FoxO TFs in establishing formative post-implantation pluripotency, a critical early embryonic cell fate transition.
Collapse
Affiliation(s)
- Laura Santini
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna, Medical University of Vienna, 1030, Vienna, Austria
| | - Saskia Kowald
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria
| | - Luis Miguel Cerron-Alvan
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna, Medical University of Vienna, 1030, Vienna, Austria
| | - Michelle Huth
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna, Medical University of Vienna, 1030, Vienna, Austria
| | - Anna Philina Fabing
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria
| | - Giovanni Sestini
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna, Medical University of Vienna, 1030, Vienna, Austria
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, 1030, Vienna, Austria
| | - Nicolas Rivron
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, 1030, Vienna, Austria
| | - Martin Leeb
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria.
| |
Collapse
|
8
|
Jang EH, Kim SA. Long-Term Epigenetic Regulation of Foxo3 Expression in Neonatal Valproate-Exposed Rat Hippocampus with Sex-Related Differences. Int J Mol Sci 2024; 25:5287. [PMID: 38791325 PMCID: PMC11121443 DOI: 10.3390/ijms25105287] [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: 03/14/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Perinatal exposure to valproic acid is commonly used for autism spectrum disorder (ASD) animal model development. The inhibition of histone deacetylases by VPA has been proposed to induce epigenetic changes during neurodevelopment, but the specific alterations in genetic expression underlying ASD-like behavioral changes remain unclear. We used qPCR-based gene expression and epigenetics tools and Western blotting in the hippocampi of neonatal valproic acid-exposed animals at 4 weeks of age and conducted the social interaction test to detect behavioral changes. Significant alterations in gene expression were observed in males, particularly concerning mRNA expression of Foxo3, which was significantly associated with behavioral changes. Moreover, notable differences were observed in H3K27ac chromatin immunoprecipitation, quantitative PCR (ChIP-qPCR), and methylation-sensitive restriction enzyme-based qPCR targeting the Foxo3 gene promoter region. These findings provide evidence that epigenetically regulated hippocampal Foxo3 expression may influence social interaction-related behavioral changes. Furthermore, identifying sex-specific gene expression and epigenetic changes in this model may elucidate the sex disparity observed in autism spectrum disorder prevalence.
Collapse
Affiliation(s)
| | - Soon-Ae Kim
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon 34824, Republic of Korea;
| |
Collapse
|
9
|
Kaise T, Kageyama R. Transcriptional control of neural stem cell activity. Biochem Soc Trans 2024; 52:617-626. [PMID: 38477464 DOI: 10.1042/bst20230439] [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: 12/20/2023] [Revised: 02/26/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024]
Abstract
In the adult brain, neural stem cells (NSCs) are under the control of various molecular mechanisms to produce an appropriate number of neurons that are essential for specific brain functions. Usually, the majority of adult NSCs stay in a non-proliferative and undifferentiated state known as quiescence, occasionally transitioning to an active state to produce newborn neurons. This transition between the quiescent and active states is crucial for the activity of NSCs. Another significant state of adult NSCs is senescence, in which quiescent cells become more dormant and less reactive, ceasing the production of newborn neurons. Although many genes involved in the regulation of NSCs have been identified using genetic manipulation and omics analyses, the entire regulatory network is complicated and ambiguous. In this review, we focus on transcription factors, whose importance has been elucidated in NSCs by knockout or overexpression studies. We mainly discuss the transcription factors with roles in the active, quiescent, and rejuvenation states of adult NSCs.
Collapse
Affiliation(s)
- Takashi Kaise
- RIKEN Center for Brain Science, Wako 351-0198, Japan
| | | |
Collapse
|
10
|
Cheng M, Nie Y, Song M, Chen F, Yu Y. Forkhead box O proteins: steering the course of stem cell fate. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:7. [PMID: 38466341 DOI: 10.1186/s13619-024-00190-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/26/2024] [Indexed: 03/13/2024]
Abstract
Stem cells are pivotal players in the intricate dance of embryonic development, tissue maintenance, and regeneration. Their behavior is delicately balanced between maintaining their pluripotency and differentiating as needed. Disruptions in this balance can lead to a spectrum of diseases, underscoring the importance of unraveling the complex molecular mechanisms that govern stem cell fate. Forkhead box O (FOXO) proteins, a family of transcription factors, are at the heart of this intricate regulation, influencing a myriad of cellular processes such as survival, metabolism, and DNA repair. Their multifaceted role in steering the destiny of stem cells is evident, as they wield influence over self-renewal, quiescence, and lineage-specific differentiation in both embryonic and adult stem cells. This review delves into the structural and regulatory intricacies of FOXO transcription factors, shedding light on their pivotal roles in shaping the fate of stem cells. By providing insights into the specific functions of FOXO in determining stem cell fate, this review aims to pave the way for targeted interventions that could modulate stem cell behavior and potentially revolutionize the treatment and prevention of diseases.
Collapse
Affiliation(s)
- Mengdi Cheng
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Yujie Nie
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Min Song
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Fulin Chen
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China
| | - Yuan Yu
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China.
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China.
| |
Collapse
|
11
|
Bhogale S, Seward C, Stubbs L, Sinha S. SEAMoD: A fully interpretable neural network for cis-regulatory analysis of differentially expressed genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.09.565900. [PMID: 38014229 PMCID: PMC10680628 DOI: 10.1101/2023.11.09.565900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
A common way to investigate gene regulatory mechanisms is to identify differentially expressed genes using transcriptomics, find their candidate enhancers using epigenomics, and search for over-represented transcription factor (TF) motifs in these enhancers using bioinformatics tools. A related follow-up task is to model gene expression as a function of enhancer sequences and rank TF motifs by their contribution to such models, thus prioritizing among regulators. We present a new computational tool called SEAMoD that performs the above tasks of motif finding and sequence-to-expression modeling simultaneously. It trains a convolutional neural network model to relate enhancer sequences to differential expression in one or more biological conditions. The model uses TF motifs to interpret the sequences, learning these motifs and their relative importance to each biological condition from data. It also utilizes epigenomic information in the form of activity scores of putative enhancers and automatically searches for the most promising enhancer for each gene. Compared to existing neural network models of non-coding sequences, SEAMoD uses far fewer parameters, requires far less training data, and emphasizes biological interpretability. We used SEAMoD to understand regulatory mechanisms underlying the differentiation of neural stem cell (NSC) derived from mouse forebrain. We profiled gene expression and histone modifications in NSC and three differentiated cell types and used SEAMoD to model differential expression of nearly 12,000 genes with an accuracy of 81%, in the process identifying the Olig2, E2f family TFs, Foxo3, and Tcf4 as key transcriptional regulators of the differentiation process.
Collapse
|
12
|
Draijer S, Timmerman R, Pannekeet J, van Harten A, Farshadi EA, Kemmer J, van Gilst D, Chaves I, Hoekman MFM. FoxO3 Modulates Circadian Rhythms in Neural Stem Cells. Int J Mol Sci 2023; 24:13662. [PMID: 37686468 PMCID: PMC10563086 DOI: 10.3390/ijms241713662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Both FoxO transcription factors and the circadian clock act on the interface of metabolism and cell cycle regulation and are important regulators of cellular stress and stem cell homeostasis. Importantly, FoxO3 preserves the adult neural stem cell population by regulating cell cycle and cellular metabolism and has been shown to regulate circadian rhythms in the liver. However, whether FoxO3 is a regulator of circadian rhythms in neural stem cells remains unknown. Here, we show that loss of FoxO3 disrupts circadian rhythmicity in cultures of neural stem cells, an effect that is mediated via regulation of Clock transcriptional levels. Using Rev-Erbα-VNP as a reporter, we then demonstrate that loss of FoxO3 does not disrupt circadian rhythmicity at the single cell level. A meta-analysis of published data revealed dynamic co-occupancy of multiple circadian clock components within FoxO3 regulatory regions, indicating that FoxO3 is a Clock-controlled gene. Finally, we examined proliferation in the hippocampus of FoxO3-deficient mice and found that loss of FoxO3 delayed the circadian phase of hippocampal proliferation, indicating that FoxO3 regulates correct timing of NSC proliferation. Taken together, our data suggest that FoxO3 is an integral part of circadian regulation of neural stem cell homeostasis.
Collapse
Affiliation(s)
- Swip Draijer
- Swammerdam Institute of Life Sciences, University of Amsterdam, 1018 WB Amsterdam, The Netherlands (M.F.M.H.)
| | - Raissa Timmerman
- Swammerdam Institute of Life Sciences, University of Amsterdam, 1018 WB Amsterdam, The Netherlands (M.F.M.H.)
| | - Jesse Pannekeet
- Swammerdam Institute of Life Sciences, University of Amsterdam, 1018 WB Amsterdam, The Netherlands (M.F.M.H.)
| | - Alexandra van Harten
- Swammerdam Institute of Life Sciences, University of Amsterdam, 1018 WB Amsterdam, The Netherlands (M.F.M.H.)
| | - Elham Aida Farshadi
- Department of Molecular Genetics, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Julius Kemmer
- Department of Molecular Genetics, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Demy van Gilst
- Department of Molecular Genetics, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Inês Chaves
- Department of Molecular Genetics, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Marco F. M. Hoekman
- Swammerdam Institute of Life Sciences, University of Amsterdam, 1018 WB Amsterdam, The Netherlands (M.F.M.H.)
| |
Collapse
|
13
|
Singh N, Siebzehnrubl FA, Martinez-Garay I. Transcriptional control of embryonic and adult neural progenitor activity. Front Neurosci 2023; 17:1217596. [PMID: 37588515 PMCID: PMC10426504 DOI: 10.3389/fnins.2023.1217596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/10/2023] [Indexed: 08/18/2023] Open
Abstract
Neural precursors generate neurons in the embryonic brain and in restricted niches of the adult brain in a process called neurogenesis. The precise control of cell proliferation and differentiation in time and space required for neurogenesis depends on sophisticated orchestration of gene transcription in neural precursor cells. Much progress has been made in understanding the transcriptional regulation of neurogenesis, which relies on dose- and context-dependent expression of specific transcription factors that regulate the maintenance and proliferation of neural progenitors, followed by their differentiation into lineage-specified cells. Here, we review some of the most widely studied neurogenic transcription factors in the embryonic cortex and neurogenic niches in the adult brain. We compare functions of these transcription factors in embryonic and adult neurogenesis, highlighting biochemical, developmental, and cell biological properties. Our goal is to present an overview of transcriptional regulation underlying neurogenesis in the developing cerebral cortex and in the adult brain.
Collapse
Affiliation(s)
- Niharika Singh
- Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff, United Kingdom
| | - Florian A. Siebzehnrubl
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff, United Kingdom
| | - Isabel Martinez-Garay
- Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
14
|
Li L, Li X, Han R, Wu M, Ma Y, Chen Y, Zhang H, Li Y. Therapeutic Potential of Chinese Medicine for Endogenous Neurogenesis: A Promising Candidate for Stroke Treatment. Pharmaceuticals (Basel) 2023; 16:ph16050706. [PMID: 37242489 DOI: 10.3390/ph16050706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Strokes are a leading cause of morbidity and mortality in adults worldwide. Extensive preclinical studies have shown that neural-stem-cell-based treatments have great therapeutic potential for stroke. Several studies have confirmed that the effective components of traditional Chinese medicine can protect and maintain the survival, proliferation, and differentiation of endogenous neural stem cells through different targets and mechanisms. Therefore, the use of Chinese medicines to activate and promote endogenous nerve regeneration and repair is a potential treatment option for stroke patients. Here, we summarize the current knowledge regarding neural stem cell strategies for ischemic strokes and the potential effects of these Chinese medicines on neuronal regeneration.
Collapse
Affiliation(s)
- Lin Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiao Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rui Han
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Meirong Wu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yaolei Ma
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuzhao Chen
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Han Zhang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yue Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
15
|
Jang EH, Kim SA. Acute valproate exposure affects proneural factor expression by increasing FOXO3 in the hippocampus of juvenile mice with a sex-based difference. Neurosci Lett 2023; 806:137226. [PMID: 37019270 DOI: 10.1016/j.neulet.2023.137226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/13/2023] [Accepted: 04/02/2023] [Indexed: 04/05/2023]
Abstract
Valproic acid (VPA), an anticonvulsant and mood stabilizer, may affect Notch signaling and mitochondrial function. In a previous study, acute VPA exposure induced increased expression of FOXO3, a transcription factor that shares common targets with pro-neuronal ASCL1. In this study, intraperitoneal acute VPA (400 mg/kg) administration in 4-week-old mice increased and decreased FOXO3 and ASCL1 expression, respectively, in the hippocampus, associated with sex-based differences. Treatment of Foxo3 siRNA increased the mRNA expression levels of Ascl1, Ngn2, Hes6, and Notch1 in PC12 cells. Furthermore, VPA exposure induced significant expression changes of mitochondria-related genes, including COX4 and SIRT1, in hippocampal tissues, associated with sex-based differences. This study suggests that acute VPA exposure differently affects proneural gene expression via FOXO3 induction in the hippocampus based on sex.
Collapse
Affiliation(s)
- Eun Hye Jang
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon, Republic of Korea
| | - Soon Ae Kim
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon, Republic of Korea.
| |
Collapse
|
16
|
Zhang Q, Liu J, Chen L, Zhang M. Promoting Endogenous Neurogenesis as a Treatment for Alzheimer's Disease. Mol Neurobiol 2023; 60:1353-1368. [PMID: 36445633 DOI: 10.1007/s12035-022-03145-2] [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: 06/01/2022] [Accepted: 11/19/2022] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is the most universal neurodegenerative disorder characterized by memory loss and cognitive impairment. AD is biologically defined by production and aggregation of misfolded protein including extracellular amyloid β (Aβ) peptide and intracellular microtubule-associated protein tau tangles in neurons, leading to irreversible neuronal loss. At present, regulation of endogenous neurogenesis to supplement lost neurons has been proposed as a promising strategy for treatment of AD. However, the exact underlying mechanisms of impaired neurogenesis in AD have not been fully explained and effective treatments targeting neurogenesis for AD are limited. In this review, we mainly focus on the latest research of impaired neurogenesis in AD. Then we discuss the factors affecting stages of neurogenesis and the interplay between neural stem cells (NSCs) and neurogenic niche under AD pathological conditions. This review aims to explore potential therapeutic strategies that promote endogenous neurogenesis for AD treatments.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Jingyue Liu
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Li Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China. .,School of Nursing, Jilin University, Changchun, China.
| | - Ming Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
17
|
Chang ZS, He ZM, Xia JB. FoxO3 Regulates the Progress and Development of Aging and Aging-Related Diseases. Curr Mol Med 2023; 23:991-1006. [PMID: 36239722 DOI: 10.2174/1566524023666221014140817] [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: 06/20/2022] [Revised: 09/04/2022] [Accepted: 09/06/2022] [Indexed: 11/22/2022]
Abstract
Aging is an inevitable risk factor for many diseases, including cardiovascular diseases, neurodegenerative diseases, cancer, and diabetes. Investigation into the molecular mechanisms involved in aging and longevity will benefit the treatment of age-dependent diseases and the development of preventative medicine for agingrelated diseases. Current evidence has revealed that FoxO3, encoding the transcription factor (FoxO)3, a key transcription factor that integrates different stimuli in the intrinsic and extrinsic pathways and is involved in cell differentiation, protein homeostasis, stress resistance and stem cell status, plays a regulatory role in longevity and in age-related diseases. However, the precise mechanisms by which the FoxO3 transcription factor modulates aging and promotes longevity have been unclear until now. Here, we provide a brief overview of the mechanisms by which FoxO3 mediates signaling in pathways involved in aging and aging-related diseases, as well as the current knowledge on the role of the FoxO3 transcription factor in the human lifespan and its clinical prospects. Ultimately, we conclude that FoxO3 signaling pathways, including upstream and downstream molecules, may be underlying therapeutic targets in aging and age-related diseases.
Collapse
Affiliation(s)
- Zao-Shang Chang
- Department of Physiology, School of Basic Medical Sciences, Shaoyang University, Shaoyang 422000, Hunan, China
| | - Zhi-Ming He
- Department of Physiology, School of Basic Medical Sciences, Shaoyang University, Shaoyang 422000, Hunan, China
| | - Jing-Bo Xia
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Guangzhou Sport University, Guangzhou 510500, Guangdong, China
| |
Collapse
|
18
|
Endothelial cells regulate astrocyte to neural progenitor cell trans-differentiation in a mouse model of stroke. Nat Commun 2022; 13:7812. [PMID: 36535938 PMCID: PMC9763251 DOI: 10.1038/s41467-022-35498-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
The concept of the neurovascular unit emphasizes the importance of cell-cell signaling between neural, glial, and vascular compartments. In neurogenesis, for example, brain endothelial cells play a key role by supplying trophic support to neural progenitors. Here, we describe a surprising phenomenon where brain endothelial cells may release trans-differentiation signals that convert astrocytes into neural progenitor cells in male mice after stroke. After oxygen-glucose deprivation, brain endothelial cells release microvesicles containing pro-neural factor Ascl1 that enter into astrocytes to induce their trans-differentiation into neural progenitors. In mouse models of focal cerebral ischemia, Ascl1 is upregulated in endothelium prior to astrocytic conversion into neural progenitor cells. Injecting brain endothelial-derived microvesicles amplifies the process of astrocyte trans-differentiation. Endothelial-specific overexpression of Ascl1 increases the local conversion of astrocytes into neural progenitors and improves behavioral recovery. Our findings describe an unexpected vascular-regulated mechanism of neuroplasticity that may open up therapeutic opportunities for improving outcomes after stroke.
Collapse
|
19
|
Gui T, Burgering BMT. FOXOs: masters of the equilibrium. FEBS J 2022; 289:7918-7939. [PMID: 34610198 PMCID: PMC10078705 DOI: 10.1111/febs.16221] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/22/2021] [Accepted: 10/04/2021] [Indexed: 01/14/2023]
Abstract
Forkhead box O (FOXO) transcription factors (TFs) are a subclass of the larger family of forkhead TFs. Mammalians express four members FOXO1, FOXO3, FOXO4, and FOXO6. The interest in FOXO function stems mostly from their observed role in determining lifespan, where in model organisms, increased FOXO activity results in extended lifespan. FOXOs act as downstream of several signaling pathway and are extensively regulated through post-translational modifications. The transcriptional program activated by FOXOs in various cell types, organisms, and under various conditions has been described and has shed some light on what the critical transcriptional targets are in mediating FOXO function. At the cellular level, these studies have revealed a role for FOXOs in cell metabolism, cellular redox, cell proliferation, DNA repair, autophagy, and many more. The general picture that emerges hereof is that FOXOs act to preserve equilibrium, and they are important for cellular homeostasis. Here, we will first briefly summarize the general knowledge of FOXO regulation and possible functions. We will use genomic stability to illustrate how FOXOs ensure homeostasis. Genomic stability is critical for maintaining genetic integrity, and therefore preventing disease. However, genomic mutations need to occur during lifetime to enable evolution, yet their accumulation is believed to be causative to aging. Therefore, the role of FOXO in genomic stability may underlie its role in lifespan and aging. Finally, we will come up with questions on some of the unknowns in FOXO function, the answer(s) to which we believe will further our understanding of FOXO function and ultimately may help to understand lifespan and its consequences.
Collapse
Affiliation(s)
- Tianshu Gui
- Molecular Cancer Research, Center Molecular Medicine, University Medical Center Utrecht and the Oncode Institute, The Netherlands
| | - Boudewijn M T Burgering
- Molecular Cancer Research, Center Molecular Medicine, University Medical Center Utrecht and the Oncode Institute, The Netherlands
| |
Collapse
|
20
|
Angelopoulos I, Gakis G, Birmpas K, Kyrousi C, Habeos EE, Kaplani K, Lygerou Z, Habeos I, Taraviras S. Metabolic regulation of the neural stem cell fate: Unraveling new connections, establishing new concepts. Front Neurosci 2022; 16:1009125. [PMID: 36340763 PMCID: PMC9634649 DOI: 10.3389/fnins.2022.1009125] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/03/2022] [Indexed: 11/30/2022] Open
Abstract
The neural stem cell niche is a key regulator participating in the maintenance, regeneration, and repair of the brain. Within the niche neural stem cells (NSC) generate new neurons throughout life, which is important for tissue homeostasis and brain function. NSCs are regulated by intrinsic and extrinsic factors with cellular metabolism being lately recognized as one of the most important ones, with evidence suggesting that it may serve as a common signal integrator to ensure mammalian brain homeostasis. The aim of this review is to summarize recent insights into how metabolism affects NSC fate decisions in adult neural stem cell niches, with occasional referencing of embryonic neural stem cells when it is deemed necessary. Specifically, we will highlight the implication of mitochondria as crucial regulators of NSC fate decisions and the relationship between metabolism and ependymal cells. The link between primary cilia dysfunction in the region of hypothalamus and metabolic diseases will be examined as well. Lastly, the involvement of metabolic pathways in ependymal cell ciliogenesis and physiology regulation will be discussed.
Collapse
Affiliation(s)
| | - Georgios Gakis
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| | - Kyriakos Birmpas
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| | - Christina Kyrousi
- First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, Eginition Hospital, Athens, Greece
- University Mental Health, Neurosciences and Precision Medicine Research Institute “Costas Stefanis”, Athens, Greece
| | - Evagelia Eva Habeos
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| | - Konstantina Kaplani
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| | - Zoi Lygerou
- Department of General Biology, School of Medicine, University of Patras, Patras, Greece
| | - Ioannis Habeos
- Division of Endocrinology, Department of Internal Medicine, University of Patras, Patras, Greece
| | - Stavros Taraviras
- Department of Physiology, Medical School, University of Patras, Patras, Greece
- *Correspondence: Stavros Taraviras,
| |
Collapse
|
21
|
Lalli M, Yen A, Thopte U, Dong F, Moudgil A, Chen X, Milbrandt J, Dougherty JD, Mitra RD. Measuring transcription factor binding and gene expression using barcoded self-reporting transposon calling cards and transcriptomes. NAR Genom Bioinform 2022; 4:lqac061. [PMID: 36062164 PMCID: PMC9428926 DOI: 10.1093/nargab/lqac061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/04/2022] [Accepted: 08/24/2022] [Indexed: 11/29/2022] Open
Abstract
Calling cards technology using self-reporting transposons enables the identification of DNA-protein interactions through RNA sequencing. Although immensely powerful, current implementations of calling cards in bulk experiments on populations of cells are technically cumbersome and require many replicates to identify independent insertions into the same genomic locus. Here, we have drastically reduced the cost and labor requirements of calling card experiments in bulk populations of cells by introducing a DNA barcode into the calling card itself. An additional barcode incorporated during reverse transcription enables simultaneous transcriptome measurement in a facile and affordable protocol. We demonstrate that barcoded self-reporting transposons recover in vitro binding sites for four basic helix-loop-helix transcription factors with important roles in cell fate specification: ASCL1, MYOD1, NEUROD2 and NGN1. Further, simultaneous calling cards and transcriptional profiling during transcription factor overexpression identified both binding sites and gene expression changes for two of these factors. Lastly, we demonstrated barcoded calling cards can record binding in vivo in the mouse brain. In sum, RNA-based identification of transcription factor binding sites and gene expression through barcoded self-reporting transposon calling cards and transcriptomes is an efficient and powerful method to infer gene regulatory networks in a population of cells.
Collapse
Affiliation(s)
- Matthew Lalli
- Department of Genetics, School of Medicine, Washington University in St. Louis School of Medicine, Saint Louis, MO 63110, USA.,Edison Family Center for Genome Sciences and Systems Biology Washington University in St. Louis School of Medicine, Saint Louis, MO 63110, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Allen Yen
- Department of Genetics, School of Medicine, Washington University in St. Louis School of Medicine, Saint Louis, MO 63110, USA.,Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Urvashi Thopte
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Fengping Dong
- Department of Genetics, School of Medicine, Washington University in St. Louis School of Medicine, Saint Louis, MO 63110, USA.,Edison Family Center for Genome Sciences and Systems Biology Washington University in St. Louis School of Medicine, Saint Louis, MO 63110, USA
| | - Arnav Moudgil
- Department of Genetics, School of Medicine, Washington University in St. Louis School of Medicine, Saint Louis, MO 63110, USA.,Edison Family Center for Genome Sciences and Systems Biology Washington University in St. Louis School of Medicine, Saint Louis, MO 63110, USA
| | - Xuhua Chen
- Department of Genetics, School of Medicine, Washington University in St. Louis School of Medicine, Saint Louis, MO 63110, USA.,Edison Family Center for Genome Sciences and Systems Biology Washington University in St. Louis School of Medicine, Saint Louis, MO 63110, USA
| | - Jeffrey Milbrandt
- Department of Genetics, School of Medicine, Washington University in St. Louis School of Medicine, Saint Louis, MO 63110, USA
| | - Joseph D Dougherty
- Department of Genetics, School of Medicine, Washington University in St. Louis School of Medicine, Saint Louis, MO 63110, USA.,Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Robi D Mitra
- Department of Genetics, School of Medicine, Washington University in St. Louis School of Medicine, Saint Louis, MO 63110, USA.,Edison Family Center for Genome Sciences and Systems Biology Washington University in St. Louis School of Medicine, Saint Louis, MO 63110, USA
| |
Collapse
|
22
|
ASCL1 activates neuronal stem cell-like lineage programming through remodeling of the chromatin landscape in prostate cancer. Nat Commun 2022; 13:2282. [PMID: 35477723 PMCID: PMC9046280 DOI: 10.1038/s41467-022-29963-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 04/08/2022] [Indexed: 12/21/2022] Open
Abstract
Treatment with androgen receptor pathway inhibitors (ARPIs) in prostate cancer leads to the emergence of resistant tumors characterized by lineage plasticity and differentiation toward neuroendocrine lineage. Here, we find that ARPIs induce a rapid epigenetic alteration mediated by large-scale chromatin remodeling to support activation of stem/neuronal transcriptional programs. We identify the proneuronal transcription factor ASCL1 motif to be enriched in hyper-accessible regions. ASCL1 acts as a driver of the lineage plastic, neuronal transcriptional program to support treatment resistance and neuroendocrine phenotype. Targeting ASCL1 switches the neuroendocrine lineage back to the luminal epithelial state. This effect is modulated by disruption of the polycomb repressive complex-2 through UHRF1/AMPK axis and change the chromatin architecture in favor of luminal phenotype. Our study provides insights into the epigenetic alterations induced by ARPIs, governed by ASCL1, provides a proof of principle of targeting ASCL1 to reverse neuroendocrine phenotype, support luminal conversion and re-addiction to ARPIs. Following androgen receptor pathway inhibition prostate cancers can differentiate towards the neuroendocrine lineage. Here, the authors identify epigenetic alterations regulated by ASCL1 and suggest targeting ASCL1 to reverse the neuroendocrine phenotype.
Collapse
|
23
|
Leung RF, George AM, Roussel EM, Faux MC, Wigle JT, Eisenstat DD. Genetic Regulation of Vertebrate Forebrain Development by Homeobox Genes. Front Neurosci 2022; 16:843794. [PMID: 35546872 PMCID: PMC9081933 DOI: 10.3389/fnins.2022.843794] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/14/2022] [Indexed: 01/19/2023] Open
Abstract
Forebrain development in vertebrates is regulated by transcription factors encoded by homeobox, bHLH and forkhead gene families throughout the progressive and overlapping stages of neural induction and patterning, regional specification and generation of neurons and glia from central nervous system (CNS) progenitor cells. Moreover, cell fate decisions, differentiation and migration of these committed CNS progenitors are controlled by the gene regulatory networks that are regulated by various homeodomain-containing transcription factors, including but not limited to those of the Pax (paired), Nkx, Otx (orthodenticle), Gsx/Gsh (genetic screened), and Dlx (distal-less) homeobox gene families. This comprehensive review outlines the integral role of key homeobox transcription factors and their target genes on forebrain development, focused primarily on the telencephalon. Furthermore, links of these transcription factors to human diseases, such as neurodevelopmental disorders and brain tumors are provided.
Collapse
Affiliation(s)
- Ryan F. Leung
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Ankita M. George
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Enola M. Roussel
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Maree C. Faux
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Jeffrey T. Wigle
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - David D. Eisenstat
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
24
|
Woods LM, Ali FR, Gomez R, Chernukhin I, Marcos D, Parkinson LM, Tayoun ANA, Carroll JS, Philpott A. Elevated ASCL1 activity creates de novo regulatory elements associated with neuronal differentiation. BMC Genomics 2022; 23:255. [PMID: 35366798 PMCID: PMC8977041 DOI: 10.1186/s12864-022-08495-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 03/23/2022] [Indexed: 12/14/2022] Open
Abstract
Background The pro-neural transcription factor ASCL1 is a master regulator of neurogenesis and a key factor necessary for the reprogramming of permissive cell types to neurons. Endogenously, ASCL1 expression is often associated with neuroblast stem-ness. Moreover, ASCL1-mediated reprogramming of fibroblasts to differentiated neurons is commonly achieved using artificially high levels of ASCL1 protein, where ASCL1 acts as an “on-target” pioneer factor. However, the genome-wide effects of enhancing ASCL1 activity in a permissive neurogenic environment has not been thoroughly investigated. Here, we overexpressed ASCL1 in the neuronally-permissive context of neuroblastoma (NB) cells where modest endogenous ASCL1 supports the neuroblast programme. Results Increasing ASCL1 in neuroblastoma cells both enhances binding at existing ASCL1 sites and also leads to creation of numerous additional, lower affinity binding sites. These extensive genome-wide changes in ASCL1 binding result in significant reprogramming of the NB transcriptome, redirecting it from a proliferative neuroblastic state towards one favouring neuronal differentiation. Mechanistically, ASCL1-mediated cell cycle exit and differentiation can be increased further by preventing its multi-site phosphorylation, which is associated with additional changes in genome-wide binding and gene activation profiles. Conclusions Our findings show that enhancing ASCL1 activity in a neurogenic environment both increases binding at endogenous ASCL1 sites and also results in additional binding to new low affinity sites that favours neuronal differentiation over the proliferating neuroblast programme supported by the endogenous protein. These findings have important implications for controlling processes of neurogenesis in cancer and cellular reprogramming. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08495-8.
Collapse
|
25
|
Audesse AJ, Karashchuk G, Gardell ZA, Lakis NS, Maybury-Lewis SY, Brown AK, Leeman DS, Teo YV, Neretti N, Anthony DC, Brodsky AS, Webb AE. FOXO3 regulates a common genomic program in aging and glioblastoma stem cells. AGING AND CANCER 2021; 2:137-159. [PMID: 36303712 PMCID: PMC9601604 DOI: 10.1002/aac2.12043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 11/14/2021] [Indexed: 01/14/2023]
Abstract
Background Glioblastoma (GBM) is an aggressive, age-associated malignant glioma that contains populations of cancer stem cells. These glioma stem cells (GSCs) evade therapeutic interventions and repopulate tumors due to their existence in a slowly cycling quiescent state. Although aging is well known to increase cancer initiation, the extent to which the mechanisms supporting GSC tumorigenicity are related to physiological aging remains unknown. Aims Here, we investigate the transcriptional mechanisms by which Forkhead Box O3 (FOXO3), a transcriptional regulator that promotes healthy aging, affects GSC function and the extent to which FOXO3 transcriptional networks are dysregulated in aging and GBM. Methods and results We performed transcriptome analysis of clinical GBM tumors and observed that high FOXO3 activity is associated with gene expression signatures of stem cell quiescence, reduced oxidative metabolism, and improved patient outcomes. Consistent with these findings, we show that elevated FOXO3 activity significantly reduces the proliferation of GBM-derived GSCs. Using RNA-seq, we find that functional ablation of FOXO3 in GSCs rewires the transcriptional circuitry associated with metabolism, epigenetic stability, quiescence, and differentiation. Since FOXO3 has been implicated in healthy aging, we then investigated the extent to which it regulates common transcriptional programs in aging neural stem cells (NSCs) and GSCs. We uncover a shared transcriptional program and, most strikingly, find that FOXO3-regulated pathways are associated with altered mitochondrial functions in both aging and GBM. Conclusions This work identifies a FOXO-associated transcriptional program that correlates between GSCs and aging NSCs and is enriched for metabolic and stemness pathways connected with GBM and aging.
Collapse
Affiliation(s)
- Amanda J. Audesse
- Neuroscience Graduate Program, Brown University, Providence, Rhode Island, USA
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Galina Karashchuk
- Department of Pathology and Laboratory Medicine, Lifespan Academic Medical Center and Warren Alpert Medical School at Brown University, Providence, Rhode Island, USA
| | - Zachary A. Gardell
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Nelli S. Lakis
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Sun Y. Maybury-Lewis
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, USA
- Molecular Biology, Cell Biology, and Biochemistry Graduate Program, Brown University, Providence, Rhode Island, USA
| | - Abigail K. Brown
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, USA
- Molecular Biology, Cell Biology, and Biochemistry Graduate Program, Brown University, Providence, Rhode Island, USA
| | - Dena S. Leeman
- Department of Discovery Immunology, Genentech, Inc., South San Francisco, California, USA
| | - Yee Voan Teo
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, USA
- Molecular Biology, Cell Biology, and Biochemistry Graduate Program, Brown University, Providence, Rhode Island, USA
| | - Nicola Neretti
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, USA
- Center on the Biology of Aging, Brown University, Providence, Rhode Island, USA
| | - Douglas C. Anthony
- Department of Pathology and Laboratory Medicine, Lifespan Academic Medical Center and Warren Alpert Medical School at Brown University, Providence, Rhode Island, USA
- Department of Neurology, Brown University, Providence, Rhode Island, USA
| | - Alexander S. Brodsky
- Department of Pathology and Laboratory Medicine, Lifespan Academic Medical Center and Warren Alpert Medical School at Brown University, Providence, Rhode Island, USA
| | - Ashley E. Webb
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, USA
- Center on the Biology of Aging, Brown University, Providence, Rhode Island, USA
- Carney Institute for Brain Science, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
26
|
Maybury‐Lewis SY, Brown AK, Yeary M, Sloutskin A, Dhakal S, Juven‐Gershon T, Webb AE. Changing and stable chromatin accessibility supports transcriptional overhaul during neural stem cell activation and is altered with age. Aging Cell 2021; 20:e13499. [PMID: 34687484 PMCID: PMC8590101 DOI: 10.1111/acel.13499] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 06/25/2021] [Accepted: 09/13/2021] [Indexed: 12/22/2022] Open
Abstract
Neural stem cells (NSCs) in the adult and aged brain are largely quiescent, and require transcriptional reprogramming to re-enter the cell cycle. However, the mechanisms underlying these changes and how they are altered with age remain undefined. Here, we identify the chromatin accessibility differences between primary neural stem/progenitor cells in quiescent and activated states. These distinct cellular states exhibit shared and unique chromatin profiles, both associated with gene regulation. Accessible chromatin states specific to activation or quiescence are active enhancers bound by key pro-neurogenic and quiescence factors. In contrast, shared sites are enriched for core promoter elements associated with translation and metabolism. Unexpectedly, through integrated analysis, we find that many sites that become accessible during NSC activation are linked to gene repression and associated with pro-quiescence factors, revealing a novel mechanism that may preserve quiescence re-entry. Furthermore, we report that in aged NSCs, chromatin regions associated with metabolic and transcriptional functions bound by key pro-quiescence transcription factors lose accessibility, suggesting a novel mechanism of age-associated NSC dysfunction. Together, our findings reveal how accessible chromatin states regulate the transcriptional switch between NSC quiescence and activation, and how this switch is affected with age.
Collapse
Affiliation(s)
- Sun Y. Maybury‐Lewis
- Department of Molecular Biology, Cell Biology, and Biochemistry Brown University Providence Rhode Island USA
| | - Abigail K. Brown
- Department of Molecular Biology, Cell Biology, and Biochemistry Brown University Providence Rhode Island USA
| | - Mitchell Yeary
- Department of Molecular Biology, Cell Biology, and Biochemistry Brown University Providence Rhode Island USA
| | - Anna Sloutskin
- The Mina and Everard Goodman Faculty of Life Sciences Bar‐Ilan University Ramat Gan Israel
| | - Shleshma Dhakal
- Department of Molecular Biology, Cell Biology, and Biochemistry Brown University Providence Rhode Island USA
| | - Tamar Juven‐Gershon
- The Mina and Everard Goodman Faculty of Life Sciences Bar‐Ilan University Ramat Gan Israel
| | - Ashley E. Webb
- Department of Molecular Biology, Cell Biology, and Biochemistry Brown University Providence Rhode Island USA
- Carney Institute for Brain Science Brown University Providence Rhode Island USA
- Center on the Biology of Aging Brown University Providence Rhode Island USA
| |
Collapse
|
27
|
Gillotin S, Sahni V, Lepko T, Hanspal MA, Swartz JE, Alexopoulou Z, Marshall FH. Targeting impaired adult hippocampal neurogenesis in ageing by leveraging intrinsic mechanisms regulating Neural Stem Cell activity. Ageing Res Rev 2021; 71:101447. [PMID: 34403830 DOI: 10.1016/j.arr.2021.101447] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/14/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023]
Abstract
Deficits in adult neurogenesis may contribute to the aetiology of many neurodevelopmental, psychiatric and neurodegenerative diseases. Genetic ablation of neurogenesis provides proof of concept that adult neurogenesis is required to sustain complex and dynamic cognitive functions, such as learning and memory, mostly by providing a high degree of plasticity to neuronal circuits. In addition, adult neurogenesis is reactive to external stimuli and the environment making it particularly susceptible to impairment and consequently contributing to comorbidity. In the human brain, the dentate gyrus of the hippocampus is the main active source of neural stem cells that generate granule neurons throughout life. The regulation and preservation of the pool of neural stem cells is central to ensure continuous and healthy adult hippocampal neurogenesis (AHN). Recent advances in genetic and metabolic profiling alongside development of more predictive animal models have contributed to the development of new concepts and the emergence of molecular mechanisms that could pave the way to the implementation of new therapeutic strategies to treat neurological diseases. In this review, we discuss emerging molecular mechanisms underlying AHN that could be embraced in drug discovery to generate novel concepts and targets to treat diseases of ageing including neurodegeneration. To support this, we review cellular and molecular mechanisms that have recently been identified to assess how AHN is sustained throughout life and how AHN is associated with diseases. We also provide an outlook on strategies for developing correlated biomarkers that may accelerate the translation of pre-clinical and clinical data and review clinical trials for which modulation of AHN is part of the therapeutic strategy.
Collapse
|
28
|
Samoilova EM, Belopasov VV, Baklaushev VP. Transcription Factors of Direct Neuronal Reprogramming in Ontogenesis and Ex Vivo. Mol Biol 2021; 55:645-669. [DOI: 10.1134/s0026893321040087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 03/07/2025]
|
29
|
Kim SA, Chai JH, Jang EH. Prenatal Trimethyltin Exposure Induces Long-Term DNA Methylation Changes in the Male Mouse Hippocampus. Int J Mol Sci 2021; 22:ijms22158009. [PMID: 34360774 PMCID: PMC8348768 DOI: 10.3390/ijms22158009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/14/2021] [Accepted: 07/23/2021] [Indexed: 12/12/2022] Open
Abstract
Trimethyltin (TMT) is an irreversible neurotoxicant. Because prenatal TMT exposure has been reported to induce behavioral changes, this study was conducted to observe gender differences and epigenetic changes using a mouse model. In behavioral testing of offspring at 5 weeks of age, the total times spent in the center, corner, or border zones in the male prenatal TMT-exposed mice were less than those of control unexposed mice in the open-field test. Female TMT-exposed mice scored lower on total numbers of arm entries and percentages of alternations than controls in the Y-maze test with lower body weight. We found that only TMT-exposed males had fewer copies of mtDNA in the hippocampus and prefrontal cortex region than controls. Additional epigenetic changes, including increased 5-methyl cytosine/5-hydroxymethyl cytosine levels in the male TMT hippocampus, were observed. After methylation binding domain (MBD) sequencing, multiple signaling pathways related to metabolism and neurodevelopment, including FoxO signaling, were identified by pathway analysis for differentially methylated regions (DMRs). Increased FOXO3 and decreased ASCL1 expression were also observed in male TMT hippocampi. This study suggests that sex differences and epigenetics should be more carefully considered in prenatal toxicology studies.
Collapse
Affiliation(s)
- Soon-Ae Kim
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon 34824, Korea;
- Correspondence: ; Tel.: +82-42-259-1672
| | - Jung-Hoon Chai
- Center for Sport Science in Seoul, Seoul Sports Council, Seoul 02119, Korea;
| | - Eun-Hye Jang
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon 34824, Korea;
| |
Collapse
|
30
|
Rana T, Behl T, Sehgal A, Mehta V, Singh S, Sharma N, Bungau S. Elucidating the Possible Role of FoxO in Depression. Neurochem Res 2021; 46:2761-2775. [PMID: 34075521 DOI: 10.1007/s11064-021-03364-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/23/2021] [Accepted: 05/25/2021] [Indexed: 12/21/2022]
Abstract
Forkhead box-O (FoxO) transcriptional factors perform essential functions in several physiological and biological processes. Recent studies have shown that FoxO is implicated in the pathophysiology of depression. Changes in the upstream mediators of FoxOs including brain-derived neurotrophic factor (BDNF) and protein kinase B have been associated with depressive disorder and the antidepressant agents are known to alter the phosphorylation of FoxOs. Moreover, FoxOs might be regulated by serotonin or noradrenaline signaling and the hypothalamic-pituitary-adrenal (HPA)-axis,both of them are associated with the development of the depressive disorder. FoxO also regulates neural morphology, synaptogenesis, and neurogenesis in the hippocampus, which accounts for the pathogenesis of the depressive disorder. The current article underlined the potential functions of FoxOs in the etiology of depressive disorder and formulate few essential proposals for further investigation. The review also proposes that FoxO and its signal pathway might establish possible therapeutic mediators for the management of depressive disorder.
Collapse
Affiliation(s)
- Tarapati Rana
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India.,Government Pharmacy College, Seraj, Mandi, Himachal Pradesh, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Vineet Mehta
- Government College of Pharmacy, Rohru, Distt., Shimla, Himachal Pradesh, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
31
|
Soh R, Hardy A, Zur Nieden NI. The FOXO signaling axis displays conjoined functions in redox homeostasis and stemness. Free Radic Biol Med 2021; 169:224-237. [PMID: 33878426 PMCID: PMC9910585 DOI: 10.1016/j.freeradbiomed.2021.04.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023]
Abstract
Previous views of reactive oxygen species (ROS) depicted them as harmful byproducts of metabolism as uncontrolled levels of ROS can lead to DNA damage and cell death. However, recent studies have shed light into the key role of ROS in the self-renewal or differentiation of the stem cell. The interplay between ROS levels, metabolism, and the downstream redox signaling pathways influence stem cell fate. In this review we will define ROS, explain how they are generated, and how ROS signaling can influence transcription factors, first and foremost forkhead box-O transcription factors, that shape not only the cellular redox state, but also stem cell fate. Now that studies have illustrated the importance of redox homeostasis and the role of redox signaling, understanding the mechanisms behind this interplay will further shed light into stem cell biology.
Collapse
Affiliation(s)
- Ruthia Soh
- Department of Molecular, Cell and Systems Biology, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, 92521, CA, USA
| | - Ariana Hardy
- Department of Molecular, Cell and Systems Biology, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, 92521, CA, USA
| | - Nicole I Zur Nieden
- Department of Molecular, Cell and Systems Biology, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, 92521, CA, USA; Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, 92521, CA, USA.
| |
Collapse
|
32
|
Regulation of Adult Mammalian Neural Stem Cells and Neurogenesis by Cell Extrinsic and Intrinsic Factors. Cells 2021; 10:cells10051145. [PMID: 34068607 PMCID: PMC8150395 DOI: 10.3390/cells10051145] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 05/07/2021] [Indexed: 12/27/2022] Open
Abstract
Tissue-specific stem cells give rise to new functional cells to maintain tissue homeostasis and restore damaged tissue after injury. To ensure proper brain functions in the adult brain, neural stem cells (NSCs) continuously generate newborn neurons that integrate into pre-existing neuronal networks. Proliferation, as well as neurogenesis of NSCs, are exquisitely controlled by extrinsic and intrinsic factors, and their underlying mechanisms have been extensively studied with the goal of enhancing the neurogenic capacity of NSCs for regenerative medicine. However, neurogenesis of endogenous NSCs alone is insufficient to completely repair brains damaged by neurodegenerative diseases and/or injury because neurogenic areas are limited and few neurons are produced in the adult brain. An innovative approach towards replacing damaged neurons is to induce conversion of non-neuronal cells residing in injured sites into neurons by a process referred to as direct reprogramming. This review describes extrinsic and intrinsic factors controlling NSCs and neurogenesis in the adult brain and discusses prospects for their applications. It also describes direct neuronal reprogramming technology holding promise for future clinical applications.
Collapse
|
33
|
A FOXO-dependent replication checkpoint restricts proliferation of damaged cells. Cell Rep 2021; 34:108675. [PMID: 33503422 DOI: 10.1016/j.celrep.2020.108675] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 10/30/2020] [Accepted: 12/30/2020] [Indexed: 01/05/2023] Open
Abstract
DNA replication is challenged by numerous exogenous and endogenous factors that can interfere with the progression of replication forks. Substantial accumulation of single-stranded DNA during DNA replication activates the DNA replication stress checkpoint response that slows progression from S/G2 to M phase to protect genomic integrity. Whether and how mild replication stress restricts proliferation remains controversial. Here, we identify a cell cycle exit mechanism that prevents S/G2 phase arrested cells from undergoing mitosis after exposure to mild replication stress through premature activation of the anaphase promoting complex/cyclosome (APC/CCDH1). We find that replication stress causes a gradual decrease of the levels of the APC/CCDH1 inhibitor EMI1/FBXO5 through Forkhead box O (FOXO)-mediated inhibition of its transcription factor E2F1. By doing so, FOXOs limit the time during which the replication stress checkpoint is reversible and thereby play an important role in maintaining genomic stability.
Collapse
|
34
|
Voisin J, Farina F, Naphade S, Fontaine M, Tshilenge K, Galicia Aguirre C, Lopez‐Ramirez A, Dancourt J, Ginisty A, Sasidharan Nair S, Lakshika Madushani K, Zhang N, Lejeune F, Verny M, Campisi J, Ellerby LM, Neri C. FOXO3 targets are reprogrammed as Huntington's disease neural cells and striatal neurons face senescence with p16 INK4a increase. Aging Cell 2020; 19:e13226. [PMID: 33156570 PMCID: PMC7681055 DOI: 10.1111/acel.13226] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 06/26/2020] [Accepted: 07/26/2020] [Indexed: 12/21/2022] Open
Abstract
Neurodegenerative diseases (ND) have been linked to the critical process in aging—cellular senescence. However, the temporal dynamics of cellular senescence in ND conditions is unresolved. Here, we show senescence features develop in human Huntington's disease (HD) neural stem cells (NSCs) and medium spiny neurons (MSNs), including the increase of p16INK4a, a key inducer of cellular senescence. We found that HD NSCs reprogram the transcriptional targets of FOXO3, a major cell survival factor able to repress cell senescence, antagonizing p16INK4a expression via the FOXO3 repression of the transcriptional modulator ETS2. Additionally, p16INK4a promotes cellular senescence features in human HD NSCs and MSNs. These findings suggest that cellular senescence may develop during neuronal differentiation in HD and that the FOXO3‐ETS2‐p16INK4a axis may be part of molecular responses aimed at mitigating this phenomenon. Our studies identify neuronal differentiation with accelerated aging of neural progenitors and neurons as an alteration that could be linked to NDs.
Collapse
Affiliation(s)
- Jessica Voisin
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| | - Francesca Farina
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| | | | - Morgane Fontaine
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| | | | | | | | - Julia Dancourt
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| | - Aurélie Ginisty
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| | - Satish Sasidharan Nair
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| | | | | | - François‐Xavier Lejeune
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| | - Marc Verny
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| | - Judith Campisi
- Buck Institute for Research on Aging Novato CA USA
- Lawrence Berkeley National Laboratory Berkeley CA USA
| | | | - Christian Neri
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| |
Collapse
|
35
|
García-Prat L, Perdiguero E, Alonso-Martín S, Dell'Orso S, Ravichandran S, Brooks SR, Juan AH, Campanario S, Jiang K, Hong X, Ortet L, Ruiz-Bonilla V, Flández M, Moiseeva V, Rebollo E, Jardí M, Sun HW, Musarò A, Sandri M, Del Sol A, Sartorelli V, Muñoz-Cánoves P. FoxO maintains a genuine muscle stem-cell quiescent state until geriatric age. Nat Cell Biol 2020; 22:1307-1318. [PMID: 33106654 DOI: 10.1038/s41556-020-00593-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 09/15/2020] [Indexed: 02/07/2023]
Abstract
Tissue regeneration declines with ageing but little is known about whether this arises from changes in stem-cell heterogeneity. Here, in homeostatic skeletal muscle, we identify two quiescent stem-cell states distinguished by relative CD34 expression: CD34High, with stemness properties (genuine state), and CD34Low, committed to myogenic differentiation (primed state). The genuine-quiescent state is unexpectedly preserved into later life, succumbing only in extreme old age due to the acquisition of primed-state traits. Niche-derived IGF1-dependent Akt activation debilitates the genuine stem-cell state by imposing primed-state features via FoxO inhibition. Interventions to neutralize Akt and promote FoxO activity drive a primed-to-genuine state conversion, whereas FoxO inactivation deteriorates the genuine state at a young age, causing regenerative failure of muscle, as occurs in geriatric mice. These findings reveal transcriptional determinants of stem-cell heterogeneity that resist ageing more than previously anticipated and are only lost in extreme old age, with implications for the repair of geriatric muscle.
Collapse
Affiliation(s)
- Laura García-Prat
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.,Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain.,Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Eusebio Perdiguero
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Sonia Alonso-Martín
- Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain.,Neurosciences Area, Biodonostia Health Research Institute, Donostia-San Sebastián, San Sebastián, Spain
| | - Stefania Dell'Orso
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH Bethesda, Bethesda, MD, USA
| | - Srikanth Ravichandran
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Stephen R Brooks
- Biodata Mining and Discovery Section, NIAMS, NIH Bethesda, Bethesda, MD, USA
| | - Aster H Juan
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH Bethesda, Bethesda, MD, USA
| | - Silvia Campanario
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.,Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain
| | - Kan Jiang
- Biodata Mining and Discovery Section, NIAMS, NIH Bethesda, Bethesda, MD, USA
| | - Xiaotong Hong
- Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain
| | - Laura Ortet
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Vanessa Ruiz-Bonilla
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Marta Flández
- Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain.,Grupo de Investigación en Oncología Clínico Traslacional, Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Victoria Moiseeva
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Elena Rebollo
- Molecular Imaging Platform, Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Mercè Jardí
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Hong-Wei Sun
- Biodata Mining and Discovery Section, NIAMS, NIH Bethesda, Bethesda, MD, USA
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Marco Sandri
- Department of Biomedical Science, University of Padova, Padova, Italy
| | - Antonio Del Sol
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.,CIC bioGUNE, Derio, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Vittorio Sartorelli
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH Bethesda, Bethesda, MD, USA.
| | - Pura Muñoz-Cánoves
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain. .,Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain. .,ICREA, Barcelona, Spain.
| |
Collapse
|
36
|
Kobayashi T, Kageyama R. Lysosomes and signaling pathways for maintenance of quiescence in adult neural stem cells. FEBS J 2020; 288:3082-3093. [PMID: 32902139 PMCID: PMC8246936 DOI: 10.1111/febs.15555] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/18/2020] [Accepted: 09/01/2020] [Indexed: 12/28/2022]
Abstract
Quiescence is a cellular strategy for maintaining somatic stem cells in a specific niche in a low metabolic state without senescence for a long period of time. During development, neural stem cells (NSCs) actively proliferate and self-renew, and their progeny differentiate into both neurons and glial cells to form mature brain tissues. On the other hand, most NSCs in the adult brain are quiescent and arrested in G0/G1 phase of the cell cycle. Quiescence is essential in order to avoid the precocious exhaustion of NSCs, ensuring a sustainable source of available stem cells in the brain throughout the lifespan. After receiving activation signals, quiescent NSCs reenter the cell cycle and generate new neurons. This switching between quiescence and proliferation is tightly regulated by diverse signaling pathways. Recent studies suggest significant involvement of cellular proteostasis (homeostasis of the proteome) in the quiescent state of NSCs. Proteostasis is the result of integrated regulation of protein synthesis, folding, and degradation. In this review, we discuss regulation of quiescence by multiple signaling pathways, especially bone morphogenetic protein and Notch signaling, and focus on the functional involvement of the lysosome, an organelle governing cellular degradation, in quiescence of adult NSCs.
Collapse
Affiliation(s)
- Taeko Kobayashi
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Ryoichiro Kageyama
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto, Japan.,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto, Japan
| |
Collapse
|
37
|
Audesse AJ, Webb AE. Mechanisms of enhanced quiescence in neural stem cell aging. Mech Ageing Dev 2020; 191:111323. [PMID: 32781077 DOI: 10.1016/j.mad.2020.111323] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 12/24/2022]
Abstract
The maintenance of neural stem cell function is vital to ensure neurogenesis throughout adulthood. During aging, there is a significant reduction in adult neurogenesis that correlates with a decline in cognitive function. Although recent studies have revealed novel extrinsic and intrinsic mechanisms that regulate the adult neural stem cell (NSC) pool and lineage progression, the precise molecular mechanisms that drive dysregulation of adult neurogenesis in the context of aging are only beginning to emerge. Recent studies have shed light on mechanisms that regulate the earliest step of adult neurogenesis, the activation of quiescent NSCs. Interestingly, the ability of NSCs to enter the cell cycle in the aged brain significantly declines suggesting a deepend state of quiescence. Given the likely contribution of adult neurogenesis to supporting cognitive function in humans, enhancing neurogenesis may be a strategy to combat age-related cognitive decline. This review highlights the mechanisms that regulate the NSC pool throughout adulthood and discusses how dysregulation of these processes may contribute to the decline in neurogenesis and cognitive function throughout aging.
Collapse
Affiliation(s)
- Amanda J Audesse
- Graduate Program in Neuroscience, Brown University, USA; Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Ashley E Webb
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA; Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA; Center on the Biology of Aging, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
38
|
Labusch M, Mancini L, Morizet D, Bally-Cuif L. Conserved and Divergent Features of Adult Neurogenesis in Zebrafish. Front Cell Dev Biol 2020; 8:525. [PMID: 32695781 PMCID: PMC7338623 DOI: 10.3389/fcell.2020.00525] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/03/2020] [Indexed: 12/14/2022] Open
Abstract
Adult neurogenesis, i.e., the generation of neurons from neural stem cells (NSCs) in the adult brain, contributes to brain plasticity in all vertebrates. It varies, however, greatly in extent, location and physiological characteristics between species. During the last decade, the teleost zebrafish (D. rerio) was increasingly used to study the molecular and cellular properties of adult NSCs, in particular as a prominent NSC population was discovered at the ventricular surface of the dorsal telencephalon (pallium), in territories homologous to the adult neurogenic niches of rodents. This model, for its specific features (large NSC population, amenability to intravital imaging, high regenerative capacity) allowed rapid progress in the characterization of basic adult NSC features. We review here these findings, with specific comparisons with the situation in rodents. We specifically discuss the cellular nature of NSCs (astroglial or neuroepithelial cells), their heterogeneities and their neurogenic lineages, and the mechanisms controlling NSC quiescence and fate choices, which all impact the neurogenic output. We further discuss the regulation of NSC activity in response to physiological triggers and non-physiological conditions such as regenerative contexts.
Collapse
Affiliation(s)
- Miriam Labusch
- Zebrafish Neurogenetics Unit, Institut Pasteur, UMR 3738, CNRS, Team Supported by the Ligue Nationale Contre le Cancer, Paris, France.,Sorbonne Université, Collège Doctoral, Paris, France
| | - Laure Mancini
- Zebrafish Neurogenetics Unit, Institut Pasteur, UMR 3738, CNRS, Team Supported by the Ligue Nationale Contre le Cancer, Paris, France.,Sorbonne Université, Collège Doctoral, Paris, France
| | - David Morizet
- Zebrafish Neurogenetics Unit, Institut Pasteur, UMR 3738, CNRS, Team Supported by the Ligue Nationale Contre le Cancer, Paris, France.,Sorbonne Université, Collège Doctoral, Paris, France
| | - Laure Bally-Cuif
- Zebrafish Neurogenetics Unit, Institut Pasteur, UMR 3738, CNRS, Team Supported by the Ligue Nationale Contre le Cancer, Paris, France
| |
Collapse
|
39
|
Pro-neuronal activity of Myod1 due to promiscuous binding to neuronal genes. Nat Cell Biol 2020; 22:401-411. [PMID: 32231311 DOI: 10.1038/s41556-020-0490-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 02/18/2020] [Indexed: 12/25/2022]
Abstract
The on-target pioneer factors Ascl1 and Myod1 are sequence-related but induce two developmentally unrelated lineages-that is, neuronal and muscle identities, respectively. It is unclear how these two basic helix-loop-helix (bHLH) factors mediate such fundamentally different outcomes. The chromatin binding of Ascl1 and Myod1 was surprisingly similar in fibroblasts, yet their transcriptional outputs were drastically different. We found that quantitative binding differences explained differential chromatin remodelling and gene activation. Although strong Ascl1 binding was exclusively associated with bHLH motifs, strong Myod1-binding sites were co-enriched with non-bHLH motifs, possibly explaining why Ascl1 is less context dependent. Finally, we observed that promiscuous binding of Myod1 to neuronal targets results in neuronal reprogramming when the muscle program is inhibited by Myt1l. Our findings suggest that chromatin access of on-target pioneer factors is primarily driven by the protein-DNA interaction, unlike ordinary context-dependent transcription factors, and that promiscuous transcription factor binding requires specific silencing mechanisms to ensure lineage fidelity.
Collapse
|
40
|
Chen Y, Bravo JI, Son JM, Lee C, Benayoun BA. Remodeling of the H3 nucleosomal landscape during mouse aging. TRANSLATIONAL MEDICINE OF AGING 2020; 4:22-31. [PMID: 32462102 DOI: 10.1016/j.tma.2019.12.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In multi-cellular organisms, the control of gene expression is key not only for development, but also for adult cellular homeostasis, and deregulation of gene expression correlates with aging. A key layer in the study of gene regulation mechanisms lies at the level of chromatin: cellular chromatin states (i.e. the 'epigenome') can tune transcriptional profiles, and, in line with the prevalence of transcriptional alterations with aging, accumulating evidence suggests that the chromatin landscape is altered with aging across cell types and species. However, although alterations in the chromatin make-up of cells are considered to be a hallmark of aging, little is known of the genomic loci that are specifically affected by age-related chromatin state remodeling and of their biological significance. Here, we report the analysis of genome-wide profiles of core histone H3 occupancy in aging male mouse tissues (i.e. heart, liver, cerebellum and olfactory bulb) and primary cultures of neural stem cells. We find that, although no drastic changes in H3 levels are observed, local changes in H3 occupancy occur with aging across tissues and cells with both regions of increased or decreased occupancy. These changes are compatible with a general increase in chromatin accessibility at pro-inflammatory genes and may thus mechanistically underlie known shift in gene expression programs during aging.
Collapse
Affiliation(s)
- Yilin Chen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.,Master of Science in Nutrition, Healthspan, and Longevity, University of Southern California, Los Angeles, CA 90089, USA
| | - Juan I Bravo
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.,Graduate program in the Biology of Aging, University of Southern California, Los Angeles, CA 90089, USA
| | - Jyung Mean Son
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Changhan Lee
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.,USC Norris Comprehensive Cancer Center, Epigenetics and Gene Regulation, Los Angeles, CA 90089, USA.,Biomedical Sciences, Graduate School, Ajou University, Suwon 16499, Republic of Korea
| | - Bérénice A Benayoun
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.,USC Norris Comprehensive Cancer Center, Epigenetics and Gene Regulation, Los Angeles, CA 90089, USA.,USC Stem Cell Initiative, Los Angeles, CA 90089, USA
| |
Collapse
|
41
|
Quiescence of Adult Mammalian Neural Stem Cells: A Highly Regulated Rest. Neuron 2019; 104:834-848. [DOI: 10.1016/j.neuron.2019.09.026] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/05/2019] [Accepted: 09/17/2019] [Indexed: 12/14/2022]
|
42
|
Traxler L, Edenhofer F, Mertens J. Next-generation disease modeling with direct conversion: a new path to old neurons. FEBS Lett 2019; 593:3316-3337. [PMID: 31715002 PMCID: PMC6907729 DOI: 10.1002/1873-3468.13678] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/20/2019] [Accepted: 11/07/2019] [Indexed: 12/13/2022]
Abstract
Within just over a decade, human reprogramming-based disease modeling has developed from a rather outlandish idea into an essential part of disease research. While iPSCs are a valuable tool for modeling developmental and monogenetic disorders, their rejuvenated identity poses limitations for modeling age-associated diseases. Direct cell-type conversion of fibroblasts into induced neurons (iNs) circumvents rejuvenation and preserves hallmarks of cellular aging. iNs are thus advantageous for modeling diseases that possess strong age-related and epigenetic contributions and can complement iPSC-based strategies for disease modeling. In this review, we provide an overview of the state of the art of direct iN conversion and describe the key epigenetic, transcriptomic, and metabolic changes that occur in converting fibroblasts. Furthermore, we summarize new insights into this fascinating process, particularly focusing on the rapidly changing criteria used to define and characterize in vitro-born human neurons. Finally, we discuss the unique features that distinguish iNs from other reprogramming-based neuronal cell models and how iNs are relevant to disease modeling.
Collapse
Affiliation(s)
- Larissa Traxler
- Department of GenomicsStem Cell Biology & Regenerative MedicineInstitute of Molecular Biology & CMBILeopold‐Franzens‐University InnsbruckInnsbruckAustria
- Laboratory of GeneticsThe Salk Institute for Biological StudiesLa JollaCAUSA
| | - Frank Edenhofer
- Department of GenomicsStem Cell Biology & Regenerative MedicineInstitute of Molecular Biology & CMBILeopold‐Franzens‐University InnsbruckInnsbruckAustria
| | - Jerome Mertens
- Department of GenomicsStem Cell Biology & Regenerative MedicineInstitute of Molecular Biology & CMBILeopold‐Franzens‐University InnsbruckInnsbruckAustria
- Laboratory of GeneticsThe Salk Institute for Biological StudiesLa JollaCAUSA
| |
Collapse
|
43
|
Rizk M, Rizq O, Oshima M, Nakajima-Takagi Y, Koide S, Saraya A, Isshiki Y, Chiba T, Yamazaki S, Ma A, Jin J, Iwama A, Mimura N. Akt inhibition synergizes with polycomb repressive complex 2 inhibition in the treatment of multiple myeloma. Cancer Sci 2019; 110:3695-3707. [PMID: 31571328 PMCID: PMC6890440 DOI: 10.1111/cas.14207] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/18/2019] [Accepted: 09/23/2019] [Indexed: 12/19/2022] Open
Abstract
Polycomb repressive complex 2 (PRC2) components, EZH2 and its homolog EZH1, and PI3K/Akt signaling pathway are focal points as therapeutic targets for multiple myeloma. However, the exact crosstalk between their downstream targets remains unclear. We herein elucidated some epigenetic interactions following Akt inhibition and demonstrated the efficacy of the combined inhibition of Akt and PRC2. We found that TAS-117, a potent and selective Akt inhibitor, downregulated EZH2 expression at the mRNA and protein levels via interference with the Rb-E2F pathway, while EZH1 was compensatively upregulated to maintain H3K27me3 modifications. Consistent with these results, the dual EZH2/EZH1 inhibitor, UNC1999, but not the selective EZH2 inhibitor, GSK126, synergistically enhanced TAS-117-induced cytotoxicity and provoked myeloma cell apoptosis. RNA-seq analysis revealed the activation of the FOXO signaling pathway after TAS-117 treatment. FOXO3/4 mRNA and their downstream targets were upregulated with the enhanced nuclear localization of FOXO3 protein after TAS-117 treatment. ChIP assays confirmed the direct binding of FOXO3 to EZH1 promoter, which was enhanced by TAS-117 treatment. Moreover, FOXO3 knockdown repressed EZH1 expression. Collectively, the present results reveal some molecular interactions between Akt signaling and epigenetic modulators, which emphasize the benefits of targeting PRC2 full activity and the Akt pathway as a therapeutic option for multiple myeloma.
Collapse
Affiliation(s)
- Mohamed Rizk
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.,Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ola Rizq
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.,Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Motohiko Oshima
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.,Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yaeko Nakajima-Takagi
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.,Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shuhei Koide
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Atsunori Saraya
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yusuke Isshiki
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.,Department of Hematology, Chiba University Hospital, Chiba, Japan.,Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tetsuhiro Chiba
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Satoshi Yamazaki
- Division of Stem Cell Biology, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Anqi Ma
- Department of Pharmacological Sciences, Mount Sinai Center for Therapeutics Discovery, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Mount Sinai Center for Therapeutics Discovery, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jian Jin
- Department of Pharmacological Sciences, Mount Sinai Center for Therapeutics Discovery, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Mount Sinai Center for Therapeutics Discovery, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Atsushi Iwama
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.,Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Naoya Mimura
- Department of Transfusion Medicine and Cell Therapy, Chiba University Hospital, Chiba, Japan
| |
Collapse
|
44
|
Abbott LC, Nigussie F. Adult neurogenesis in the mammalian dentate gyrus. Anat Histol Embryol 2019; 49:3-16. [PMID: 31568602 DOI: 10.1111/ahe.12496] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 06/03/2019] [Accepted: 08/22/2019] [Indexed: 12/30/2022]
Abstract
Earlier observations in neuroscience suggested that no new neurons form in the mature central nervous system. Evidence now indicates that new neurons do form in the adult mammalian brain. Two regions of the mature mammalian brain generate new neurons: (a) the border of the lateral ventricles of the brain (subventricular zone) and (b) the subgranular zone (SGZ) of the dentate gyrus of the hippocampus. This review focuses only on new neuron formation in the dentate gyrus of the hippocampus. During normal prenatal and early postnatal development, neural stem cells (NSCs) give rise to differentiated neurons. NSCs persist in the dentate gyrus SGZ, undergoing cell division, with some daughter cells differentiating into functional neurons that participate in learning and memory and general cognition through integration into pre-existing neural networks. Axons, which emanate from neurons in the entorhinal cortex, synapse with dendrites of the granule cells (small neurons) of the dentate gyrus. Axons from granule cells synapse with pyramidal cells in the hippocampal CA3 region, which send axons to synapse with CA1 hippocampal pyramidal cells that send their axons out of the hippocampus proper. Adult neurogenesis includes proliferation, differentiation, migration, the death of some newly formed cells and final integration of surviving cells into neural networks. We summarise these processes in adult mammalian hippocampal neurogenesis and discuss the roles of major signalling molecules that influence neurogenesis, including neurotransmitters and some hormones. The recent controversy raised concerning whether or not adult neurogenesis occurs in humans also is discussed.
Collapse
Affiliation(s)
- Louise C Abbott
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Fikru Nigussie
- College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| |
Collapse
|
45
|
Liu YZ, Peng W, Chen JK, Su WJ, Yan WJ, Wang YX, Jiang CL. FoxO1 is a critical regulator of hepatocyte lipid deposition in chronic stress mice. PeerJ 2019; 7:e7668. [PMID: 31579588 PMCID: PMC6768057 DOI: 10.7717/peerj.7668] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/13/2019] [Indexed: 01/28/2023] Open
Abstract
Forkhead box O1 (FoxO1) is involved in lipid metabolisms. However, its role in chronic stress-related nonalcoholic fatty liver disease (NAFLD) is unclear. The scientific premise of our study was based on the finding that FoxO1 expression is increased in the liver of mice after chronic stress. It is important to understand the mechanisms involved in the activation of FoxO1 and how its function affects the liver lipid deposition. We employed a murine chronic stress model, in which mice were treated by plantar electrical stimulation and restraint for 6 weeks, and a cellular model, in which Hepa1–6 cells were treated with corticosterone. We also used a pharmacologic approach as1842856, a highly specific FoxO1 inhibitor. Lipid metabolism related genes levels were measured by qRT-PCR and the lipid levels by biochemical detection. We show that the level of FoxO1 is significantly elevated in the liver of chronic stress mice. Transcription factor FoxO1 regulates a lipid synthesis phenotype of hepatocyte that is involved in the development and progression of NAFLD. We have shown that inhibition of FoxO1 induced phenotypic conversion of hepatocytes and down-regulates lipid synthesis genes expression by hepatocytes, which contribute to lipid deposition in NAFLD. At the cellular level, the inhibitor of FoxO1 as1842856 can also attenuate the lipid deposition of Hepa1–6 cells induced by corticosterone. Targeting FoxO1 is a novel therapeutic target for chronic stress-related NAFLD.
Collapse
Affiliation(s)
- Yun-Zi Liu
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, Shanghai, China
| | - Wei Peng
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, Shanghai, China
| | - Ji-Kuai Chen
- Department of Health Toxicology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Wen-Jun Su
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, Shanghai, China
| | - Wen-Jie Yan
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, Shanghai, China
| | - Yun-Xia Wang
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, Shanghai, China
| | - Chun-Lei Jiang
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, Shanghai, China
| |
Collapse
|
46
|
Coller HA. The paradox of metabolism in quiescent stem cells. FEBS Lett 2019; 593:2817-2839. [PMID: 31531979 DOI: 10.1002/1873-3468.13608] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/05/2019] [Accepted: 09/10/2019] [Indexed: 12/12/2022]
Abstract
The shift between a proliferating and a nonproliferating state is associated with significant changes in metabolic needs. Proliferating cells tend to have higher metabolic rates, and their metabolic profiles facilitate biosynthesis, as compared to those of nondividing cells of the same sort. Recent studies have elucidated specific molecules that control metabolic changes while cells shift between proliferation and quiescence. Embryonic stem cells, which are rapidly proliferating, tend to have metabolic patterns that are similar to those of nonstem cells in a proliferative state. Moreover, although adult stem cells tend to be quiescent, their metabolic profiles have been reported in multiple organs to more closely resemble those of proliferating than those of nondividing cells in some respects. The findings raise questions about whether there are metabolic profiles that are required for stemness, and whether these profiles relate to the metabolic properties that may be required for quiescence. Here, we review the literature on how metabolism changes upon commitment to proliferation and compare the proliferating and nonproliferating metabolic states of differentiated cells and embryonic and adult stem cells.
Collapse
Affiliation(s)
- Hilary A Coller
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA.,Department of Biological Chemistry, David Geffen School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
47
|
FOXO1 Confers Maintenance of the Dark Zone Proliferation and Survival Program and Can Be Pharmacologically Targeted in Burkitt Lymphoma. Cancers (Basel) 2019; 11:cancers11101427. [PMID: 31557894 PMCID: PMC6826697 DOI: 10.3390/cancers11101427] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/16/2019] [Accepted: 09/20/2019] [Indexed: 12/19/2022] Open
Abstract
The FOXO1 transcription factor plays a central role in the proliferation and survival of B cells at several stages of differentiation. B cell malignancies, with exception of classical Hodgkin lymphoma, maintain expression of FOXO1 at levels characteristic for their non-malignant counterparts. Extensive expression profiling had revealed that Burkitt lymphoma (BL) show many characteristics of the dark zone (DZ) germinal center (GC) B cell program. Here we show that FOXO1 knockdown inhibits proliferation of human BL cell lines. The anti-proliferative effect of the FOXO1 knockdown is associated with the repression of the DZ B cell program including expression of MYB, CCND3, RAG2, BACH2, and CXCR4. In addition, the induction of signaling pathways of the light zone (LZ) program like NF-κB and PI3K-AKT was observed. Using a rescue experiment we identified downregulation of the proto-oncogene MYB as a critical factor contributing to the antiproliferative effect of FOXO1 knockdown. In an attempt to estimate the feasibility of pharmacological FOXO1 repression, we found that the small molecular weight FOXO1 inhibitor AS1842856 induces cell death and growth arrest in BL cell lines at low concentrations. Interestingly, we found that overactivation of FOXO1 also induces growth inhibition in BL cell lines, indicating the importance of a tight regulation of FOXO1 activity in BL.
Collapse
|
48
|
Kim SA, Jang EH, Mun JY, Choi H. Propionic acid induces mitochondrial dysfunction and affects gene expression for mitochondria biogenesis and neuronal differentiation in SH-SY5Y cell line. Neurotoxicology 2019; 75:116-122. [PMID: 31526819 DOI: 10.1016/j.neuro.2019.09.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/27/2019] [Accepted: 09/12/2019] [Indexed: 12/20/2022]
Abstract
Studies in animal models have shown that the short-chain fatty acid, propionic acid (PPA), interferes with mitochondrial metabolism leading to mitochondrial dysfunction and behavioral abnormalities. The aim of this study was to investigate the effects of PPA on mitochondrial function and gene expression in neuronal cells. SH-SY5Y cells and normal human neural progenitor (NHNP) cells were exposed to 1, 5 mM PPA for 4 or 24 h and we found that the mitochondrial potential measured in SH-SY5Y cells decreased in a dose-dependent manner after PPA treatment. Electron microscopy analysis revealed that the size of the mitochondria was significantly reduced following PPA treatment. A dose-dependent increase in the mitochondrial DNA copy number was observed in the PPA-treated cells. The expression of the mitochondrial biogenesis-related proteins PGC-1α, TFAM, SIRT3, and COX4 was significantly increased after PPA treatment. Transcriptome analysis revealed that mRNA expression in the notch signaling-related genes ASCL1 and LFNG changed after PPA treatment and the positive correlated protein expression changes were also observed. These results revealed that PPA treatment may affect neurodevelopment by altering mitochondrial function and notch signaling-related gene expression.
Collapse
Affiliation(s)
- Soon Ae Kim
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon, Republic of Korea.
| | - Eun Hye Jang
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon, Republic of Korea
| | - Ji Young Mun
- Neural Circuits Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Hyosun Choi
- Neural Circuits Research Group, Korea Brain Research Institute, Daegu, Republic of Korea; BK21 Plus Program, Department of Senior Healthcare, Graduate School, Eulji University, Daejeon, Republic of Korea
| |
Collapse
|
49
|
A statistical approach to fine-mapping for the identification of potential causal variants related to human intelligence. J Hum Genet 2019; 64:781-787. [PMID: 31165785 DOI: 10.1038/s10038-019-0623-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 04/28/2019] [Accepted: 05/15/2019] [Indexed: 01/10/2023]
Abstract
Genome-wide association studies (GWASs) have identified >20 genetic loci associated with human intelligence. However, due to correlations between the trait-associated SNPs, only a few of the loci are confirmed to have a true biological effect. In order to distinguish the SNPs that have a causal effect on human intelligence, we must eliminate the noise from the high degree of linkage disequilibrium that persists throughout the genome. In this study, we apply a novel PAINTOR fine-mapping method, which uses a Bayesian approach to determine the SNPs with the highest probability of causality. This technique incorporates the GWAS summary statistics, linkage disequilibrium structure, and functional annotations to compute the posterior probability of causality for all SNPs in the GWAS-associated regions. We found five SNPs (rs6002620, rs41352752, rs6568547, rs138592330, and rs28371699) with a high probability of causality, three of which have posterior probabilities >0.60. The SNP rs6002620 (NDUFA6), which is involved in mitochondrial function, has the highest likelihood of causality. These findings provide important insight into the genetic determinants contributing to human intelligence.
Collapse
|
50
|
Birnbaum A, Wu X, Tatar M, Liu N, Bai H. Age-Dependent Changes in Transcription Factor FOXO Targeting in Female Drosophila. Front Genet 2019; 10:312. [PMID: 31134124 PMCID: PMC6514159 DOI: 10.3389/fgene.2019.00312] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 03/21/2019] [Indexed: 11/13/2022] Open
Abstract
FOXO transcription factors have long been associated with longevity control and tissue homeostasis. Although the transcriptional regulation of FOXO have been previously characterized (especially in long-lived insulin mutants and under stress conditions), how normal aging impacts the transcriptional activity of FOXO is poorly understood. Here, we conducted a chromatin immunoprecipitation sequencing (ChIP-Seq) analysis in both young (2-week-old) and aged (5-week-old) wild-type female fruit flies, Drosophila melanogaster, to evaluate the dynamics of FOXO gene targeting during aging. Intriguingly, the number of FOXO-bound genes dramatically decreases with age (from 2617 to 224). Consistent to the reduction of FOXO binding activity, many genes targeted by FOXO in young flies are transcriptionally altered with age, either up-regulated (FOXO-repressing genes) or down-regulated (FOXO-activating genes) in adult head tissue. In addition, we show that many FOXO-bound genes in wild-type flies are unique from those in insulin receptor substrate chico mutants. Distinct from chico mutants, FOXO targets specific cellular processes (e.g., actin cytoskeleton) and signaling pathways (e.g., Hippo, MAPK) in young wild-type female flies. FOXO targeting on these pathways decreases with age. Interestingly, FOXO targets in aged flies are enriched in cellular processes like chromatin organization and nucleosome assembly. Furthermore, FOXO binding to core histone genes is well maintained at aged flies. Together, our findings provide new insights into dynamic FOXO targeting under normal aging and highlight the diverse and understudied regulatory mechanisms for FOXO transcriptional activity.
Collapse
Affiliation(s)
- Allison Birnbaum
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, United States
| | - Xiaofen Wu
- Interdisciplinary Research Center on Biology and Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Marc Tatar
- Department of Ecology and Evolutionary Biology, Brown University, Providence, RI, United States
| | - Nan Liu
- Interdisciplinary Research Center on Biology and Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Hua Bai
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, United States
| |
Collapse
|